12.07.2015 Views

Adult Neurogenesis: From Precursors to Network and Physiology

Adult Neurogenesis: From Precursors to Network and Physiology

Adult Neurogenesis: From Precursors to Network and Physiology

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

Physiol Rev 85: 523–569, 2005;doi:10.1152/physrev.00055.2003.<strong>Adult</strong> <strong>Neurogenesis</strong>: <strong>From</strong> <strong>Precursors</strong><strong>to</strong> <strong>Network</strong> <strong>and</strong> <strong>Physiology</strong>DJOHER NORA ABROUS, MURIEL KOEHL, AND MICHEL LE MOALLabora<strong>to</strong>ire de Physiopathologie des Comportements, Institut National de la Santéet de la Recherche Médicale, U588, Université de Bordeaux 2, Bordeaux, FranceI. <strong>Neurogenesis</strong> in the <strong>Adult</strong> Brain: a New Paradigm for Structure-Function Relationships 523II. <strong>From</strong> Newly Born Cells <strong>to</strong> <strong>Network</strong> 524A. Definitions 524B. In vivo evaluation of adult neurogenesis: methodological issues 525C. Integration of the newly born cells in<strong>to</strong> neurogenic site networks 527D. Fac<strong>to</strong>rs regulating adult neurogenesis 533III. The Ana<strong>to</strong>mo-Functional Approach 539A. Environmental <strong>and</strong> physiological influences on neurogenesis 539B. Activation of the stress axis: acute <strong>and</strong> long-term effects on neurogenesis 543C. Aging <strong>and</strong> longitudinal observations 545D. Involvement of neurogenesis in pathologies 548IV. Conclusion 553Abrous, Djoher Nora, Muriel Koehl, <strong>and</strong> Michel Le Moal. <strong>Adult</strong> <strong>Neurogenesis</strong>: <strong>From</strong> <strong>Precursors</strong> <strong>to</strong> <strong>Network</strong> <strong>and</strong><strong>Physiology</strong>. Physiol Rev 85: 523–569, 2005; doi:10.1152/physrev.00055.2003.—The discovery that the adult mammalianbrain creates new neurons from pools of stemlike cells was a breakthrough in neuroscience. Interestingly, thisparticular new form of structural brain plasticity seems specific <strong>to</strong> discrete brain regions, <strong>and</strong> most investigationsconcern the subventricular zone (SVZ) <strong>and</strong> the dentate gyrus (DG) of the hippocampal formation (HF). Overall, twomain lines of research have emerged over the last two decades: the first aims <strong>to</strong> underst<strong>and</strong> the fundamentalbiological properties of neural stemlike cells (<strong>and</strong> their progeny) <strong>and</strong> the integration of the newly born neurons in<strong>to</strong>preexisting networks, while the second focuses on underst<strong>and</strong>ing its relevance in brain functioning, which has beenmore extensively approached in the DG. Here, we propose an overview of the current knowledge on adultneurogenesis <strong>and</strong> its functional relevance for the adult brain. We first present an analysis of the methodologicalissues that have hampered progress in this field <strong>and</strong> describe the main neurogenic sites with their specificities. Wewill see that despite considerable progress, the levels of ana<strong>to</strong>mic <strong>and</strong> functional integration of the newly bornneurons within the host circuitry have yet <strong>to</strong> be elucidated. Then the intracellular mechanisms controlling neuronalfate are presented briefly, along with the extrinsic fac<strong>to</strong>rs that regulate adult neurogenesis. We will see that agrowing list of epigenetic fac<strong>to</strong>rs that display a specificity of action depending on the neurogenic site underconsideration has been identified. Finally, we review the progress accomplished in implicating neurogenesis inhippocampal functioning under physiological conditions <strong>and</strong> in the development of hippocampal-related pathologiessuch as epilepsy, mood disorders, <strong>and</strong> addiction. This constitutes a necessary step in promoting the development oftherapeutic strategies.Downloaded from physrev.physiology.org on November 25, 2005I. NEUROGENESIS IN THE ADULT BRAIN: ANEW PARADIGM FOR STRUCTURE-FUNCTION RELATIONSHIPSDevelopmental biology teaches us that brain functioningrequires neurons of appropriate types <strong>to</strong> be generatedin appropriate places <strong>and</strong> numbers, <strong>and</strong> then <strong>to</strong>migrate <strong>to</strong> their final positions <strong>and</strong> refine their synapticcontacts <strong>to</strong> a high degree of precision <strong>and</strong> avoid aberrantconnections. Finally, the brain must accommodategrowth of the organism <strong>and</strong> new behavioral or cognitivecapacities. For decades, neurobiologists have known thatneuronal network organization as well as individual performancesare altered by environmental events, experience,or internal signals such as hormones, aging, <strong>and</strong>various injuries (322).The adult brain faces two seemingly contradic<strong>to</strong>rychallenges. On the one h<strong>and</strong> it must maintain behaviorwww.prv.org 0031-9333/05 $18.00 Copyright © 2005 the American Physiological Society523


524 ABROUS, KOEHL, AND LE MOAL<strong>and</strong> thus preserve the underlying circuitry, <strong>and</strong> on theother h<strong>and</strong>, it must allow circuits <strong>to</strong> adapt <strong>to</strong> environmentalchallenges. This dilemma between stability <strong>and</strong> plasticityis a subject of debate. It has been suggested that thestructural changes underlying plasticity exist at the levelsof dendritic spines, dendrites, <strong>and</strong> axons (28, 287, 297,383, 408). However, direct observation of dendrites <strong>and</strong>spines in the adult brain supports two divergent viewpoints, i.e., both long-term dendrite stability (192, 368)<strong>and</strong> experience-dependent synaptic plasticity (541). Datasuggest that dendritic spines are heterogeneous in shape<strong>and</strong> structure <strong>and</strong> can be roughly divided in two groups:large spines that survive for months <strong>and</strong> even years <strong>and</strong>small spines that are motile, change form rapidly, <strong>and</strong>either disappear or transform in<strong>to</strong> large spines (for review,see Refs. 132, 216, 255). During long-term potentiation(LTP) of synaptic transmission, which is known <strong>to</strong>involve modifications in dendritic spine morphology (297,337, 586), small spines are both generated <strong>and</strong> eliminated(143, 338). Given the structure-stability-function relationships,it has been suggested that the small spines aregenerated during activity-dependent processes <strong>and</strong> acquisitionof memories, providing an inexhaustible source ofnew synapses, <strong>and</strong> that the large spines are the structuralbasis of memories in the long-term (255). For many authors,all these local <strong>and</strong> cellular phenomena have beenlabeled as neuroplasticity, a concept used <strong>to</strong> account forthe functional observations.In the search for structure-function relationships,great enthusiasm followed the discovery that embryonicneurons, for instance, dopaminergic neurons, could betransplanted in<strong>to</strong> adult brains, survive <strong>and</strong> alleviate somepostlesion behavioral alterations. Although it generated aconsiderable amount of data, this approach has notproven very successful so far in living up <strong>to</strong> functional <strong>and</strong>therapeutic hopes (157, 217). However, it has generated aconsiderable amount of data on the growth <strong>and</strong> the migrationof these neurons, <strong>and</strong> on their ability <strong>to</strong> makesynapse with the surrounding neurons of the host <strong>and</strong> <strong>to</strong>influence the activity of their target neurons. However,the turning point has been the demonstration that theadult mammalian brain is also capable of mi<strong>to</strong>sis <strong>and</strong> thatthe generated cells differentiate in<strong>to</strong> neurons that migrate<strong>to</strong> integrate circuitries. Interestingly, this particular newform of structural brain plasticity seems specific <strong>to</strong> discretebrain regions <strong>and</strong> most investigations concern thesubventricular zone (SVZ) <strong>and</strong> the dentate gyrus (DG)of the hippocampal formation (HF), although an interestingdebate developed in the wake of the propositionthat neurogenesis occurs in the neocortex of the adultprimate.In brief, the discovery of neurogenesis in the adultbrain confronted the persistent assumption that adultneurons did not undergo proliferation <strong>and</strong> that structurecould not be changed in this way. In the 1960s, Altman’sfirst observations of adult neurogenesis (11), followed bythe studies of Kaplan <strong>and</strong> Hinds (for a his<strong>to</strong>rical view, seeRef. 249), were followed by negative reactions <strong>and</strong> criticalpublications that did not confirm the existence of newbornneurons in adults (452). Ironically, at about the sameperiod, data were published on neurogenesis in birdsrelated <strong>to</strong> the appearance of seasonal song, a discoverythat gave rise <strong>to</strong> new thoughts on brain plasticity <strong>and</strong>adaptation (for review, see Ref, 410).Overall, two main lines of research have emergedsince the discovery that neurogenesis persists in discreteregions of the adult brain. The first aims <strong>to</strong> isolate neuralstem cells <strong>and</strong> underst<strong>and</strong> their fundamental biologicalproperties, the ultimate objective being <strong>to</strong> manipulatethem <strong>and</strong> enhance repair <strong>and</strong> regeneration. The secondfocuses on underst<strong>and</strong>ing its functional relevance, especiallyin the DG. We will thus approach these two lines ofresearch in the following chapters. As we will see, it hasnow been unambiguously demonstrated that persistentneurogenesis occurs in the SVZ <strong>and</strong> DG of adult rodents.Most of our knowledge on the birth, proliferation, migration,<strong>and</strong> death of adult-born cells results from studiesperformed in the SVZ paradigm that easily allow the dissectionof the properties of the stem cells (<strong>and</strong> theirprogeny) <strong>and</strong> the integration of the newly born neuronsin<strong>to</strong> preexisting networks. Once the existence of the phenomenonhad been established, the important step was <strong>to</strong>underst<strong>and</strong> what triggers <strong>and</strong> inhibits it, <strong>and</strong> how it isregulated. Identifying the functional roles of these adultbornneurons in the context of structure-function relationshipswill be the main focus of the review. The considerableevidence supporting the participation of thesenew neurons in brain functioning has been collected inthe DG, <strong>and</strong> their role in information s<strong>to</strong>rage <strong>and</strong> hippocampal-relatedpathology will be examined. On theassumption that the DG transforms en<strong>to</strong>rhinal inputs <strong>to</strong>facilitate s<strong>to</strong>rage in CA3, adult neurogenesis may allowthe DG <strong>to</strong> adapt <strong>to</strong> new flows of relevant informationwhile at the same time preserving it for the processing ofold memories. In more general terms, we will ask whetherneurogenesis acts on memory consolidation in a specific<strong>and</strong> functional manner or whether it prepares the HF forgeneral experiences <strong>and</strong> challenges.II. FROM NEWLY BORN CELLS TO NETWORKA. DefinitionsThe recent interest in adult neurogenesis studies hasled <strong>to</strong> a promiscuous use of the term stem cell <strong>and</strong> abroadening of its definition. However, based on theirfunctional properties, one can distinguish “stem” cellsfrom “progeni<strong>to</strong>r or precursor” cells. Thus a stem cell iscurrently defined as an undifferentiated cell that exhibitsDownloaded from physrev.physiology.org on November 25, 2005Physiol Rev • VOL 85 • APRIL 2005 • www.prv.org


NEUROGENESIS IN THE ADULT BRAIN 525the ability <strong>to</strong> proliferate, <strong>to</strong> self-renew, <strong>and</strong> <strong>to</strong> differentiatein<strong>to</strong> multiple yet distinct lineages (356, 570). In the adultbrain, most stemlike cells are in a quiescent stage, exceptin the two neurogenic zones considered here, the SVZ <strong>and</strong>the DG of the HF, where they have a very slow dividingturnover (a few weeks). In contrast, progeni<strong>to</strong>r cells aremi<strong>to</strong>tic cells with a faster dividing cell cycle that retainthe ability <strong>to</strong> proliferate <strong>and</strong> <strong>to</strong> give rise <strong>to</strong> terminallydifferentiated cells but are not capable of indefinite selfrenewal;they are more committed than stemlike cells,<strong>and</strong> their multipotentiality is still a matter of debate.Finally, when the cell type being studied is not clear, as isthe case in vivo, both stem <strong>and</strong> progeni<strong>to</strong>r cells are referred<strong>to</strong> as precursor cells. For an overview of the characteristicsthat can be used <strong>to</strong> differentiate neural stemfrom progeni<strong>to</strong>r cells, <strong>and</strong> of the controversies that stillpersist around definitions, we refer the reader <strong>to</strong> reviewsby Gage (160), Geuna et al. (167), <strong>and</strong> Seaberg <strong>and</strong> van derKooy (494).B. In Vivo Evaluation of <strong>Adult</strong> <strong>Neurogenesis</strong>:Methodological Issues1. Original studies: DNA synthesis staining <strong>and</strong>BrdU utilizationThe original studies on in vivo cell proliferation reliedon the use of [ 3 H]thymidine ([ 3 H]dT), which is incorporatedin<strong>to</strong> the cell during DNA synthesis, i.e., the Sphase of the cell cycle. Mi<strong>to</strong>tically active cells are subsequentlyrevealed by au<strong>to</strong>radiography. More recently,BrdU, an analog of thymidine, has been used in lieu of[ 3 H]dT (412). These markers have comparable availabilitytimes <strong>and</strong> labeling efficiencies (411). Although [ 3 H]dTpresents the advantage of good s<strong>to</strong>ichiometry (allowingdetermination of the number of mi<strong>to</strong>tic events after labeling),BrdU revealed by immunohis<strong>to</strong>chemistry is morefrequently used as the cost is lower, section processing isfaster, <strong>and</strong> stereological analysis is possible. In addition,BrdU is easier <strong>to</strong> combine with other cellular markers,which makes it possible <strong>to</strong> phenotype the newly borncells by double or triple immunohis<strong>to</strong>chemistry.The original pro<strong>to</strong>cols used for BrdU labeling, establishedby Gage <strong>and</strong> colleagues (268, 267), consisted of adozen BrdU injections (50 mg · kg 1 · day 1 ip). Since thispioneering work, with the flurry of data, pro<strong>to</strong>cols havediversified. As we will see in section III, the number <strong>and</strong>the doses of BrdU injections as well as the frequency ofadministration vary considerably from one experiment <strong>to</strong>another depending on the area of interest (fewer injectionsare required for studying the SVZ compared with theDG), the species, the phenomena being examined (a decreaseor an increase in neurogenesis), <strong>and</strong> the subjecttype (young intact subjects, impaired or old individuals).To make the picture more complex, BrdU is sometimesadministered in mice via drinking water (1–1.5 mg/ml for2–4 wk) <strong>to</strong> minimize animal pain <strong>and</strong> distress (80, 332),with the disadvantage that individual daily intake is unknown.Following BrdU injections, the killing of the animal isadjusted depending on the step of neurogenesis that isbeing investigated: cell proliferation, cell determination<strong>to</strong>ward a given phenotype, cell migration <strong>and</strong> differentiation,or cell death. In this way, <strong>to</strong> study proliferation,animals are killed following the last BrdU injection withina short time lapse ranging from a few hours <strong>to</strong> a few days.As early as 2 h after a single injection, mi<strong>to</strong>tic figures canbe seen, <strong>and</strong> this time point has been proposed as a truemeasure of proliferation. However, in the case of senescentrats, multiple injections are required as cell proliferation<strong>and</strong> the frequency of labeled clusters are <strong>to</strong>o low.Furthermore, when animals are killed within a short timeafter the last BrdU injection, newly born cells are small,irregular, <strong>and</strong> clustered (the clusters being more numerouswith multiple BrdU injections), which renders quantitativeanalysis challenging. For this reason, a washoutpro<strong>to</strong>col of a few days allowing cells <strong>to</strong> migrate is appliedin many experiments. Cell migration is followed by killingthe animals at different days (1–10 days) after BrdU injection,in which case it is necessary <strong>to</strong> limit the numberof BrdU injections (208). Survival of the newborn cells isusually studied by killing the animals after a long delay(several weeks) following BrdU administration, when thephenotype of the cells has been determined. Less frequently,retroviral tracing has also been used for “developmental”studies (81, 123, 439, 501). However, becauseretroviral infection is not controlled, labeling is quitevariable <strong>and</strong> is thus unsuitable for quantitative analysisbetween different experimental groups (556). Finally, inmost pharmacological <strong>and</strong> behavioral studies, the treatmentunder investigation is interrupted after BrdU pulses,<strong>and</strong> animals are killed after a “chase” period of severalweeks. It is important <strong>to</strong> be aware that in this case, theinfluence of the treatment on cell determination <strong>and</strong> cellsurvival is not addressed. To do so, the “treatment” shouldbe prolonged over the “chase” period time (for example,see Refs. 267, 335).2. Phenotyping cellsOne of the most important tasks is <strong>to</strong> phenotypecells, <strong>and</strong> markers specific <strong>to</strong> the dividing cells or thestate of maturation of newly born cells are available for invivo studies.A) PRECURSOR CELLS. So far, no marker that is exclusive <strong>to</strong>stemlike cells has been identified, which certainly reflectsthe divergence in opinion on the identity of the stemlikecell population. However, markers of stem-cell c<strong>and</strong>idatesor simply dividing cells are now available <strong>and</strong> canbe divided in<strong>to</strong> two categories depending on the purposeDownloaded from physrev.physiology.org on November 25, 2005Physiol Rev • VOL 85 • APRIL 2005 • www.prv.org


526 ABROUS, KOEHL, AND LE MOALof the studies. The first category covers “universal” orgeneral molecular markers that are used <strong>to</strong> define thebiological characteristics of stemlike cells (for an extensivereview, see Ref. 440). Among these markers, the mostwidely used is nestin, which defines classes of intermediatefilament proteins; it was first described as the produc<strong>to</strong>f a gene whose expression distinguishes precursors frommore differentiated cells in the neural tube (156) <strong>and</strong> wasfound <strong>to</strong> be specifically expressed in neural progeni<strong>to</strong>rcells (307). More recently, a family of molecular markershas been identified, which includes mouse-Musashi1(m-Msi1), a neural RNA-binding protein expressed predominantlyin proliferating neuronal <strong>and</strong>/or glial precursorcells but not in newly generated postmi<strong>to</strong>tic neurons<strong>and</strong> oligodendrocytes (246, 479).The second category of markers is more specificallyused in research aimed at uncovering the functional roleof adult neurogenesis <strong>and</strong> consists of antigens present incycling cells that are used as proliferative markers. Theyare often used in combination with BrdU, since incorporationof BrdU alone identifies cells undergoing DNAreplication but does not formally provide evidence thatthese cells are actually capable of division. They includeKi-67, a nuclear protein expressed in dividing cells for theentire duration of their mi<strong>to</strong>tic activity, the expression ofwhich is neither linked <strong>to</strong> DNA repair nor <strong>to</strong> apop<strong>to</strong>ticprocesses (142, 259). Although less frequently used, proliferatingcell nuclear antigen (PCNA) allows assessmen<strong>to</strong>f cell proliferation as well. It is an auxiliary protein ofDNA polymerase which is increasingly expressedthrough G 1 , peaks at the G 1 /S interface, <strong>and</strong> decreasesthrough G 2 (146, 162, 294). This marker has been usedsuccessfully <strong>to</strong> study cell proliferation <strong>and</strong> cell cyclelength in conjunction with BrdU cumulative labeling (39).Finally <strong>and</strong> much less used are P34cdc2, a key player inthe initiation of mi<strong>to</strong>sis (415), <strong>and</strong> phosphorylated His<strong>to</strong>neH3 (HH3) expression, which is confined <strong>to</strong> cells inthe G 2 <strong>and</strong> M phases of the cell cycle. In general, cells inthe G 2 phase exhibit punctuate HH 3 nuclear staining thatchanges <strong>to</strong> a more condensed pattern once they enter theM phase (214).B) IMMATURE NEURONS. The mammalian RNA-binding proteinHu, the homolog of the Drosophila neuron-specificRNA-binding protein Elav, is exclusively expressed inpostmi<strong>to</strong>tic neurons (7, 479) <strong>and</strong> thus is frequently usedas an early neuronal marker (31, 414). Other markersinclude neuron-specific class III -tubulin (TuJ1), a cy<strong>to</strong>skeletalprotein expressed in all postmi<strong>to</strong>tic neurons(360, 361), doublecortin (DCX), which encodes a microtubule-associatedprotein expressed in migrating neuroblasts(155, 168, 389), TOAD 64 (turned on after division,also called TUC-4, CRMP4, PUlip1), a cy<strong>to</strong>plasmic proteinexpressed transiently by postmi<strong>to</strong>tic neurons (366), <strong>and</strong>the polysialylated-neuronal cell adhesion molecule PSA-NCAM (277, 498). Another c<strong>and</strong>idate for immature neuronalmarkers is the basic helix-loop-helix (bHLH) transcriptionfac<strong>to</strong>r NeuroD, which seems <strong>to</strong> be expressedfrom a very early stage <strong>to</strong> the stage where new neuronsdevelop dendrites (496, 497). Some of these markers stainnonneuronal cells, <strong>and</strong> some of them (TOAD 64, PSA-NCAM, DCX) are present in the brain, for example, in thepiriform cortex, independently of neurogenesis (113, 389,392, 423, 498, 560), which makes the interpretation ofresults more difficult.C) MATURE NEURONS. The most frequently used specificmarkers for mature neurons are neuron specific enolase(NSE), neuronal specific nuclear protein (NeuN) (386),<strong>and</strong> microtubule-associated protein (MAP-2). However,the identification of the neuronal phenotype of newlygenerated cells using these markers is considered ambiguousas they also label other types of cells (189, 411, 453,454). An alternative approach consists in demonstratingthat adult-born cells do not express any marker for glialcells such as 2,3-cyclic nucleotide (CNP) for oligodendrocytes,<strong>and</strong> calcium binding proteins (S100, S100) orglial fibrillary acidic protein (GFAP) for astrocytes.3. Current identified pitfallsIn addition <strong>to</strong> the problem of neuronal identification,several methodological pitfalls have been identified. First,embryonic <strong>and</strong> postnatal injections of BrdU have beenreported <strong>to</strong> induce physical, morphological, <strong>and</strong> behavioralabnormalities (282, 495). Although only a few studieshave addressed the deleterious effects of BrdU injectionsin adulthood, a neuro<strong>to</strong>xic effect has been reported inaged rats, loss of body weight, <strong>and</strong> deterioration in thestate of the coat being induced by repeated administrationsof 150 mg · kg 1 · day 1 of BrdU for 5 days (130).Together with its early tera<strong>to</strong>genic effects, this suggeststhat incorporation of BrdU in<strong>to</strong> mi<strong>to</strong>tically active cellsmay inhibit cell formation <strong>and</strong> affect stemlike cell population.This raises the as yet unresolved problem thatBrdU may alter the functioning of the labeled cells. Second,changes in neurogenesis detected by variations inBrdU staining may be related <strong>to</strong> a modification in BrdUavailability through an alteration in blood flow or inblood-brain barrier permeability. To rule out such confoundingparameters, using endogenous markers of thecell cycle (Ki67, PCNA, HH 3 , <strong>and</strong> P34cdc2) <strong>and</strong> evaluatingchanges in different brain regions may be helpful. Forbehavioral studies, adequate control groups should beused <strong>to</strong> measure the involvement of a potential modificationof blood flow by exercise. Third, pharmacologicaltreatments or behavioral <strong>and</strong> environment-inducedchanges in neurogenesis may exert their effects by modifyingthe length of the cell cycle, the number of proliferatingcells, or both. However, in most “functional” studies,evaluating the cell cycle parameters is difficult if notimpossible, <strong>and</strong> one might question the relevance of thisDownloaded from physrev.physiology.org on November 25, 2005Physiol Rev • VOL 85 • APRIL 2005 • www.prv.org


NEUROGENESIS IN THE ADULT BRAIN 527“exercise.” Finally, BrdU or [ 3 H]dT labeling might producefalse negatives <strong>and</strong> positives. The most commonlyused dose of BrdU (50 mg/kg) labels only a fraction of theproliferating cells (75), which may result in an apparentabsence of effects of a given experimental setting on cellproliferation. However, multiple injections of this lowdose may overcome this problem. Furthermore, becausethe precursor cell population most certainly does notdivide synchronically, labeling all dividing cells with asingle injection of BrdU, whatever the dose, appears u<strong>to</strong>pist.On the other h<strong>and</strong>, higher doses of BrdU associatedwith an enhanced sensitivity of immunohis<strong>to</strong>chemicalmethods might lead <strong>to</strong> labeling apop<strong>to</strong>tic cells or nonproliferatingcells that synthesize DNA for repair (594), <strong>and</strong>thus <strong>to</strong> false positives. This problem is particularly relevantfor studies on lesions, epilepsy, or ischemia, where ahigh rate of cell death is observed. There are severalpossible ways <strong>to</strong> rule out BrdU labeling of nonproliferativecells: 1) observing mi<strong>to</strong>tic figures a few hours afterBrdU or [ 3 H]dT application; 2) using other endogenouscell cycle markers; 3) using electronic microscopy; 4)analyzing a time course of BrdU labeling (95); 5) combiningBrdU with markers of cell death, although in this casefalse negative results due <strong>to</strong> the downregulation of markersby dying cells cannot be ruled out; 6) using retrovirallabeling (81, 123, 439, 501, 556), which is not devoid ofdisadvantages (the type of cells incorporating the virusmay not be fully controlled, <strong>and</strong> stereotaxic injection,known <strong>to</strong> cause injury, is required); <strong>and</strong> 7) visualizingneurogenic sites in nestin-promoter-GFP transgenic mice(577).4. SummaryThis last decade has been marked by tremendousadvances including the development of BrdU labelingmethods, the discovery of selective markers that differentiateneurons from glial cells, <strong>and</strong> the use of new molecular<strong>and</strong> genetic <strong>to</strong>ols <strong>to</strong> follow the “development” ofthe newly born cells. As we will see in the followingsections, this technical progress has led <strong>to</strong> a lot of research<strong>and</strong> data that have confirmed observations made inthe early 1960s <strong>and</strong> considered at that time by manyspecialists as nonconclusive. However, researchers arenow aware of problems <strong>and</strong> agree that proper interpretationsdepend on accepted rules: 1) [ 3 H]dT <strong>and</strong> BrdUlabeling are not sufficient <strong>to</strong> unambiguously differentiateold neurons from newborn cells, 2) the neuron-specificmarkers presently used must be complemented by appropriatecontrols <strong>to</strong> detect false positives <strong>and</strong> by electrophysiologicalrecordings <strong>to</strong> assert real neuronal phenotypes,<strong>and</strong> 3) the maturation <strong>and</strong> the integration of theadult-born neurons must be followed up.C. Integration of the Newly Born CellsIn<strong>to</strong> Neurogenic Site <strong>Network</strong>s1. The SVZA) IDENTIFICATION OF THE STEMLIKE CELLS. The SVZ, locatedthroughout the lateral wall of the lateral ventricle, harborsthe largest population of proliferating cells in the adultbrain of rodents (12, 448, 511), monkeys (178, 179, 182,248, 286, 434), <strong>and</strong> humans (47, 144), <strong>and</strong> it has beenestimated that 30,000 cells are generated bilaterally dailyin the mouse SVZ (317). This region exhibits a rostrocaudalgradient of proliferative activity: proliferation ishigher in the dorsolateral corner of the rostral ventricle<strong>and</strong> falls caudally, moving from the striatum <strong>to</strong>wards theHF (511). This gradient has been correlated with thecapability of the constitutively proliferative cells <strong>to</strong> divide<strong>and</strong> <strong>to</strong> form neurospheres (381, 493) <strong>and</strong> certainly reflectsthe existence of two populations of dividing cells, one ofquiescent stemlike cells <strong>and</strong> one of rapidly proliferatingprogeni<strong>to</strong>r cells (381, 382). Four cell types have beendescribed in the SVZ: 1) ependymal ciliated cells (type E)facing the lumen of the ventricle, whose function is <strong>to</strong>circulate the cerebrospinal fluid; 2) proliferating type Aneuroblasts, expressing PSA-NCAM, Tuj1, <strong>and</strong> Hu, <strong>and</strong>migrating in “chains” <strong>to</strong>ward the olfac<strong>to</strong>ry bulb (OB); 3)slowly proliferating type B cells expressing nestin <strong>and</strong>GFAP, <strong>and</strong> unsheathing migrating type A neuroblasts; <strong>and</strong>4) actively proliferating type C cells or “transit amplifyingprogeni<strong>to</strong>rs” expressing nestin, <strong>and</strong> forming clusters interspacedamong chains throughout the SVZ (15, 55, 122,124, 166, 220, 474).Although it has been proposed that the stemlike cellsmay be ependymal type E cells, which were shown <strong>to</strong>divide in vivo <strong>and</strong> <strong>to</strong> differentiate in<strong>to</strong> neurons in the OB(242), this view has been challenged by van der Kooy <strong>and</strong>colleagues (85) who have found that ependymal cellsgenerating spheres do not have the ability <strong>to</strong> self-renew or<strong>to</strong> produce neurons, <strong>and</strong> by Capela <strong>and</strong> Temple (79) whohave shown that ependymal cells do not form neurospheres.It is more likely that the type B cells represent astemlike cell type (123, 124). Indeed, after 1 wk of intracerebroventricular(icv) infusion of the antimi<strong>to</strong>tic drugcy<strong>to</strong>sine--D-arabinofuranoside (AraC), type C <strong>and</strong> type Acells are eliminated, whereas type B cells continue <strong>to</strong>divide. Between 1 <strong>and</strong> 2 days after treatment cessation,type C cells reappear, followed 2 days later by type Acells, suggesting that type B cells give rise <strong>to</strong> type C cells,which in turn generate neuroblasts (125). Furthermore,following the targeted introduction of a retrovirus in<strong>to</strong>GFAP-positive cells, which include type B cells, labeledcells migrate <strong>to</strong>wards the OB where they give rise <strong>to</strong> newneurons (123). Thus the lineage progression B 3 C 3 Ahas been proposed (123, 124). However, Doetsch et al.(126) have also shown that after exposure <strong>to</strong> high concentrationsof EGF, the type C cells retain stemlike cellDownloaded from physrev.physiology.org on November 25, 2005Physiol Rev • VOL 85 • APRIL 2005 • www.prv.org


528 ABROUS, KOEHL, AND LE MOALproperties (126). In fact, there are opposing views onwhether neurospheres are derived from GFAP-positivecells (type B cells) or from fast proliferating type C cells(225, 380) (see Fig. 1).B) MIGRATION OF THE NEWLY BORN CELLS THROUGH THE ROSTRALMIGRATORY STREAM. The newly generated cells originatingfrom different levels of the SVZ migrate in chains rostrally,up <strong>to</strong> 5 mm in rodents <strong>and</strong> <strong>to</strong> 20 mm in monkeys, <strong>to</strong>reach the OB (122, 286, 317). This migration, which followsthe rostral migration stream (RMS) <strong>and</strong> requires 2–6days in rodents (220, 317, 325), involves PSA-NCAM (100,220, 418, 474, 540) <strong>and</strong> a chemorepulsion mechanismthrough Slit-Robo signaling (574). Although a role of theOB as a chemoattractant structure has been suggested, itsinvolvement in proliferation <strong>and</strong> guidance of the newlyborn cells still remains unclear. Indeed, whereas OB removal(275), transection of the olfac<strong>to</strong>ry peduncle (231),or unilateral nostril occlusion (96) does not prevent SVZprecursors from proliferating <strong>and</strong> migrating <strong>to</strong>wards theOB, a transection through the RMS (9) or a removal of therostral OB (314) impedes neuroblast migration. It has thusbeen proposed that a diffusible attractant is secreted inspecific layers in the OB, including the glomerular layer(314).After reaching the middle of the OB, the newborncells detach from chains, migrate radially, <strong>and</strong> progressin<strong>to</strong> one of the overlying cell layers whereupon they undergoterminal differentiation. Neuroblast detachmentfrom chains is initiated by Reelin <strong>and</strong> tenascin, whereasradial migration depends on tenascin-R (198, 478).C) LIFE AND DEATH OF THE NEWLY BORN CELLS. Massive celldeath has been observed during the first 2 mo after a BrdUpulse (572). This elimination mechanism is prominent inthe OB compared with the RMS <strong>and</strong> the SVZ (39, 50, 439)<strong>and</strong> may maintain constant OB cell number by a continuouscell turnover, as was suggested during earlier development(419). The number of newly born cells that survive1 mo after a BrdU pulse (50 mg · kg 1 · day 1 of BrdUfor 4 consecutive days in 2-mo-old female wistar rats) hasbeen estimated <strong>to</strong> be 60,000 per granule cell layer in onestudy (50) <strong>and</strong> 120,000 in another (572). In the latter case,it was shown that 50% of the newly generated neurons(80,000 per granule cell layer <strong>and</strong> 800 per periglomerularlayer) that survived the initial period of cell deathsurvived for at least 19 mo (572), confirming earlier work(253). With the use of retroviral labeling of precursors inthe SVZ, it was confirmed that one-half of the labeled cellsdied shortly after their arrival in the OB (between 15 <strong>and</strong>45 days after neuronal birth) <strong>and</strong> that most dying cellswere mature, harboring dendritic arborization, <strong>and</strong> receivingconnections (439). It was further shown in thisstudy that survival of the newly generated granule cellsdepends on sensory input.D) DIFFERENTIATION OF THE NEWLY BORN CELLS IN THE OLFACTORYBULB. The newly born cells differentiate mainly in<strong>to</strong> neuronsthat grow dendritic trees <strong>and</strong> differentiate in<strong>to</strong> twotypes of intrabulbar interneurons. Most of the cells (75–99%) differentiate in<strong>to</strong> GABA granule cells (GCs),whereas a smaller number (1–25%) differentiate in<strong>to</strong> periglomerularcells expressing GABA <strong>and</strong>/or tyrosine hydroxylase(38, 81, 256, 326, 439, 475, 572). Several monthsafter their birth, 10,000 GABAergic granule neurons, 100periglomerular GABAergic neurons, <strong>and</strong> 60 new dopa-Downloaded from physrev.physiology.org on November 25, 2005FIG. 1. <strong>Neurogenesis</strong> in the subventricularzone (SVZ)/olfac<strong>to</strong>ry bulb (OB) system. Top panel:a sagittal view of a rodent brain showing the sites ofneurogenesis in the SVZ/OB system is given. Cellsproliferate mainly in the SVZ, migrate along therostral migra<strong>to</strong>ry stream (RMS) <strong>to</strong> reach the OB,where they migrate radially <strong>and</strong> undergo terminaldifferentiation. Bot<strong>to</strong>m panel: a sequence of celltypes involved in neuronal lineage <strong>and</strong> specificmarkers allowing cell identification are presented.Markers appearing in bold are specific <strong>to</strong> each stage(see text for further details).Physiol Rev • VOL 85 • APRIL 2005 • www.prv.org


530 ABROUS, KOEHL, AND LE MOALThe clusters of dividing cells have been shown <strong>to</strong>include several cell types: neural precursors, committedneuroblasts, glial precursors, <strong>and</strong> endothelial precursors(angioblasts representing 37% of proliferative cells) thatare proximal <strong>to</strong> vessels (256, 363, 424). This suggests thatneurogenesis most probably occurs within the context ofvascular recruitment. Within this niche, endothelial cellsmay constitute a critical regula<strong>to</strong>r for self-renewal ofstemlike cells (505) through the release of trophic fac<strong>to</strong>rs(see sect. IID2C <strong>and</strong> Table 1).In the rodent DG, the vast majority of newly borncells are postmi<strong>to</strong>tic <strong>and</strong> at least partially differentiatedbetween 3 <strong>and</strong> 7 days (111, 416), since during this periodBrdU-labeled cells express NeuroD (417). The cells thatdo not terminally differentiate die within 1 wk of theirgeneration, a process affecting 60% of the newborn cells(111, 204). In the macaque, a similar time course has beendemonstrated with an initial rise in BrdU-labeled cellsfollowed by a fall after 5 wk (182). Interestingly, the GCLharbors a much lower number of proliferating cells comparedwith the SVZ. Thus, in a study performed in 9- <strong>to</strong>10-wk-old rats, 9,000 newborn cells were found <strong>to</strong> begenerated per day (75). In a more recent study, only4,000 new cells, out of which 3,000 were found <strong>to</strong> benew neurons, were shown <strong>to</strong> be added daily in the DG of4-mo-old rats (456); this discrepancy in the number ofnewborn cells is certainly linked <strong>to</strong> the difference in theage of the animals used in the studies, as the productionof new neurons in the DG diminishes with age (see sect.IIIC1A). In the macaque, the number of newborn cells perday (200) represents 0.004% of one GCL (284).C) FATE OF THE NEWLY BORN CELLS. The newborn cells differentiatemainly in<strong>to</strong> neurons in the GCL. In his originalFIG. 2. <strong>Neurogenesis</strong> in the hippocampalsystem. Top panel: a frontalview of a rodent brain showing the sitesof neurogenesis in the dentate gyrus(DG) of the hippocampal formation (HF)is given. The HF forms a trisynaptic network:multimodal inputs are transferredfrom neocortical regions <strong>to</strong> the DG viathe en<strong>to</strong>rhinal cortex; information is thentransferred from the DG <strong>to</strong> CA3 throughthe mossy fibers, onward through theSchaffer collaterals <strong>to</strong> CA1 (<strong>and</strong> the subiculum)<strong>and</strong> then via the en<strong>to</strong>rhinal cortexback out <strong>to</strong> the cortical associativeareas (16). Cells proliferate in the subgranularlayer (SGL) located at the interfacebetween the hilus <strong>and</strong> the granularlayer (GL), where they migrate <strong>and</strong> differentiatein<strong>to</strong> mature neurons. Bot<strong>to</strong>mpanel: a sequence of cell types involvedin neuronal development, along with specificmarkers allowing cell identification,is proposed [see text <strong>and</strong> recent reviewfrom Kempermann et al. (265) for furtherdetails].study using electron microscopy, Kaplan <strong>and</strong> co-workers(249–252) reported that the newborn cells exhibit theultrastructural characteristics of neurons. Then, theywere shown <strong>to</strong> express neuronal markers. During the first2 days after their birth, most BrdU-labeled cells expressnestin (95), then TuJ1 (75) or TOAD-64 (75, 536) in thefollowing 3 days, <strong>and</strong> DCX between 1 <strong>and</strong> 14 days aftertheir generation (65). DCX-BrdU-colabeled cells show featuresof progeni<strong>to</strong>r cells as some of them coexpress Ki-67<strong>and</strong> are thus still able <strong>to</strong> divide (58, 154, 521). This rapidpostmi<strong>to</strong>tic status of new cells has been confirmed withcalretinin, a calcium binding protein that is transientlyexpressed by postmi<strong>to</strong>tic cells, the expression of which isobserved as early as 1 day after labeling dividing cells, apeak being reached after 1 wk (58). Although newly borncells express NeuN as early as 72 h after their generation(58, 193), only half of the BrdU-IR cells expressed NeuNover 3 wk (65). Using NSE, half of the newly born cellswere found <strong>to</strong> be neurons 2 wk after their birth (78).Calbindin is expressed later, by 3 wk after birth (180, 290,536, 537). A low percentage of adult-born cells differentiatesin<strong>to</strong> astrocytes (GFAP/S100), <strong>and</strong> rare are thosethat adopt a microglial or oligodendrocyte phenotype(521). It should be emphasized that even 4 mo after theirlabeling a nonnegligible proportion of BrdU-IR cells havean unknown phenotype. In monkeys, most newly generatedcells are claimed <strong>to</strong> be neurons as well since theyexpress TuJ1, TOAD-64, NeuN, NSE, <strong>and</strong> calbindin, <strong>and</strong>rarely markers of astrocytes (GFAP) or oligodendrocytes(CNP) (178, 182, 284). It was first thought that the newlyborn cells did not survive, thereby constituting a continuouslyrefreshed pool of neurons (189). However, it wasDownloaded from physrev.physiology.org on November 25, 2005Physiol Rev • VOL 85 • APRIL 2005 • www.prv.org


NEUROGENESIS IN THE ADULT BRAIN 531Extrinsic fac<strong>to</strong>rs regulating in vivo cell proliferation <strong>and</strong> neurogenesis in the subventricularzone/olfac<strong>to</strong>ry bulb system <strong>and</strong> the dentate gyrus of the hippocampal formation under physiological conditionsTABLE 1.Subventricular Zone/Olfac<strong>to</strong>ry BulbDentate GyrusFac<strong>to</strong>rsCellproliferation <strong>Neurogenesis</strong>NeuronaldifferentiationLong-termsurvivalCellproliferation <strong>Neurogenesis</strong>NeuronaldifferentiationLong-termsurvivalReference Nos.Hormones <strong>and</strong> peptidesCorticosteroneSuppression (adx) 0 m m 0 0 71, 74, 176, 465Cort. treatment n n 0 17, 71, 76, 212EstradiolEstrus cycle 0 mproE 0 n 536OVX 0 n 29, 536OVX acute 17-E m m then n 0 29, 422, 536OVX chronic 17-E 0 0 0 43517-E m (4h) 421Prolactin m m 0 506PACAP m m 362NeurosteroidsDHEA m m 0 0 254PregS m m m 349Allopreg n 349NeurotransmittersGlutamateEnt. Cx. lesion m 73NMDA activation n 73, 76NMDA blockade m m 46, 73, 76, 177, 390AMPA potentiation m 27mGluR II blockade m 581Sero<strong>to</strong>nin5-HT depletion n n 29, 61Lesion graft m 63Reuptake inhibition 0 m m 0 0 10, 335, 4835-HT 1A blockade n 4515-HT 1A activation m m 0 m m 0 30, 4835-HT 1B blockade m 0 305-HT 1B activation m 05-HT 2A/2c blockade 0 n5-HT 2A/2c activation m 05-HT 2c blockade 0 05-HT 2c activation m m 0 0 0NorepinephrineDepletion 0 n n 0 0 292Reuptake inhibition m 335No donor m m m 587Growth or trophic fac<strong>to</strong>rsbFGF m m m 0 0 0 238, 240, 291EGF m n n 0 n n 291HB-EGF m m 0 m m 236, 238, 240TGF- m 98IGF-I m m (ovx) 0 or m (hpx) 0 1, 310, 435, 542BDNF m m 0 593CNTF m m m m 141VEGF m m 241Morphogenic fac<strong>to</strong>rsShh m m 0 0 295BMP n n n 311Noggin m m m 311Vitamins <strong>and</strong> retinoidsVitamin EDeficiency m n 86, 87Supplementation (<strong>to</strong>copherol) n m m 83, 102Retinoic acid (chronictreatment)n n n n n 99Downloaded from physrev.physiology.org on November 25, 2005Adx, adrenalec<strong>to</strong>my; Cort, corticosterone; OVX, ovariec<strong>to</strong>my; 17-E, 17-estradiol; proE, proestrus; PregS, pregnenolone sulfate; Allopreg,allopregnanolone; Ent. Cx., en<strong>to</strong>rhinal cortex; Hpx, hypophysiec<strong>to</strong>my; 0, no change; blank case, not determined; m, increase; n, decrease.Physiol Rev • VOL 85 • APRIL 2005 • www.prv.org


532 ABROUS, KOEHL, AND LE MOALmore recently found that they do not have an ephemeralexistence <strong>and</strong> survive for at least 11 mo (263).Recently, it has been shown that beside glutamatergicgranule neurons, a small percentage of newly borncells (14%) differentiate in<strong>to</strong> GABAergic basket cells(316). Furthermore, under specific circumstances (ischemiaor neonatal kainate administration), pyramidal cellsin Ammon’s horn are also generated (see also sect. IIID1B;Refs. 128, 399, 489). However, the progeni<strong>to</strong>rs responsiblefor the regenerating pyramidal neurons originate from theperiventricular region rather than from the SGZ (399).D) INTEGRATION OF THE NEWLY BORN CELLS. The newborn cellsintegrate the GCL 4–10 days after their generation. Asthey form dendrites, they receive synaptic contacts <strong>and</strong>extend axons in<strong>to</strong> the CA3 region, suggesting that theymake synapses long before being fully mature (75, 204,252, 344, 517). Indeed, using virus-based transynaptic activityneuronal tracing (PVR GS518), the neurons generatedin the DG have been shown <strong>to</strong> be synaptically integratedin<strong>to</strong> the preexisting circuitry 4–8 wk after theirbirth (80), whereas they reach a mature morphology(soma size, <strong>to</strong>tal dendritic length, dendritic branching,<strong>and</strong> spine density) only 4 mo after their birth (513).E) FUNCTIONAL PROPERTIES OF THE NEWLY BORN CELLS. When stilllocated at the interface of the hilus, the newly born cellsexhibit electrophysiological properties characteristic ofimmature neurons: they are completely unaffected byGABA A recep<strong>to</strong>r inhibition, <strong>and</strong> they exhibit paired-pulsefacilitation, have a lower threshold for induction of longtermpotentiation (LTP), <strong>and</strong> display robust LTP (564).With the use of nestin-promoter GFP transgenic mice,type I cells (putative B cells) were found <strong>to</strong> have low inputresistance values, whereas the type II cells (type D cells,expressing PSA-NCAM) exhibited higher input resistance<strong>and</strong> voltage-dependent sodium currents (159). Recently,PSA-NCAM expressing cells have been shown <strong>to</strong> differfrom mature neurons in their passive (input resistance)<strong>and</strong> active membrane properties (such as calcium spikesthat boost fast sodium action potentials) <strong>and</strong> in theirenhanced ability <strong>to</strong> develop LTP (490). One month aftertheir birth, the “newborn” neurons, labeled by a retroviralvec<strong>to</strong>r expressing GFP, exhibit some electrophysiologicalproperties (input resistance, threshold potential for spiking<strong>and</strong> firing) similar <strong>to</strong> those of mature granule neurons(556). The stimulation of the perforant pathway, the mainexcita<strong>to</strong>ry afference <strong>to</strong> the DG, elicits responses in “newborn”GFP-labeled cells, indicating that they receive functionalsynaptic inputs <strong>and</strong> are therefore functionally integratedin<strong>to</strong> the preexisting network. However, a depressionof synaptic currents is evoked in these cells followingthe paired-pulse stimulation of the perforant pathway,whereas a facilitation response is observed in maturecells, a discrepancy attributed <strong>to</strong> differences in presynapticrelease properties (556). The functionality of the adultborngranule cells has been further demonstrated byshowing, using c-fos, that 40% of 1-mo-old newborn granuleneurons were able <strong>to</strong> respond <strong>to</strong> various chemoconvulsants(232, 234). In contrast, following a more physiologicalform of hippocampal stimulation consisting in ahippocampal-dependent learning task, only 3% of cells areactivated (232). Finally, besides these functional excita<strong>to</strong>ryadult-born granule cells, it has been shown thatGABAergic newborn basket cells form functional inhibi<strong>to</strong>rysynapses with the granule cells (316). This discoveryraises the question of the mechanisms <strong>and</strong> fac<strong>to</strong>rs involvedin the production of excita<strong>to</strong>ry granule neuronsversus inhibi<strong>to</strong>ry interneurons.3. The cortexAlthough very controversial, the existence of adultneurogenesis in the cortex deserves attention. Indeed,Altman <strong>and</strong> co-workers in the 1960s (11, 12, 14) <strong>and</strong>Kaplan in the early 1980s (247–249) reported evidence forneurogenesis in the cortex of rats <strong>and</strong> cats. More recently,neurogenesis has been described in the rat anterior neocortex(182) <strong>and</strong> in the prefrontal, inferior temporal, <strong>and</strong>posterior parietal cortex of macaques (179, 182). Within 2wk after BrdU injection(s), the number of BrdU-labeledcells increased <strong>and</strong> then fell after 5 wk, indicating that alarge number of newly born cells died. The surviving cellswere assumed <strong>to</strong> be neurons since they extended axonslocally <strong>and</strong> expressed markers of immature (TOAD 64,TuJ1) <strong>and</strong> mature (38–52% of BrdU-NeuN cells) neuronsbut did not express GFAP or CNP. Although the site oforigin of the newly born neurons remains unclear, it hasbeen proposed that they may be generated in<strong>to</strong> the SVZ<strong>and</strong> migrate <strong>to</strong> neocortical areas through the white matteror may be recruited from local quiescent stemlike cells(179, 182, 284).In contrast <strong>to</strong> these data, other groups have reporteda complete absence of neurogenesis in the mouse (134,332) <strong>and</strong> monkey cortex (281, 453). <strong>Neurogenesis</strong> wasobserved in the neocortex of mice only following a targetedapop<strong>to</strong>tic degeneration of corticothalamic neurons(332), <strong>and</strong> in this case, endogenous neural precursorsmigrated <strong>and</strong> differentiated in<strong>to</strong> neocortical neurons in alayer- <strong>and</strong> region-specific manner <strong>and</strong> reformed appropriatelong-distance corticothalamic connections; thus theyappeared <strong>to</strong> reconstruct the lesioned circuits. In the primateneocortex, although cell proliferation occurs, thenewly born cells do not seem <strong>to</strong> differentiate in<strong>to</strong> neurons(285). These discrepancies may be due <strong>to</strong> differences inhousing conditions, the animals’ his<strong>to</strong>ries, <strong>and</strong> geneticbackground, <strong>and</strong> other technical considerations (survivaltimes, BrdU dosage <strong>and</strong> administration mode, immunohis<strong>to</strong>chemistry.. .), but generally, the existence of thiscortical neurogenesis is still a matter of debate (411).Downloaded from physrev.physiology.org on November 25, 2005Physiol Rev • VOL 85 • APRIL 2005 • www.prv.org


NEUROGENESIS IN THE ADULT BRAIN 5334. SummaryFollowing a century of doubt <strong>and</strong> controversies,there is now a consensus that neurogenesis occurs in theadult brain in at least two regions, the SVZ <strong>and</strong> the DG. Inboth structures, stemlike cells proliferate, migrate, <strong>and</strong>differentiate mainly in<strong>to</strong> granule neurons that will synthesizeGABA in the OB <strong>and</strong> glutamate in the DG. Althoughless studied, a small proportion of adult-born cells differentiatein<strong>to</strong> other types of interneurons (the periglomerularin the OB <strong>and</strong> the basket cells in the DG). Al<strong>to</strong>getherthis adult neurogenesis leads <strong>to</strong> the birth of 30,000 newneurons per day in the SVZ <strong>and</strong> between 3,000 <strong>and</strong> 9,000in the DG of young adult rats, depending on their age. Thereasons for which, 1) the SVZ <strong>and</strong> the DG harbor adultneurogenesis, 2) neurogenesis is curtailed in the DG comparedwith the OB, <strong>and</strong> 3) genesis rates are much lower inprimates, are currently unknown. Furthermore, becausethese structures do not grow in size, a homeostatic compensa<strong>to</strong>ryequilibrium must be attained through an increasein cell death that must be equivalent <strong>to</strong> the initialaddition of neurons. This poorly unders<strong>to</strong>od phenomenon,in particular in the DG, deserves more attention forit is an important partner of neurogenesis. Finally, recentevidence indicates that the adult-born neurons of the OB<strong>and</strong> the DG are functional <strong>and</strong> thus play a physiologicalrole. Although these findings suggest a relevant contributionof these newly generated neurons <strong>to</strong> the bulbar orhippocampal function, further studies are needed <strong>to</strong> confirmthese reports <strong>and</strong> fully unravel their fundamentalconsequences on the animals’ behavior (see sect. III).D. Fac<strong>to</strong>rs Regulating <strong>Adult</strong> <strong>Neurogenesis</strong>Although various fac<strong>to</strong>rs that affect the division, migration,<strong>and</strong> differentiation of neural precursor cells havebeen isolated, the precise mechanisms that control neuronalfate in the adult nervous system remain largelyunknown. Both cell intrinsic programs <strong>and</strong> extracellular/environmental fac<strong>to</strong>rs, which we will review below, are atplay.1. Intrinsic programs controlling neurogenesisTwo fundamental decisions are involved in order fora precursor cell <strong>to</strong> generate a neural cell. The first one is<strong>to</strong> decide whether <strong>to</strong> self-renew or <strong>to</strong> undergo mi<strong>to</strong>ticarrest, <strong>and</strong> the second is <strong>to</strong> interpret mi<strong>to</strong>tic arrest, usingcell au<strong>to</strong>nomous cues that direct <strong>to</strong>ward a particular fate.Much of our current underst<strong>and</strong>ing of these cell intrinsicprograms comes from work performed when neurogenesisis at its best, i.e., during development. However, it isimportant <strong>to</strong> emphasize that the rules that govern neuronalspecification during development may not be the samein the adult brain. Furthermore, since a description of thedetailed molecular mechanisms involved in cell proliferation<strong>and</strong> determination is beyond the scope of this review,we here propose only a brief overview of the intrinsicfac<strong>to</strong>rs involved, with a special mention whenevertheir involvement has been extended <strong>to</strong> the adult neurogeniczones.A) CONTROL OF CELL PROLIFERATION. Among the cell cyclefac<strong>to</strong>rs regulating cellular proliferation, Rb (retinoblas<strong>to</strong>ma)<strong>and</strong> its related proteins (p107, p130), necdin, <strong>and</strong>the E2F protein families are key players (for a review, seeRef. 580). Thus, during the G 1 phase, Rb predominates ina hypophosphorylated form that can bind <strong>to</strong> E2F, a positiveregula<strong>to</strong>r of the cell cycle (403), thereby repressingits transcriptional activity <strong>and</strong> preventing the cells fromentering the S phase. In cycling cells, phosphorylated Rbaccumulated during the late G 1 phase releases E2F, thusallowing S phase entry. This phosphorylation depends onthe activation of cyclin-dependent kinases (CDK) actingsequentially. Early <strong>to</strong> mid-G 1 , cyclins of the D class (D1,D2, <strong>and</strong> D3) activate CDK4/CDK6, <strong>and</strong> in late G 1 , cyclin Eactivates CDK2, leading <strong>to</strong> hyperphosphorylation of theRb protein. Two families of CDK inhibi<strong>to</strong>rs (CDKIs) thatcan suppress cell proliferation by inhibiting Rb phosphorylationhave been identified (566): members of the inhibi<strong>to</strong>rsof CDK4 family (INK4 family), including p15 Ink4b , p16 Ink4a ,p18 Ink4c , <strong>and</strong> p19 Ink4d , <strong>and</strong> members of the kinase inhibi<strong>to</strong>ryprotein family (Cip/Kip family) such as p21 Waf1/Cip1 , p27 Kip1or p57 Kip2 (see Fig. 3).It is now clear that the Rb <strong>and</strong> E2F protein familiesare differentially expressed in proliferative <strong>and</strong> postmi<strong>to</strong>ticcells of the adult brain. Thus, in the two neurogenicsites, the DG <strong>and</strong> the SVZ, Rb immunoreactivity is high inproliferating neuronal precursors <strong>and</strong> reduced during terminaldifferentiation (415), which implies that the transientincrease in the level of Rb is an important step in theinitiation of terminal mi<strong>to</strong>sis in neuronal progeni<strong>to</strong>rs.Moreover, the Rb protein family was found <strong>to</strong> be essentialfor the development of a neural lineage <strong>and</strong> the exit fromthe cell cycle, whereas it does not seem <strong>to</strong> be involved inthe maintenance of postmi<strong>to</strong>tic neurons (69, 89, 228, 299).The role of E2F1 as a key fac<strong>to</strong>r in regulating adultneurogenesis has been further emphasized in a recentstudy performed in mice lacking E2F1. These mice, whenadult, exhibit a lower level of cell proliferation <strong>and</strong> areduction in the number of neurons generated in adultneurogenic areas (94).The role of cell cycle regula<strong>to</strong>rs in the control ofneuron production has been studied as well. Transgenicmice that lack p27 Kip1 expression display a higher rate ofcell proliferation versus differentiation in the SVZ, leading<strong>to</strong> an increased number of type C cells, a reduced numberof type A neuroblasts, <strong>and</strong> no change in the number oftype B cells (127). Finally, distinct functions for CDKinhibi<strong>to</strong>rs, either in the control of cell cycle exit <strong>and</strong>differentiation during neurogenesis (respectively, p27 Kip1Downloaded from physrev.physiology.org on November 25, 2005Physiol Rev • VOL 85 • APRIL 2005 • www.prv.org


534 ABROUS, KOEHL, AND LE MOAL<strong>and</strong> p19 Ink4d ) or in the maintenance of a quiescent state inneural progeni<strong>to</strong>rs (p18 Ink4c ) or neurons (p21 Cip1 ) inadults have been underlined (303).B) CONTROL OF CELL FATE. Data from developmental biologyhave clearly indicated that a core genetic programinvolving multiple bHLH transcription fac<strong>to</strong>rs is requiredfor both neuronal differentiation <strong>and</strong> determination.These fac<strong>to</strong>rs, deriving from proneural <strong>and</strong> neurogenicgenes, antagonistically control the switch from cell proliferation<strong>to</strong> neural differentiation: cascades of neuronalbHLH genes promote differentiation, whereas antineuronalbHLH genes repress them under the control of Notch<strong>and</strong> keep cells at a precursor stage (for recent generalreviews, see Refs. 358, 473).Two classes of proneural genes can be distinguished:the determination fac<strong>to</strong>rs, such as Mash1 (mammalianachaete-scute homolog), Math1 (mammalian a<strong>to</strong>nal homolog),<strong>and</strong> Ngns (Neurogenins) including Ngn2, expressedearly in mi<strong>to</strong>tic neural precursor cells, <strong>and</strong> thedifferentiation fac<strong>to</strong>rs, including NeuroD, NeuroD2, <strong>and</strong>Math2, expressed later in postmi<strong>to</strong>tic cells (for reviews,see Refs. 49, 379). It was recently determined that theseproneural genes are downstream effec<strong>to</strong>rs of Pax6 (for areview, see Ref. 174), a transcription fac<strong>to</strong>r that promotesneurogenesis (210).These proneural genes are components of a cell-cellsignaling mechanism whereby a cell that becomes committed<strong>to</strong> a neural fate inhibits its neighbor from doinglikewise. This process of lateral inhibition, which restrictsthe domains of the proneural gene activity <strong>and</strong> involvesneurogenic genes, is mediated by the Notch pathway (forreviews, see Refs. 23, 32, 161, 244, 491, 567). Among theeffec<strong>to</strong>rs of Notch, three families of negative regula<strong>to</strong>rs ofFIG. 3. Schematic representation ofthe mammalian cell cycle. The cyclinD/cdk4–6 complex phosphorylates the Rbprotein leading <strong>to</strong> sequential phosphorylationby cyclinE/cdk2 <strong>and</strong> the release of freeE2F. Phosphorylation of Rb relieves transcriptionalrepression of genes involved inthe induction of S-phase entry. The abilityof the cyclin/cdk enzymes <strong>to</strong> phosphorylateRb is inhibited by cyclin-dependentkinase inhibi<strong>to</strong>rs (cdkis), the activation ofwhich thus suppresses cell proliferation.The basic helix-loop-helix (bHLH) proneuralgenes NeuroD, Mash1, <strong>and</strong> Ngn1 controlthe switch from cell proliferation <strong>to</strong>neural differentiation by activation of theKip/Cip Cdki family, thus preventing progressionthrough the G 1 -S phases. The HLHproteins Id1 <strong>and</strong> Id3 (inhibi<strong>to</strong>r of differentiation)repress neuronal determination bydirect binding with proneural bHLH proteins,but also favor progression throughthe cell cycle via inhibition of the INK4 <strong>and</strong>Kip/Cip Cdki families. Activating <strong>and</strong> inhibitinginfluences are represented by blue arrows<strong>and</strong> red lines, respectively.the bHLH transcription fac<strong>to</strong>rs are known: the HES, Id,<strong>and</strong> HES-related (HESR or HERP) families (226, 413,481, 545). These effec<strong>to</strong>rs repress neuronal determination<strong>and</strong> differentiation in those cells not destined <strong>to</strong>become neuroblasts. Paralleling this regulation of neuronalproliferation by inhibiting neuronal fate, Notchwas also found <strong>to</strong> be involved in determining glial fate(186, 398, 467).Recent studies have highlighted that some of thesefac<strong>to</strong>rs, which are at play during development, may beinvolved in neuronal cell fate during adulthood. ThusNotch1 <strong>and</strong> Hes5 were found <strong>to</strong> be expressed at highlevels <strong>and</strong> with a similar pattern in both the adult SVZ<strong>and</strong> DG, whereas numb <strong>and</strong> numblike, which negativelyregulate the Notch signal transduction, were not, suggestingan active Notch signal in the adult neurogeniczones (523). Similarly, the mRNA expression of severalbHLH fac<strong>to</strong>rs was found <strong>to</strong> be present at various degreesin the adult HF, ranging from the restricted expressionof Mash1 within the proliferative SGZ, which is consistentwith its role in maintaining a precursor cell phenotype,<strong>to</strong> the widespread profile of Hes5 throughout theHF (140).In conclusion, despite the considerable advancesachieved recently with the development of molecular<strong>to</strong>ols, the complex intrinsic machinery that controls <strong>and</strong>coordinates proliferation <strong>and</strong> differentiation is still poorlyunders<strong>to</strong>od. As is the case for hema<strong>to</strong>poiesis, for whichmuch progress has been achieved, underst<strong>and</strong>ing thegene expression patterns of progeni<strong>to</strong>r cells <strong>and</strong> theirprogeny will be a critical step in elucidating the mechanismsunderlying cell fate.Downloaded from physrev.physiology.org on November 25, 2005Physiol Rev • VOL 85 • APRIL 2005 • www.prv.org


NEUROGENESIS IN THE ADULT BRAIN 5352. Extrinsic fac<strong>to</strong>rs regulating neurogenesisA) HORMONES AND NEUROSTEROIDS. I) Adrenal corticosteroids.His<strong>to</strong>rically, corticosteroids were the first fac<strong>to</strong>rs<strong>to</strong> be studied for their influence on adult neurogenesis(354). Corticosteroids are released in<strong>to</strong> the blood circulationfollowing the activation of the hypothalamo-pituitary-adrenal(HPA) axis, primarily by stress (351). Corticosterone,the main corticosteroid in rodents, regulatesits own secretion through negative feedback, by interactingwith two recep<strong>to</strong>rs (the mineralocorticoid MR, theglucocorticoid GR), present in the DG. Suppression ofcorticosterone secretion after bilateral adrenalec<strong>to</strong>my(adx) increases glial <strong>and</strong> neuronal births in the DG (71,176), whereas mi<strong>to</strong>tic activity in the SVZ remains unchanged(465), suggesting a site-specific inhibi<strong>to</strong>ry influenceof corticosteroids. It was further shown that proliferationincreases within 24 h after adx <strong>and</strong> remains constan<strong>to</strong>ver the 6 subsequent days; the newly generatedcells survive for at least 4 wk in the absence of corticosterone,indicating that their survival is corticosteroneindependent (465). Cell death is also enhanced, but thepopulations of cells undergoing mi<strong>to</strong>sis or apop<strong>to</strong>sis aredistinct: immature cells divide at the interface of the hilus,whereas more mature neurons, located at the interface ofthe molecular layer, die (72). These alterations are preventedby corticosterone replacement (176, 465). The respectiveroles of MR <strong>and</strong> GR on adrenalec<strong>to</strong>my-inducedstructural modifications have recently been examined(374). Treatment with a low dose of the MR agonist,aldosterone, prevents adrenalec<strong>to</strong>my-induced increase incell death, whereas a higher dose is necessary <strong>to</strong> normalizecell proliferation. Furthermore, treatment with a GRagonist, RU 28362, at doses that should fully occupy thisrecep<strong>to</strong>r prevents both adrenalec<strong>to</strong>my-induced cell death<strong>and</strong> birth. Thus the stimulation of both MR <strong>and</strong> GR mediatesthe effects of corticosterone on cell proliferation<strong>and</strong> protects mature cells from cell death.This inhibi<strong>to</strong>ry action of corticosterone on neurogenesishas also been found in other models: 1) acute corticosteroneadministration {2 40 mg/kg administered 1 hbefore a single [H 3 ]dT injection (71, 76); implantation of a200-mg corticosterone pellet followed by 1 200 mg/kgBrdU (17)}, 2) chronic corticosterone treatment [1 40mg/kg for 21 days; BrdU 10 50 mg/kg every 12 h on days17–21 (212)], 3) stress (see sect. IIIB1), <strong>and</strong> 4) interindividualdifferences in the activity of the HPA axis (305; seesect. IIIB2).The mechanisms by which corticosterone hamperscell proliferation remain unknown. In vitro, glucocorticoidsblock the G 1 phase of the cell cycle, notably throughrepression of cyclin D1, cyclin D2, Cdk4 <strong>and</strong> Cdk6, <strong>and</strong>induction of the CDKIs p21 Cip1 <strong>and</strong> p27 Kip1 (438, 585, 592).However, it remains <strong>to</strong> be determined which of thesemechanisms is involved in vivo. Because only a smallminority of precursor cells express corticosteroid recep<strong>to</strong>rs(77), corticosterone may not act directly on proliferatingprecursors, although it cannot be excluded thatimmature forms of recep<strong>to</strong>rs that are not recognized bycurrent antibodies are involved. Corticosterone could ac<strong>to</strong>n neighboring glial or neuronal cells expressing corticosteroidrecep<strong>to</strong>rs, which could control the cell cycle byreleasing growth fac<strong>to</strong>rs (515). Alternatively, corticosteronecould regulate proliferation indirectly by increasingglutamate release in the DG (520) (see sect. IID2B) astheeffects of adx or high levels of corticosterone can beblocked by NMDA recep<strong>to</strong>r activation or inactivation,respectively (76). Corticosterone may also inhibit cellproliferation by dowregulating the production of insulinlikegrowth fac<strong>to</strong>r I (IGF-I) (585, 592).II) Gonadal hormones. The influence of female gonadalhormones has been investigated in the DG sinceestrogen replacement therapy seems <strong>to</strong> reduce the risk ofage-related cognitive impairments (213). Although cellproliferation in the GCL (<strong>and</strong> not the hilus) is higher infemale than in male rats, the newly born cells do notsurvive, which explains the lack of sex differences in thenumber of BrdU-IR cells 2 wk after labeling (536). Sexdependentproliferative activity involves the stimula<strong>to</strong>ryinfluence of estrogens, since the number of BrdU-labeledcells is highest during the proestrus phase of the estruscycle, when circulating levels of estrogens are highest(536), <strong>and</strong> acute administration of 17-estradiol reversesthe ovariec<strong>to</strong>my-induced decrease in cell birth (29, 536).In contrast, using TOAD-64 <strong>and</strong> calbindin as early <strong>and</strong> lateneuronal markers, it has been shown that estradiol doesnot alter neuronal differentiation (536).However, discrepant results have been reported followingchronic administration of estradiol <strong>to</strong> ovariec<strong>to</strong>mizedadult female rats (435), spontaneous hypertensiverats (436), or adult wild meadow voles (163). Cell proliferationin the GCL (measured 24 h after a single intraperi<strong>to</strong>nealinjection of [ 3 H]dT) is lower in female meadowvoles captured during the breeding season, when estradiollevels are high, compared with reproductively inactivefemales. A similar relationship has been confirmed inlabora<strong>to</strong>ry-reared female meadow voles (421).These apparently contradic<strong>to</strong>ry results might be explainedby a complex regula<strong>to</strong>ry mechanism. In fact, asingle administration of estradiol initially enhances(within 4 h) <strong>and</strong> subsequently suppresses (within 48 h)cell proliferation in the DG of ovariec<strong>to</strong>mized female ratsor meadow voles (420, 421). The increase in cell proliferationis mediated by sero<strong>to</strong>nin (29), whereas the decreaseis prevented by adx (422), suggesting an involvement ofcorticosterone. However, although corticosterone-inducedregulation of cell birth certainly involves NMDArecep<strong>to</strong>rs, estradiol influences on cell proliferation arenot mediated by these recep<strong>to</strong>rs (420). Finally, estrogenmay act directly on estrogen recep<strong>to</strong>rs subtype (ER )Downloaded from physrev.physiology.org on November 25, 2005Physiol Rev • VOL 85 • APRIL 2005 • www.prv.org


536 ABROUS, KOEHL, AND LE MOALpresent on hippocampal precursors (435) or throughIGF-I recep<strong>to</strong>rs (see sect. IID2C).III) Neurosteroids. Neurosteroids represent a subclassof steroids synthesized de novo in many regions ofthe brain, independently of peripheral sources (36). Theyare synthesized in the HF, by glial cells mainly, <strong>and</strong> influencehippocampal-mediated functions (349). Two principalfamilies can be distinguished according <strong>to</strong> their pharmacologicalproperties (333). In particular, dehydroxyepi<strong>and</strong>rosterone(DHEA) <strong>and</strong> pregnenolone sulfate(Preg-S) act as allosteric antagonists of the GABA A recep<strong>to</strong>rswhile allopregnanolone (AlloP) is a positive modula<strong>to</strong>rof these recep<strong>to</strong>rs.Chronic treatment of young adult male rats withsubcutaneous pellets of DHEA (200 mg/pellet for 12 days)has been shown <strong>to</strong> stimulate hippocampal cell proliferationmeasured 24 h after BrdU injections (50 mg · kg 1 ·day 1 ) given on the last 4 days of the steroid treatment(254). After an additional 16 days of treatment, the newlyborn cells survive <strong>and</strong> express the neuronal marker NeuN.Interestingly, DHEA treatment is also able <strong>to</strong> reverse thesuppressive effect of corticosterone on neurogenesis, suggestingthat it may act as an antistress neurohormone.Recently, we have shown that icv infusion of Preg-S(2 12 nmol/7 l) increases cell proliferation in the DGof young adult rats within 24 h, whereas treatment withAlloP (2 6.3 nmol/7 l) decreases it (348). The newlygenerated cells survive for at least 1 mo <strong>and</strong> differentiatemainly in<strong>to</strong> neurons (expressing NeuN). These actions ofPreg-S are mediated by GABA A recep<strong>to</strong>rs present on hippocampalprecursors, since icv administration of muscimol,a GABA A agonist, blocks Preg-S-induced cell proliferation.Furthermore, a direct action of Preg-S was suggestedas this compound stimulates in vitro theproliferation of spheres derived from the adult SVZ (348).Al<strong>to</strong>gether, the reported effects of DHEA <strong>and</strong> Preg-Sare congruent with the observations that in vitro activationof GABA A recep<strong>to</strong>rs inhibits cortical cell proliferation,whereas GABA A recep<strong>to</strong>rs antagonists stimulate it(21, 318, 327).B) NEUROTRANSMITTERS AND NEUROREGULATORS. I) Glutamate.Destruction of the perforant pathway, the main glutamatergicafference <strong>to</strong> the DG arising from the en<strong>to</strong>rhinalcortex, increases cell proliferation, thus indicating thatunder these experimental conditions glutamate exerts aninhibi<strong>to</strong>ry influence (73). As expected, blockade of NMDAsubtypes of glutamate recep<strong>to</strong>rs by injection of a noncompetitiveantagonist (MK801) increases cell genesis withina few hours in rats (1 mg/kg; Refs. 73, 76), tree shrews (1mg/kg; Ref. 177), gerbils (3 3 mg/kg; Ref. 46), <strong>and</strong>ovariec<strong>to</strong>mized adult female meadow voles (30 mg/kg;Ref. 420), <strong>and</strong> treatment with the competitive NMDA recep<strong>to</strong>rantagonist CGP 37849 (5 mg/kg) increases bothcell proliferation <strong>and</strong> granule neurons density (73). Conversely,administration of NMDA (30 mg/kg) decreasescell proliferation in several species within hours (73, 76,420), which is congruent with the antiproliferative propertiesof glutamate on in vitro cortical cell proliferation(318). However, it should be mentioned that an inhibi<strong>to</strong>ryinfluence of glutamate recep<strong>to</strong>r blockade on neurogenesishas been reported in stroke-damaged brains (see sect.IIID1B). The newly born cells induced by the inactivationof NMDA recep<strong>to</strong>rs differentiate in<strong>to</strong> neurons, expressingDCX, TOAD-64, NSE, or NeuN markers (73, 177, 390, 417).Furthermore, the MK-801-induced neurons are functionalas they respond <strong>to</strong> NMDA stimulation, measured by thephosphorylation of extracellular signal-regulated kinase(ERK), 29 days after their birth date (417). This indicatesthat newly born neurons acquire components for the intracellularsignal transduction cascade linking NMDA recep<strong>to</strong>rs<strong>to</strong> phosphorylation of ERK (114).The mechanisms by which precursor proliferation isinhibited by glutamate in vivo through NMDA recep<strong>to</strong>rsremain unknown, but they most probably do not involvecollateral deleterious effects as treatment with CGP 43487upregulates cell birth without inducing cell death or astrogliosis(390).Because glutamate also acts on<strong>to</strong> AMPA recep<strong>to</strong>rs,detected in neural progeni<strong>to</strong>rs (165), the influence ofpotentia<strong>to</strong>rs of AMPA recep<strong>to</strong>rs such as LY451646 hasbeen evaluated on hippocampal mi<strong>to</strong>tic activity. Acuteadministration of LY451646 (0.025, 0.05, 0.125 mg/kg)does not influence the number of newly born cells observed24 h after BrdU pulse (4 75 mg/kg every 2 h).However, the median dose increases the number of cellsper cluster (64%) <strong>and</strong> the number of clusters (45%) (27).Furthermore, chronic treatment (21 days) with LY451646(0.05, 0.125, 0.500 mg/kg) enhances the number of proliferatingcells, of cells per cluster, <strong>and</strong> of clusters in adose-dependent manner (27).Al<strong>to</strong>gether, these results suggest that glutamate exertsa complex influence on hippocampal cell proliferation,increasing it through activation of AMPA recep<strong>to</strong>rs,<strong>and</strong> inhibiting it through activation of NMDA recep<strong>to</strong>rs.Finally, the recent discovery that GABA <strong>and</strong> glutamate arecotransmitted at the mossy fibers synapses adds furthercomplexity <strong>to</strong> the respective role of each neurotransmitterin neurogenesis regulation (196).II) Sero<strong>to</strong>nin. In the 1970s, it was proposed that earlyforming sero<strong>to</strong>nin (5-HT) neurons act as humoral signalsgoverning neuronal development <strong>and</strong> neurogenesis in particular(26, 298). Since then, several approaches haveshown that sero<strong>to</strong>nin upregulates cell proliferation in theadult DG <strong>and</strong> SVZ (see also sect. IIID2A). Inhibition of 5-HTsynthesis (by subchronic injections of PCPA for 6 days)<strong>and</strong> selective lesions of 5-HT neurons of the raphe decreasethe number of BrdU- <strong>and</strong> PSA-NCAM-IR cells in theDG <strong>and</strong> the SVZ (BrdU 50 mg/kg ip injected on the last 3days of PCPA treatment at 1–2h intervals <strong>and</strong> animalskilled 6 h after the last injection; Ref. 61). This upregula-Downloaded from physrev.physiology.org on November 25, 2005Physiol Rev • VOL 85 • APRIL 2005 • www.prv.org


NEUROGENESIS IN THE ADULT BRAIN 537tion of hippocampal cell proliferation depends on sero<strong>to</strong>ninas it is reversed by intrahippocampal grafts of embryonic5-HT neurons (63). Partial lesions of 5-HT rapheneurons lead, after an initial deprivation of hippocampal5-HT fibers, <strong>to</strong> a spontaneous reinnervation of this structureby 5-HT fibers. At this time, the fall of mi<strong>to</strong>tic activity(<strong>and</strong> PSA-NCAM expression) is reversed (62). These actionsof sero<strong>to</strong>nin are mediated by 5-HT 1A recep<strong>to</strong>rs inboth the DG <strong>and</strong> the SVZ (30, 451) while 5-HT 2A <strong>and</strong>5-HT2Crecep<strong>to</strong>rs are selectively involved in the regulationof cell proliferation in the DG <strong>and</strong> SVZ, respectively (30).Finally, 4 wk after chronic treatment with 5-HT 1A or5-HT2Aagonists (daily injection of 8-OH-DPAT or RO600175 for 15 days, 75 mg/kg of BrdU injected twice onthe last 8 days of the treatment), the newly born cells in theDG <strong>and</strong>/or the OB express NeuN (30).III) Nitric oxide. Nitric oxide (NO), a free radicalmolecule synthesized from L-arginine by NO synthase,serves as a neurotransmitter in the brain (8). In the SVZ ofadult mice, neuronal precursors have been found <strong>to</strong> bewithin the sphere of influence of a NO source <strong>and</strong> <strong>to</strong>express NO synthase at the sites of terminal differentiation(377). Administration of DETA/NONOate, a NO donor,<strong>to</strong> young adult rats significantly increases both cellproliferation <strong>and</strong> migration in the SVZ <strong>and</strong> the DG (587).C) TROPHIC FACTORS. Many trophic fac<strong>to</strong>rs have beenshown <strong>to</strong> have mi<strong>to</strong>genic actions in the adult brain neurogenicregions. Thus the proliferative effects of basicfibroblast growth fac<strong>to</strong>r (bFGF or FGF-2) have beenclearly demonstrated for the SVZ. Chronic administrationof bFGF intracerebroventrically (360 ng/day for 14 days,50mg·kg 1 · day 1 of BrdU being injected during the last12 days of the treatment; Ref. 291) or intranasally (5 0.2g/day at 5-min intervals for 1 wk concurrent with 2 50mg · kg 1 · day 1 of BrdU at 8-h intervals; Ref. 240)increases the number of mi<strong>to</strong>tic nuclei in the SVZ within24 h. This leads <strong>to</strong> an increased number of newly bornneurons (expressing DCX in the SVZ; Ref. 240) that reachthe OB where they express NeuN (4 wk after their birth;Ref. 291). In contrast, in the DG, a lack of effect (291) ora small but nonsignificant increase in cell birth (25%,following 3 days icv infusion of 10 g/ml) was reported(238). Consistently, in mice lacking bFGF, basal hippocampalneurogenesis is “normal,” suggesting that otherfac<strong>to</strong>rs may maintain low levels of precursor proliferationunder resting conditions (582). However, overexpressionof bFGF by gene transfer in wild-type <strong>and</strong> bFGF-deficientmice upregulates DG cell proliferation, which indicatesthat a robust <strong>and</strong> constant bFGF expression is a necessarycondition for increasing cell birth in this structure(582).Epidermal growth fac<strong>to</strong>r (EGF; 350 ng/day for 14days, 50 mg · kg 1 · day 1 of BrdU being injected duringthe last 12 days of the treatment; Ref. 291) <strong>and</strong> heparinbindingEGF (HB-EGF; 3 days icv infusion of 10 g/mlconcurrent with 2 50 mg · kg 1 · day 1 of BrdU at 8-hintervals; Refs. 236, 238) stimulate cell birth in the SVZ (1<strong>and</strong> 7 days after growth fac<strong>to</strong>r infusion, respectively)certainly via EGF recep<strong>to</strong>rs (381, 416, 502). However,these two fac<strong>to</strong>rs probably have distinct actions, sinceHB-EGF increases the number of BrdU-IR cells expressingDCX or NeuroD in the SVZ as well as the number ofnewly generated cells reaching the OB (236, 240), whereasEGF reduces the number of newly born neurons reachingthe OB (291). In the DG, HB-EGF (236, 238) but not EGF(291) increases cell proliferation by interacting mostprobably with EGF recep<strong>to</strong>rs expressed by the dividingcells (416). The adult-born cells express the neuronalmarker NeuroD (236).Transforming growth fac<strong>to</strong>r (TGF)- is required forSVZ precursor proliferation. Indeed, icv administration ofTGF- (400 ng/day for 6 days) induces a dramatic increasein precursor proliferation (98), whereas TGF- null miceshow a decrease in proliferating cells in the SVZ, <strong>and</strong> inthe <strong>to</strong>tal number of newly born cells within their target,the OB (543). No data on the influence of TGF- onhippocampal precursors are yet available.IGF-I is a growth-promoting peptide hormone that isproduced in the CNS by neurons <strong>and</strong> glial cells (20) <strong>and</strong>exhibits neurotrophic properties in adulthood (405). Itsinfluence on neurogenesis has been examined in hypophysiec<strong>to</strong>mizedrats presenting low levels of circulatingIGF-I (1). Thus peripheral administration of IGF-I(1.25 mg · kg 1 · day 1 ) induces an increase of cell proliferationin the dentate GCL <strong>and</strong> the hilus after 6 days.Long-term treatment (0.39 mg · kg 1 · day 1 for 20 days)increases both BrdU-labeled cell number <strong>and</strong> their differentiationin<strong>to</strong> neurons as measured by the percentage ofBrdU-IR cells expressing calbindin. In contrast, an IGF-Iinducedincrease in cell proliferation in nonhypophysiec<strong>to</strong>mizedrats is not associated with an enhancement oftheir differentiation potential <strong>to</strong>ward a neuronal fate(542). In vitro experiments (2) strongly suggest that IGF-Iregulates cell proliferation in vivo by acting directly onIGF-I recep<strong>to</strong>rs expressed by newly born cells (542), buta recent study has also shown that estrogen recep<strong>to</strong>rs arenecessary for the induction of in vivo hippocampal cellproliferation by IGF-I (435).Brain-derived neurotrophic fac<strong>to</strong>r (BDNF) is a memberof a family of related neurotrophic proteins, the functionof which is <strong>to</strong> prevent neurons from dying duringdevelopment. Chronic icv infusion of BDNF (0.012 l/dayfor 12 days) increased BrdU-labeled cells in the SVZ <strong>and</strong>in the GCL of the OB (593). The newly born cells expressedTuJ1 or MAP2. Survival (169, 276) <strong>and</strong>/or differentiation(6, 22) of the neuronal precursors <strong>and</strong> theirprogeny rather than proliferation seem <strong>to</strong> be influencedby BDNF. Concerning the DG, it has been shown thatheterozygous BDNF knockout mice exhibit reduced proliferation<strong>and</strong> survival of BrdU-labeled cells (301), sug-Downloaded from physrev.physiology.org on November 25, 2005Physiol Rev • VOL 85 • APRIL 2005 • www.prv.org


538 ABROUS, KOEHL, AND LE MOALgesting a role for BDNF as a positive regula<strong>to</strong>r of bothproliferation <strong>and</strong> survival. However, repeated, but notsingle, administration of a compound that stimulates endogenousBDNF, riluzole, increases cell birth (3-fold) butnot cell survival. Most of the newly generated cells (90%)differentiate in<strong>to</strong> granule neurons (expressing NeuN)(257). This effect is blocked by icv administration ofBDNF-specific antibodies, suggesting that an increase inBDNF is necessary for the promoting effect of riluzole onprecursor proliferation. Al<strong>to</strong>gether these data show thatBDNF plays an important role in the maintenance of basalneurogenesis, a conclusion similar <strong>to</strong> that obtained followinga developmental loss of function of this gene(312).Vascular endothelial growth fac<strong>to</strong>r (VEGF) is ahypoxia-induced angiogenic protein that exhibits neurotrophic<strong>and</strong> neuroprotective properties (359). Given thatneurogenesis occurs in close proximity <strong>to</strong> blood vessels<strong>and</strong> that clusters of dividing cells contain endothelialprecursors (see sect. IIC2B), VEGF may constitute the linkbetween neurogenesis <strong>and</strong> angiogenesis. This is supportedby the observation that icv administration of VEGF(0.24 g/day for 3 days concurrent with 2 50 mg · kg 1· day 1 BrdU) stimulates cell proliferation in the rodentSVZ <strong>and</strong> SGZ by 7 days after treatment onset (241). At thattime, the newly born cells express DCX <strong>and</strong> NeuN markers.The neuroproliferative effect of VEGF is associatedwith an upregulation of cyclin D1, Cyclin E <strong>and</strong> cdc25, <strong>and</strong>an increase in the nuclear expression of E2F1, E2F2, <strong>and</strong>E2F3 (591), which is consistent with a regulation of theG 1 /S phase transition of the cell cycle.D) MORPHOGENIC FACTORS. Sonic hedgehog (Shh), an importantmorphogen in development, is a signaling glycoproteinthat acts through Patched 1-Smoothened (Ptc1-Smo) recep<strong>to</strong>r complex <strong>to</strong> trigger various events duringCNS development, including determination of ventralneural phenotypes, induction of oligodendrocyte precursors,proliferation of specific neuron progeni<strong>to</strong>r populations,<strong>and</strong> modulation of growth cone movements (for areview, see Ref. 346). In a comprehensive study, Lai <strong>and</strong>co-workers (295) investigated the role of Shh in the adultbrain <strong>and</strong> showed that it increased proliferation of culturedadult rat hippocampal progeni<strong>to</strong>rs in a dose- <strong>and</strong>time-dependent fashion. In vivo, the exogenous administrationof Shh <strong>and</strong> the inhibition of its signaling increased<strong>and</strong> reduced cell proliferation, respectively. Finally, thepresence of the Shh recep<strong>to</strong>r Patched in the DG <strong>and</strong>Ammon’s horn in the HF, <strong>and</strong> the high levels of expressionof Shh in structures that project <strong>to</strong>wards the DGsupport the assumption that Shh could be an endogenouspositive regula<strong>to</strong>r of cell proliferation in the adult DG(295).Another group of early neural morphogens, the bonemorphogenic proteins (BMPs), belongs <strong>to</strong> the TGF- superfamily,which includes TGF-, activins, <strong>and</strong> the relativesnamed growth/differentiating fac<strong>to</strong>rs (GDF). Theseglycoproteins play a crucial role in bone remodeling <strong>and</strong>in the regulation of dorsoventral patterning of the neuraltube <strong>and</strong> cell fate during embryonic development. Furthermore,several BMPs, including BMP4, are involved inrepression of the oligodendroglial lineage <strong>and</strong> generationof the astroglial lineage during brain maturation (190).Similarly, in the adult SVZ, BMPs (BMP2, BMP4, BMP7)inhibit neurogenesis <strong>and</strong> direct astroglial differentiation,whereas their antagonist Noggin promotes neurogenesis(311). It has been proposed that Noggin produced by thetype E cells antagonizes the type B cells expressing BMP(311). Furthermore, BMP2, BMP4, <strong>and</strong> BMP7 have beenfound <strong>to</strong> activate a promoter of the gene for the HLHfac<strong>to</strong>r Id1, which is known <strong>to</strong> inhibit the function ofneurogenic transcription fac<strong>to</strong>rs such as Mash1 <strong>and</strong> neurogenin.Thus the switch from neuronal <strong>to</strong> astrocyte faterealized by BMPs certainly involves HLH proteins (578).These findings were extended <strong>to</strong> the HF. Indeed, a novelsecre<strong>to</strong>ry fac<strong>to</strong>r, neurogenesin-1 (Ng1), released by hippocampalastrocytes <strong>and</strong> dentate granule cells adjacent <strong>to</strong>progeni<strong>to</strong>r cells, was found <strong>to</strong> promote neuronal fate inthe adult HF through antagonism of BMPs, which alter thefate of neural stemlike cells from neurogenesis <strong>to</strong> astrogliogenesisby upregulating the expression of the negativeHLH fac<strong>to</strong>rs Id1, Id3, <strong>and</strong> Hes5 (548).E) REGULATION BY GLIAL CELLS. Regulation of adult neurogenesisby glial cells has been emphasized (500). Astrocytes,which play an important role as sensors of changesin their extracellular microenvironment, could regulateneurogenesis by secreting local signals (514). These signals,unknown so far, may be ionic fluxes, neurosteroids,cy<strong>to</strong>kines, growth fac<strong>to</strong>rs, <strong>and</strong> glutamate metabolites (48,243, 245, 296, 486, 516, 579, 595). The observation thatsome proliferating cells in the DG express a recep<strong>to</strong>r forS100, a small acidic calcium binding neurotrophic proteinreleased by astrocytes, reinforces the putative contributionof glial cells in the regulation of adult neurogenesis(339).F) REGULATION BY CELL DEATH. An equilibrium between neurogenesis<strong>and</strong> cell death probably ensures a homeostaticbalance in the adult brain. This hypothesis, based on theobservation that the neurogenic structures do not grow insize, implies that cell death provides a stimulus for increasedneurogenesis, a hypothesis reinforced by severallines of argument: 1) during development, there is a balancebetween the birth <strong>and</strong> death of granule cells (183); 2)mechanical lesions, aspiration, <strong>and</strong> transection stimulateneuronal precursor proliferation in the GCL <strong>and</strong>/or theSVZ (180, 242, 531, 546, 568); 3) proliferation in the SVZ isupregulated under inflamma<strong>to</strong>ry conditions (70); 4) adrenalec<strong>to</strong>my,limbic seizures, <strong>and</strong> ischemia enhance bothcell death <strong>and</strong> cell birth; <strong>and</strong> 5) the apop<strong>to</strong>tic degenerationof corticothalamic neurons (by means of targetedpho<strong>to</strong>lysis) induces endogenous neural precursors <strong>to</strong> dif-Downloaded from physrev.physiology.org on November 25, 2005Physiol Rev • VOL 85 • APRIL 2005 • www.prv.org


NEUROGENESIS IN THE ADULT BRAIN 539ferentiate in<strong>to</strong> mature neurons in regions of the cortexundergoing targeted neuronal death (332). The inductionof neurogenesis in these regions of the adult neocortexthat do not normally undergo any neurogenesis (332) mayresult from the removal of a normal inhibi<strong>to</strong>ry influenceor the loss of a secreted stimula<strong>to</strong>ry fac<strong>to</strong>r produced bythe dying cells.G) SUMMARY. <strong>Adult</strong> neurogenesis within each neurogenicsite is regulated differently by a growing list of“epigenetic fac<strong>to</strong>rs” [<strong>to</strong> which should be added prolactin(506), norepinephrine (35, 292), the pituitary adenylatecyclase-activating polypeptide (PACAP) (362), ciliary neurotrophicfac<strong>to</strong>r (141), retinoic acid (99), <strong>and</strong> vitamin E(83, 86, 87); see Table 1 for a complete summary]. Althoughmost of these fac<strong>to</strong>rs modulate cell proliferationthrough unknown mechanisms, a regulation of Cyclin D1<strong>and</strong> p27 Kip1 expressions may constitute a common pathway(438).The specificities of each neurogenic site may be related<strong>to</strong> differences in the intrinsic properties of the dividingcells (for example, their intrinsic ability <strong>to</strong> senseneural activity; Ref. 114) <strong>and</strong>/or in their microenvironment,which corresponds <strong>to</strong> the summation of local neurogenicsignals expressed or synthesized “locally” byhealthy neighboring cells or by dying cells. These signalsmay also be synthesized “peripherally” <strong>and</strong> released inneurogenic sites by neuronal afferences or by blood vessels.In this context, the role of the cerebral vasculaturehas gained importance as adult neurogenesis occurswithin an “angiogenic niche,” <strong>and</strong> an alteration in thevascular microenvironment, or its ability <strong>to</strong> respond <strong>to</strong>changes in metabolic dem<strong>and</strong>s, may be responsible for adisruption in neurogenesis (371, 372, 460).III. THE ANATOMO-FUNCTIONAL APPROACHComplementary <strong>to</strong> the neuroana<strong>to</strong>mic studies thatrevealed some key biological properties of adult neuralstemlike cells, the integration of the adult newborn neuronsin<strong>to</strong> preexisting networks has prompted the fundamentalquestion of their functional relevance. So far, moststudies have focused on the role of adult neurogenesis inhippocampal functioning. Parsimoniously, this region, includedin the limbic-cognitive system, is part of an integratednetwork involved in learning <strong>and</strong> memory, attentionalprocesses, motivational states, <strong>and</strong> emotion (68,135, 136, 227, 352, 484). As pointed out by Hamp<strong>to</strong>n et al.(201), current theories of hippocampal function wrestlewith two major conceptual issues: whether the structureencodes spatial, nonspatial, or both types of information(139) <strong>and</strong> whether the information encoded is episodic orsemantic. Recent evidence infers that hippocampal neuronalnetworks could support the cognitive mechanismsof declarative memory by encoding features of experiences.Indeed, declarative memory involves a record ofeveryday experiences, which includes information aboutthe content of that experience <strong>and</strong> the spatial <strong>and</strong> temporalcontext in which it occurred (episodic memory), woven<strong>to</strong>gether in<strong>to</strong> the framework of the individual’s knowledge(semantic memory) (135). According <strong>to</strong> the memoryspace model of declarative memory, the HF plays a criticalrole in 1) representing experiences as a series ofevents <strong>and</strong> places <strong>and</strong> 2) linking memories <strong>to</strong>gether byidentifying common features in<strong>to</strong> a network that supportsinferential expression. This said, the role of the HF inlong-term memory is in dispute as the “st<strong>and</strong>ard theory”assumes that the HF becomes dispensable after the conversionof short-term memory in<strong>to</strong> long-lasting memory,whereas the “multiple trace theory” makes the assertionthat the HF is involved in the s<strong>to</strong>rage <strong>and</strong> retrieval ofepisodic memory regardless of the age of memory (394).Although current advances do not yet allow the attributionof a specific role for newborn neurons in thisconceptual framework, in the following sections we willreview the progress achieved in implicating neurogenesisin both hippocampal functioning under physiological conditions<strong>and</strong> the apparition of hippocampal-related pathologies,which are mainly studied in preclinical animal models.Operationally, two approaches have been considered:the first consists of relating changes in hippocampal neurogenesis<strong>to</strong> changes in the ability <strong>to</strong> perform hippocampal-relatedtasks, <strong>and</strong> the second consists of studying theinfluence of hippocampal-related tasks on neurogenesis.A. Environmental <strong>and</strong> Physiological Influenceson <strong>Neurogenesis</strong>1. Experience in enriched environments,neurogenesis, <strong>and</strong> learningA) OLD PROBLEM, NEW DATA. Rosenzweig <strong>and</strong> co-workers(470–472) were the first <strong>to</strong> introduce the concept of anenriched situation, i.e., a complex environment enrichedin sensorial stimulations, social experiences, <strong>and</strong> physical<strong>and</strong> cognitive exercises (555). This concept has given rise<strong>to</strong> a wealth of studies showing that exposure <strong>to</strong> suchenvironments, which are considered as enriched for st<strong>and</strong>ardlabora<strong>to</strong>ry conditions, modifies brain functioning,learning, problem-solving ability, brain chemistry, <strong>and</strong>several structural aspects of brain organization. The consequenceof environment enrichment on hippocampalneurogenesis was originally studied in “juvenile” femaleC57Bl/6J mice. Exposure <strong>to</strong> an enriched environment between3 <strong>and</strong> 13 wk of age was found <strong>to</strong> increase survivalof newly born cells (measured 4 wk after daily injectionsof 50 mg BrdU for 12 consecutive days from days 28 <strong>to</strong> 40of enrichment), whereas cell proliferation (measured 1day after the BrdU pulses) remained unchanged in theGCL <strong>and</strong> in the hilus (267). Similar results have beenDownloaded from physrev.physiology.org on November 25, 2005Physiol Rev • VOL 85 • APRIL 2005 • www.prv.org


540 ABROUS, KOEHL, AND LE MOALobtained with adult female rats (after injecting BrdU 50mg·kg 1 · day 1 on days 26–30 of enrichment) (407).These first reports led Kempermann <strong>and</strong> Gage (262) <strong>to</strong>further investigate the impact of environmental enrichmen<strong>to</strong>n mice, <strong>and</strong> it was later shown that cell proliferationin the GCL was increased only when mice were killed3 mo after their removal from the enriched environment(262). In addition, a comparison of different strains ofmice showed that enriched experience affects hippocampalneurogenesis differently according <strong>to</strong> genetic background:as reported above, it increases cell survival butnot cell proliferation in C57BL/6 mice (267) which have ahigh baseline level of neurogenesis (268), whereas it increasescell proliferation in 129/SvJ mice (261) whichhave low levels of neurogenesis (268). This interestingfinding indicates that environment has a differential impactdepending on inheritable traits, an effect still unexplained.Finally, in most experiments, enrichment led <strong>to</strong>an improvement in spatial memory in the water maze, alearning task that requires the subjects <strong>to</strong> learn multipleextra-maze visual cues, allowing them <strong>to</strong> build a dynamicspatial representation of their surroundings <strong>to</strong> navigate <strong>to</strong>a platform hidden underneath the water surface (378).However, whether these genetic variations of neurogenesishave a functional significance in spatial memory remainsunknown (266).B) MECHANISMS INVOLVED IN ENRICHMENT-INDUCED NEUROGENESIS.Exposure <strong>to</strong> an enriched environment is known <strong>to</strong> influencebrain chemistry, in particular neurotransmitters <strong>and</strong>trophic fac<strong>to</strong>rs, which may underlie its action on neurogenesis(151, 370, 442, 471, 555, 584). In addition <strong>to</strong> thesefac<strong>to</strong>rs, several components of this environment may beinvolved in the regulation of neurogenesis.Thus the effects of an enriched environment on neurogenesis<strong>and</strong> spatial memory may be related <strong>to</strong> an enhancemen<strong>to</strong>f mo<strong>to</strong>r exercise rather than learning. Toexamine this hypothesis, female mice were housed with arunning wheel, injected with BrdU (50 mg · kg 1 · day 1for the first 12 days) <strong>and</strong> killed 1 or 30 days after the lastBrdU injection. In these conditions, cell proliferation <strong>and</strong>cell survival in the GCL, <strong>and</strong> performances in spatiallearning tasks were enhanced, suggesting that “physicalactivity” is an important media<strong>to</strong>r of enrichment-inducedneurogenesis (553, 554). Further studies proposed thatthe stimula<strong>to</strong>ry effects of running are mediated by VEGF(148), blood-borne IGF-I (542), NMDA recep<strong>to</strong>r 1-subunit,<strong>and</strong> BDNF production (278).In their early studies, Rosenzweig <strong>and</strong> Bennett (471)suggested that environmental changes or novelty were amajor fac<strong>to</strong>r in the effects of enrichment on brain <strong>and</strong>behavior (471), which suggests that it could be a relevantfac<strong>to</strong>r involved in enrichment-induced neurogenesis aswell. The observation that exposure <strong>to</strong> enriched conditionsfor an extended period of 6 mo does not add furtherbenefit certainly lends weight <strong>to</strong> this hypothesis (262).The social environment may also contribute <strong>to</strong> enrichment-inducedneurogenesis. This hypothesis has beentested in rats reared for 4 wk in isolation <strong>and</strong> injected withBrdU (2 50 mg · kg 1 · day 1 ) on the last 3 days of therearing treatment. Compared with grouped-reared rats,isolation decreases hippocampal cell proliferation within24 h, the number of newly born cells surviving over anadditional period of 4 wk of isolation, <strong>and</strong> the percentageof newly born cells expressing TOAD-64. Interestingly,the isolation-induced decrease in cell proliferation is reversedby a subsequent 4 wk of group housing (BrdUbeing injected on the last 3 days of the rearing period; Ref.320). Together, these results indicate that social environmentis a highly relevant fac<strong>to</strong>r underlying the ana<strong>to</strong>micaleffects of an enriched environment.2. Reciprocal relation between learning<strong>and</strong> neurogenesisA) LEARNING INFLUENCES SEVERAL ASPECTS OF NEUROGENESIS. Theeffect of learning on survival of newly born cells has beenexamined in the water maze <strong>and</strong> in eyeblink conditioningusing a trace pro<strong>to</strong>col (175). In this latter test, whichrequires an intact HF (385, 512), animals have <strong>to</strong> associatean audi<strong>to</strong>ry <strong>to</strong>ne (conditioned stimulus) with a cornealairpuff or electrical stimulation (unconditioned stimulus)that is temporally separated by a trace interval (350). Inboth learning tasks, rats are tested 1 wk after a singleBrdU injection (200 mg/kg). The number of BrdU-labeledcells is enhanced by learning immediately or 1 wk afterthe end of training, an effect accompanied by a decreasein cell death. In associative learning, the performance ofindividual rats positively predicts the life span of 1-wk-oldBrdU-labeled cells, i.e., rats exhibiting the best behavioralscores possessed more newborn cells; this also suggeststhat learning (<strong>and</strong> not training) is a sufficient condition <strong>to</strong>increase the survival of adult-born cells (309). This learning-inducedincrease in cell survival was maintained 2 moafter acquisition of associative learning (309). The observedenhancement of cell survival is specific <strong>to</strong> learningbecause neurogenesis remains unchanged in rats exposed<strong>to</strong> unpaired conditions in eyeblink conditioning or in ratsproducing the same amount of mo<strong>to</strong>r responses in thewater maze. Furthermore, learning-induced upregulationof neurogenesis may be specifically attributed <strong>to</strong> hippocampalfunctioning as delay-eyeblink conditioning,classically attributed <strong>to</strong> the cerebellum, <strong>and</strong> training on acued test in the water maze, a hippocampal-independenttype of learning, do not modify neurogenesis (508).Although these experiments indicate an enhancingeffect of learning in the water maze on cell survival,contradictions exist concerning cell proliferation. Whenanimals are injected daily for the entire testing period (50mg · kg 1 · day 1 for 12 days; Ref. 554) or the very last dayof testing (200 mg/kg; Ref. 175), the number of BrdU-Downloaded from physrev.physiology.org on November 25, 2005Physiol Rev • VOL 85 • APRIL 2005 • www.prv.org


NEUROGENESIS IN THE ADULT BRAIN 541labeled cells remains unchanged. However, when animalsare injected at the end of the learning phase (50 mg ·kg 1 · day 1 for 3 days), when an asymp<strong>to</strong>tic level ofperformance is reached, then the number of newly bornneurons in the GCL of the DG increases (306). To reconcilethese data, we hypothesized that different phases ofthe learning process have distinct actions on cell proliferation.To test this hypothesis rats were injected withBrdU (50 mg · kg 1 · day 1 ) either during the initial phase(4 days, Fig. 4, A <strong>and</strong> E), characterized by a fast <strong>and</strong> largeimprovement in performance, or during the late phase (4days, Fig. 4C), when an asymp<strong>to</strong>tic level of performanceFIG. 4. Learning can increase or decrease the production of newborncells. The aim of these experiments was <strong>to</strong> distinguish the effectsof the early phase () of learning (characterized by a rapid <strong>and</strong> largeimprovement in performance) <strong>and</strong> of the late phase of learning (whenasymp<strong>to</strong>tic levels of performance are reached) on neurogenesis. To thisend, bromodeoxyuridine (BrdU; solid bars) was injected during the early (A, E) or the late (C) of learning <strong>and</strong> animals were killed (arrows)after partial learning (A, at the end of the early ) or when the task hadbeen mastered (C <strong>and</strong> E, at the end of the late ). The early of learningdid not modify the number of cells generated during this period (B). Thelate of learning increased the number of newly born cells generatedduring this period (D), whereas it decreased the number of newly borncells produced during the early (F). C, control group; L, learning group.**P 0.001 compared with control group. [Modified from Döbrössy etal. (121).]was reached. We found that the early phase of learningdoes not modify proliferation (Fig. 4A), whereas the lateone does (121). Indeed, the number of newly born cellsincreased contingently with the late phase of learning(Fig. 4B), <strong>and</strong> these new cells differentiated in<strong>to</strong> neuronsthat persisted in the DG for at least 5 wk after the animalshad acquired the task. In contrast, the late phase of learninginduced a decrease in the number of newly born cellsproduced during the early phase (Fig. 4C).Most importantly, the extent of BrdU cell loss wasinversely related <strong>to</strong> learning performances: rats with thehighest <strong>and</strong> lowest cell loss have the best <strong>and</strong> worstperformances, respectively. This decline in adult-borncells most probably mirrors cell death, since learninginduces an increase in numbers of pyknotic cells (129)<strong>and</strong> tunnel-positive cells (18). The observations that learningin the water maze increases the expression of proteinsinvolved in apop<strong>to</strong>sis (82) <strong>and</strong> that intrahippocampal administrationof anticaspases, which blocks cell death,impairs long-term but not short-term spatial memory(106), strengthen the hypothesis that cell death is animportant component of hippocampal learning.The mechanisms by which cell survival <strong>and</strong> proliferationare regulated during learning are so far unknown. Anonspecific effect of training or exercise on blood flow <strong>to</strong>the brain that may increase BrdU availability is unlikely,since neurogenesis is unchanged in control “swimmers”(121, 175). Trophic fac<strong>to</strong>rs known <strong>to</strong> support the viability<strong>and</strong> function of many types of neurons are likely media<strong>to</strong>rsof learning-dependent changes in the HF. Indeed,hippocampal bFGF <strong>and</strong> BDNF are increased during tasklearning <strong>and</strong> decreased once an asymp<strong>to</strong>tic performanceis reached (171, 271).B) A BLOCKADE OF NEUROGENESIS DISTURBS LEARNING. The participationof neurogenesis in memory formation has beenmore directly tested by using the DNA methylating agentmethylazoxymethanol (MAM; 5 mg/kg for 14 days concurrentwith 75 mg · kg 1 · day 1 of BrdU on days 10–12-14).This treatment decreases the number of adult-born cellsby 80% <strong>and</strong> alters trace conditioning, whereas delay conditioning,neuronal responsiveness (presynaptic neurotransmitterrelease <strong>and</strong> excitability), <strong>and</strong> synaptic plasticityare not disrupted (509). Three weeks after treatmentcessation, the replenishment of immature neurons is associatedwith a recovery <strong>to</strong> acquire the trace conditionedresponse. Al<strong>to</strong>gether, these results suggest that newlyborn cells are necessary for the formation of trace memories.Because MAM treatment for 6 days only is unable <strong>to</strong>affect trace conditioning, adult-born cells may not be“functional” until 1–2 wk after birth. Although it has beenshown that newborn cells extend their axons in the CA3subfield within 4–10 days after birth (204), a basic temporalstudy following their functional maturation (bymeans of electrophysiological properties or c-fos expression)is still lacking.Downloaded from physrev.physiology.org on November 25, 2005Physiol Rev • VOL 85 • APRIL 2005 • www.prv.org


542 ABROUS, KOEHL, AND LE MOALRecently, it has been reported that the formation ofspatial memories may not be associated with neurogenesis.Indeed, the blockade of cell proliferation by the same14-day MAM treatment as described above does not alterspatial learning in the water maze or contextual conditioningin fear conditioning (510). In our opinion, this lackof effect is due <strong>to</strong> the residual newly born cells thatcontinue <strong>to</strong> be generated under MAM (700/day for 3BrdU injections), indicating that learning can occur with avery small percentage of new neurons. This assumption isbased on the observation that some aged rats are able <strong>to</strong>acquire spatial navigation learning despite a very lownumber of newly born cells [200/day for 5 BrdU injections;see sect. IIIC2, Fig. 6 (130)].An emerging strategy <strong>to</strong> clarify the role of neurogenesisin learning consists of using ionizing-irradiationknown <strong>to</strong> induce cognitive impairments in animals (37, 66,119, 184) <strong>and</strong> in humans (372). X-irradiations (0–15 Gy)decrease the number of BrdU-labeled cells in a dosedependentmanner (following a 60 mg/kg injection ofBrdU 1 h before death) <strong>and</strong> the number of cells expressingp34 cdc2 or Ki-67 in the adult DG (369, 433, 532). Twomonths after irradiation, the production of adult-bornneurons (expressing NeuN) is significantly reduced certainlyas a consequence of an altered neurogenic microenvironment(369, 372). The reduction in hippocampalmi<strong>to</strong>tic activity is accompanied 1 wk postirradiation bybehavioral deficits in a hippocampal-dependent task ofspatial short-term memory (place-recognition). In contrast,performances in the water maze remain unaffected2 wk postirradiation, a time by which proliferative activityhas resumed, albeit at a low level (75 nascent cells dailyin the dorsal DG; Ref. 330).3. DiscussionData accumulated in the past decade suggest that thebirth of new cells is a necessary condition for the acquisitionof memory traces (see also data on prenatal stress<strong>and</strong> aging). However, a higher reduction in cell genesisseems <strong>to</strong> be required for the appearance of deficits inlearning spatial cues than in trace conditioning, a differencewhich may be related <strong>to</strong> task difficulty (508). Furthermore,hippocampal-dependent behaviors influencecell proliferation, cell survival, <strong>and</strong> cell death. The questionof how adult neurogenesis participates in memoryprocessing (i.e., encoding, consolidation, retrieval) is asFIG. 5. Influence of the different phases oflearning on neurogenesis. The early phase () oflearning increases the survival of cells that predateexposure <strong>to</strong> the task (175). These cells could beinvolved in the encoding of memory traces, allowingtask acquisition (see sect. IIIA3A). Since thelearning-induced increase in cell survival is longlasting(309), the surviving adult-born neuronscould participate in the long-term s<strong>to</strong>rage of information<strong>and</strong> <strong>to</strong> recall remote memories. Two distinctprocesses take place after completion of the latephase of learning (121; see Fig. 4 <strong>and</strong> sect. IIIA3, B<strong>and</strong> C): 1) death of the cells born during the earlyphase of learning, which may be involved in memorytrace consolidation. This cell death phenomenoncould constitute a trimming mechanism thatsuppresses old unnecessary neurons <strong>and</strong>/or the immatureneurons that are <strong>to</strong>o old or have not establishedlearning-related synaptic connections. 2)There is an increase in the number of cells generatedcontingently with the late phase training.Given that the learning-induced increase in cell proliferationis long-lasting, these newly born cellscould be involved in a resetting process renderingthe DG available for the acquisition of new memories(forgetting <strong>and</strong> flexibility).Downloaded from physrev.physiology.org on November 25, 2005Physiol Rev • VOL 85 • APRIL 2005 • www.prv.org


NEUROGENESIS IN THE ADULT BRAIN 543yet unresolved. Although speculative, the followingframework is proposed (Fig. 5).A) DO ADULT-BORN NEURONS PRODUCED BEFORE LEARNING HAVE AROLE IN MEMORY PROCESSING? Since survival of the newly borncells whose birth predates the beginning of learning isincreased during the learning task (175), the encoding ofmemory traces may be processed in these adult-bornneurons. According <strong>to</strong> the “use it or lose it” principle, thecells that survived <strong>and</strong> integrated in<strong>to</strong> the granule cells/mossy fiber system could function as postsynaptic detec<strong>to</strong>rsof correlated afferent activity <strong>and</strong> thereby update thebearing map, which is constructed in the DG from directionalcues (229). After processing, which relies on patternseparation properties (270), episodic memories ares<strong>to</strong>red temporally by virtue of the CA3 au<strong>to</strong>-associativesystem (468). The adult-born neurons could also be involvedin temporal pattern completion: if a degraded versionof a stimulus is presented after learning, the representedfeature can be recalled from this incomplete inputinformation.The role of adult-born neurons in long-term s<strong>to</strong>rage<strong>and</strong> retrieval of remote memories is still elusive as therole of the HF <strong>and</strong> the DG is still a matter of debate. Oneview stipulates that the HF has a limited s<strong>to</strong>rage capacityas 1-mo-old memories do not require the HF, memorytraces being transferred within cortical areas where theyare consolidated <strong>and</strong> permanently s<strong>to</strong>red (19, 56, 273, 274,342, 573). In relation <strong>to</strong> this apparent time-limited role ofthe HF, it was originally thought that after encoding <strong>and</strong>conversion of short-term memory in<strong>to</strong> long-lasting memory,the adult-born neurons were eliminated (189). However,adult-born neurons that have been recruited bylearning survive for several weeks (121, 309) <strong>and</strong> thusoutlive the time required for cortical transfer. This timewindowis, in our opinion, hardly reconcilable with the“consolidation transfer hypothesis.” Furthermore, if oneconsiders that adult-born neurons are integrated in<strong>to</strong> theDG, which is no longer necessary for maintaining thememory traces, they may have adopted another, as yetundetermined, role (508).However, the view that the HF becomes dispensableafter the conversion of short-term memory in<strong>to</strong> longlastingmemory is disputed, as it is supposed <strong>to</strong> be involved,<strong>to</strong>gether with neocortical areas, in the s<strong>to</strong>rage <strong>and</strong>the retrieval of remote memories (387, 388, 393–395). Inparticular, the HF seems important in the recall of old,detailed, episodic memories that are context dependent(469, 527). This scenario is consistent with the long-termsurvival of adult-born neurons (121, 309), which could actas binding detec<strong>to</strong>r cells capable of memorizing bindingsbetween items (503) or could s<strong>to</strong>re a memory index, i.e.,an index of neocortical areas that are activated by experientialevents <strong>and</strong> contain no intrinsic meaning in termsof representation of external events (538). In these cases,adult-born neurons most probably do not s<strong>to</strong>re the memorytraces themselves but may play a critical role inlinking representations of distinct experience, <strong>and</strong> longterms<strong>to</strong>rage of spatial representations could be achievedby synap<strong>to</strong>genesis occurring within this CA3 (455). Alternatively,adult-born neurons could s<strong>to</strong>re the contextualinformation regarding the episode (the so-called contextualtrace; Ref. 394).B) A ROLE OF CELL DEATH IN STABILIZATION? A surprising phenomenonis that the death of old <strong>and</strong>/or newly bornneurons also plays an important role in enabling hippocampallearning. This may constitute a trimming mechanismreplacing the unnecessary old neurons <strong>and</strong>/or theimmature neurons that have not established learning-relatedsynaptic connections with freshly minted ones. Thismay serve the function of increasing the signal-<strong>to</strong>-noiseratio, thus consolidating the memory trace. This phenomenon,which on one h<strong>and</strong> is consistent with the “use it orlose it” principle <strong>and</strong> on the other h<strong>and</strong> goes against theclassic assumption in learning-induced structural plasticitythat “more is better,” recalls the regressive eventsobserved during development (450).C) IS LEARNING-INDUCED CELL PROLIFERATION INVOLVED IN MEMORYCLEARANCE OR FLEXIBILITY? Learning-induced cell proliferationis not correlated with learning performances, suggestingthat these newly born cells do not directly sustain ongoinglearning <strong>and</strong> may rather be involved in future behaviors.This phenomenon may represent a resetting process ensuringthat the hippocampal system is available <strong>to</strong> processnew memories. This hypothesis is strengthened by theobservation that presenilin-1 knockout mice that presentnormal basal neurogenesis exhibit a reduction in enrichment-inducedcell proliferation <strong>and</strong> better retrieval ofcontextual fear memory traces compared with controlmice (153). Thus a deficiency in the upregulation of cellproliferation induced by the late phase of learning mayprevent memory clearance, thereby impairing forgettingprocesses <strong>and</strong> disabling hippocampal functions. It is alsopossible that this alteration in cell proliferation alters theinferential use of past memories in a novel situation (flexibility)<strong>and</strong> thus leads <strong>to</strong> rigidity <strong>and</strong> perseverance.B. Activation of the Stress Axis: Acute<strong>and</strong> Long-Term Effects on <strong>Neurogenesis</strong>Exposure <strong>to</strong> stressful events prepares animals <strong>to</strong> engagein fight or flight responses, <strong>and</strong> a role of the HF inthese defensive behaviors has been proposed (52, 205).Moreover, as stressful events have deleterious effects onhippocampal integrity (351, 352, 484), the influence ofdifferent types of stress on hippocampal neurogenesis hasbeen studied.1. Effects of stress on neurogenesisIn most studies, a decrease in cell proliferation hasbeen observed after stressful events, but this effect ap-Downloaded from physrev.physiology.org on November 25, 2005Physiol Rev • VOL 85 • APRIL 2005 • www.prv.org


544 ABROUS, KOEHL, AND LE MOALpears highly dependent on the nature <strong>and</strong> the timing ofstress.In male but not female rats, exposure <strong>to</strong> the odor ofa preda<strong>to</strong>r [trimethyl thiazoline (TMT), the major componen<strong>to</strong>f fox feces] downregulates the number of proliferatingcells estimated 2 h, 24 h, <strong>and</strong> 1 wk after a singleBrdU injection (100–200 mg/kg) (149, 537). However, thiseffect is transient as TMT-induced downregulation ofBrdU cell numbers disappears 3 wk after labeling whendividing cells express NeuN or NSE (149). Likewise, inmale tree shrews (Tupaia belangeri), a single exposure <strong>to</strong>an unknown congener, which results in the establishmen<strong>to</strong>f a dominant/subordinate relationship, brings down thenumber of dividing cells (labeled by one injection ofBrdU, 75 mg/kg) in the DG of the subordinate within 2 h(177). In the marmoset (Callithrix jacchus) however, asingle exposure <strong>to</strong> a resident-intruder model of stressreduces the number of newly born cells (2 h after 1 75mg/kg BrdU) in the DG of the intruder (181). Repeated (3or 6 wk) but not acute (2 or 6 h) restrain stress decreasescell proliferation duration-dependently in the SGZ of rats(2 h after 1 200 mg BrdU/kg; Ref. 441). In contrast, theacute stress of cold immobilization <strong>and</strong> a forced swimreduce cell proliferation (1 day after a 1 200 mg/kgBrdU injection before the stress) in the DG <strong>and</strong> not theSGZ, an effect that is normalized within 1 day (209).Chronic, unpredictable stress exerted over 3 wk reducesmi<strong>to</strong>tic activity in the SGZ (<strong>and</strong> not the DG) as estimatedby KI-67 staining. At this time point, another batch ofanimals was injected with BrdU (1 200 mg/kg), <strong>and</strong> 3wk later, the effect of chronic stress on the survival ofBrdU-labeled cells was examined. Because the BrdU cellnumber was unchanged, it was concluded that cell survivalis not influenced by stress. Finally, male tree shrewssubjected <strong>to</strong> a 7-day period of psychosocial stress exhibitreduced cell birth (evaluated 1 day after 1 100 mg/kgBrdU; Ref. 104). This stress-induced reduction in adultborncells leads <strong>to</strong> a decline in neurogenesis, the neuronalphenotype being inferred by the expression of NeuN (441)or NSE (177, 181) 1 mo after labeling.Because stressful events are known <strong>to</strong> activate theHPA axis, leading <strong>to</strong> a rise in plasma levels of corticosterone,<strong>and</strong> <strong>to</strong> activate the release of endogenous opioids,which are involved in defensive behaviors, the role ofthese two fac<strong>to</strong>rs has been evaluated. To confirm theinvolvement of corticosterone, male rats were adrenalec<strong>to</strong>mized,<strong>and</strong> low levels of hormone were res<strong>to</strong>red. Inthese conditions, the prevention of the TMT-induced risein corticosterone blocks the downregulation of cell proliferation(537). In contrast, administration of naltrexone,an opioid antagonist (5 mg/kg, 30 min before TMT exposure),does not attenuate the effect of TMT on cell proliferation,whereas the expression of defensive behaviors(defensive burying, stretch approach) is partially attenuated(219), which indicates a dissociation between TMTinducedcell proliferation <strong>and</strong> defensive behaviors (149).The activity of the HPA axis is known <strong>to</strong> be regulated bycorticotrophin-releasing hormone <strong>and</strong> vasopressin, theroles of which have recently been evaluated. Thus thechronic blockade of CRF1 <strong>and</strong> V1B recep<strong>to</strong>rs (bySSR125543A <strong>and</strong> SSR149415, respectively, 30 mg · kg 1 ·day 1 for 28 days) starting 3 wk after the beginning ofchronic mild stress (CMS for 21 days) reverses the stressinducedreduction of cell proliferation (measured 24 hfollowing 1 75 mg/kg of BrdU injected at the end of thedrug treatment period) (10). These antagonists are alsoable <strong>to</strong> prevent the stress-induced reduction of neurogenesis(by means of NeuN labeling) estimated 30 days afterthe cessation of the drug treatment <strong>and</strong> BrdU pulse (3.75mg/kg every 2honthelast 3 days of the CMS). Thenormalization of neurogenesis is furthermore associatedwith a prevention of the stress-induced hippocampal atrophy.2. Interindividual differences in stress reactivityInterindividual differences in behavioral reactivity <strong>to</strong>stress or <strong>to</strong> novelty <strong>and</strong> coping abilities have been evidencedin our labora<strong>to</strong>ry in an outbred population of rats.Animals are split in<strong>to</strong> two groups according <strong>to</strong> the medianscore of behavioral response, the behaviorally high-reactive(HR) rats, <strong>and</strong> the low-reactive (LR) rats (443, 446).Compared with the LR phenotype, the HR phenotype ischaracterized by a series of adaptive defects of a cognitive<strong>and</strong> motivational nature corresponding <strong>to</strong> a deficit in inhibition<strong>and</strong> control processes (115, 304, 443, 447). Theseinterindividual differences have been related <strong>to</strong> differencesin corticosterone secretion, HR rats displaying ahyperactive HPA axis (305, 443, 444, 446).The behavioral trait of reactivity <strong>to</strong> stress has beenshown <strong>to</strong> predict the level of neurogenesis in the GCL ofthe DG (305). Two weeks after their behavioral characterization,rats were injected with BrdU (3 50 mg/kgspaced by 4-h intervals) <strong>and</strong> killed the next day or 14 dayslater. Cell proliferation was found <strong>to</strong> be negatively correlatedwith locomo<strong>to</strong>r reactivity <strong>to</strong> novelty, <strong>and</strong> when thegroup was split according <strong>to</strong> the median behavioral scorein<strong>to</strong> LRs <strong>and</strong> HRs, cell proliferation in LRs was twice tha<strong>to</strong>bserved in HRs. Survival of nascent neurons was notinfluenced by this behavioral trait. A difference in theHPA axis reactivity most probably underlies the differencesin neurogenesis, primarily due <strong>to</strong> differences in cellgenesis. The functional significance of this difference inDG plasticity remains unclear but may reflect a disruptionof behavioral inhibition in the HRs (187).3. Perinatal manipulationsStudies on stress during the perinatal period havealso led <strong>to</strong> intriguing observations. It is well documentedfrom animal <strong>and</strong> human studies that during the perinatalDownloaded from physrev.physiology.org on November 25, 2005Physiol Rev • VOL 85 • APRIL 2005 • www.prv.org


NEUROGENESIS IN THE ADULT BRAIN 545period the development of an organism is subjected <strong>to</strong>complex environmental influences <strong>and</strong> that deleteriouslife events during development induce neurobiological<strong>and</strong> behavioral defects (207, 557, 569). Indeed, environmentalchanges <strong>and</strong> stimulations have the greatest impactwhen produced in the early stages of life <strong>and</strong> duringdevelopmental periods, leading <strong>to</strong> phenotypical orientationslasting throughout life. These environmental influences,which have been evidenced for decades, have beenexplored at structural <strong>and</strong> mechanistic levels only recently,revealing profound structural changes in the HF(see, for instance, Ref. 353). Stressful events during theprenatal period, such as prenatal stress which consists instressing pregnant dams, or during the postnatal period inthe rat, as for example maternal deprivation, i.e., longlastingseparations of the mum-pup dyad, alter brainchemistry, enhance emotional reactivity, produce cognitiveimpairments, <strong>and</strong> increase vulnerability <strong>to</strong> drugs ofabuse (185, 482, 569). We <strong>and</strong> others have shown thatstressful events during the prenatal (306) <strong>and</strong> the postnatal(300, 367, 431) periods downregulate hippocampalneurogenesis. In prenatally stressed rats, we found thatcell proliferation (4 50 mg/kg of BrdU over 3 days) isreduced from adolescence <strong>to</strong> senescence, indicating anacceleration in the age-related decline of cell proliferationin the GCL. Survival of the newly born cells <strong>and</strong> neuronaldifferentiation (measured with NeuN) are not modified,<strong>and</strong> thus the net reduction in the number of adult-bornneurons is associated with a decrease in the number ofgranule cells from adulthood <strong>to</strong> senescence. Given thatcorticosterone has been implicated in long-term behavioralchanges induced by prenatal stress (280), this downregulationof neurogenesis may result from heightenedcorticosterone secretion (see sect. IID2A). Similarly, maternallydeprived rats are characterized by reduced cellproliferation in the GCL both when infants (proliferationmeasured on postnatal day 21 following 7 days of maternalseparation <strong>and</strong> 7 50 mg/kg BrdU) (300, 431) <strong>and</strong>when adults [proliferation measured at 2–3 mo of agefollowing daily 3-h maternal separation bouts from postnatalday 1 <strong>to</strong> 14 <strong>and</strong> 1 200 mg/kg BrdU 2hor1wkbefore death (367)]. In this recent study, the authorsfound that maternal deprivation reduced the number ofimmature, but not mature, neurons that are added <strong>to</strong> theDG in adulthood (367). Because maternal separation isreported <strong>to</strong> alter the activity of the HPA axis (482), it hasbeen hypothesized that corticosterone mediates the effec<strong>to</strong>f maternal separation on neurogenesis. In fact, it hasbeen shown that the maternal separation-induced decreasein cell proliferation is abrogated by adrenalec<strong>to</strong>myplus corticosterone treatment aimed at res<strong>to</strong>ring basaldiurnal levels of the hormone. The authors proposed thathippocampal precursors might be hypersensitive <strong>to</strong> corticosterone,the diurnal basal <strong>and</strong> stress-induced levels ofwhich are not modified in their paradigm (367). However,because maternal separation is known <strong>to</strong> decrease corticosteroid-bindingglobulin (559), <strong>and</strong> thus enhance thefree, <strong>and</strong> active, fraction of corticosterone reaching thebrain, a higher level of “efficient” hormone could be responsiblefor the effects of maternal separation on hippocampalcell proliferation.Given the involvement of the HF in learning <strong>and</strong>memory, we hypothesized that prenatal stress would lead<strong>to</strong> cognitive deficits via an inhibition of neurogenesis inthe DG. Thus we have shown that prenatal stress is associatedwith an alteration in learning <strong>and</strong> memory performances,<strong>and</strong> more importantly, that the learning-inducedincrease in cell proliferation observed in control animalsis blocked in prenatally stressed rats (306). This result isin accordance with our previous observation that cellproliferation is increased only when the animals reach anasymp<strong>to</strong>tic level of performance, i.e., when the task hasbeen mastered (see sect. IIIA2A <strong>and</strong> Fig. 4). Furthermore,the behavioral deficits induced by prenatal stress, similar<strong>to</strong> those observed in animals exposed <strong>to</strong> X-irradiations(37), confirm an enabling role for hippocampal neurogenesisin cognition.C. Aging <strong>and</strong> Longitudinal Observations1. Neurobiology of the age-related declinein neurogenesisA) INFLUENCE OF AGING ON NEUROGENESIS. Aging is a life-longprocess beginning with conception <strong>and</strong> ending withdeath. Within this developmental continuum, sexualmaturity (2–3 mo in rodents) is used <strong>to</strong> define the star<strong>to</strong>f adulthood. Although the definition of “how old isold” varies depending on species, genetic background,<strong>and</strong> experimental conditions, studies addressing theeffects of age on biobehavioral parameters in rodentshave identified at least three ages: adult (up <strong>to</strong> 10 mo),middle-aged, <strong>and</strong> “old” or “aged” or senescent (from 20mo) (91, 92).The persistence of neurogenesis in the DG of senescentrats was first reported by Kaplan <strong>and</strong> Bell (248,250, 251). An age-related decline in DG neurogenesishas been confirmed in mice <strong>and</strong> rats using several BrdUlabeling pro<strong>to</strong>cols (54, 208, 290, 306, 310, 499), whereasconflicting results have been reported for the SVZ (238,253, 290, 543).The decline in DG neurogenesis, which mainly occursbetween 2 <strong>and</strong> 12 mo of age, does not seem <strong>to</strong> berelated <strong>to</strong> 1) an alteration in adult-born cell survival asthe percentage of surviving BrdU-labeled cells 1 moafter their birth is independent of the age of the animals(54, 208, 269, 310) or 2) a general change in metabolicconditions, as neuronal precursors respond <strong>to</strong> BDNF’sinfluence <strong>to</strong> the same extent throughout life (169).Rather, it may be a decline in proliferative activity thatDownloaded from physrev.physiology.org on November 25, 2005Physiol Rev • VOL 85 • APRIL 2005 • www.prv.org


546 ABROUS, KOEHL, AND LE MOALunderlies the age-related decline in neurogenesis, sincethe number of newly born cells a few hours after labeling(54, 290) <strong>and</strong> the number of nestin-positive cells(391) are lower in aged rats. However, it remains <strong>to</strong> bedetermined whether the reduction in proliferative activityis a consequence of a developmental lengtheningof the cell cycle time <strong>and</strong>/or of a shrinking of theproliferative cell population. Finally, neuronal differentiationseems <strong>to</strong> be delayed with advancing age assuggested by the high percentage of cells that do notexpress a neuronal or an astrocytic phenotype 1 moafter their birth (54, 208, 264, 310, 391).B) FACTORS REGULATING NEUROGENESIS IN THE SENESCENT BRAIN.The progressive decline in cell proliferation could derivefrom inadequate local environmental cues; theaged DG may be under the influence of inhibi<strong>to</strong>ry fac<strong>to</strong>rs<strong>and</strong>/or may lack stimula<strong>to</strong>ry fac<strong>to</strong>rs that sustaindivision, differentiation, <strong>and</strong>/or survival of the newlyborn cells.I) Inhibi<strong>to</strong>ry fac<strong>to</strong>rs. Corticosterone has receivedparticular attention since basal levels of this hormone,known <strong>to</strong> inhibit cell proliferation in young rats (see sect.IID2A), increase during aging (324, 484). Thus, 1 wk afteradrenalec<strong>to</strong>my in old rats, cell proliferation is upregulatedin the GCL (373) <strong>and</strong> in the DG (GCL <strong>and</strong> hilus, Ref. 74).On the other h<strong>and</strong>, survival of the newly born cells isunrelated <strong>to</strong> corticosterone levels (373), <strong>and</strong> most of themdifferentiate in<strong>to</strong> neurons expressing TOAD-64 or NeuN(74, 373).Glutamate is also known <strong>to</strong> inhibit DG neurogenesisin young rats (see sect. IID2B). Similarly, in middleaged<strong>and</strong> aged rats, a single injection of a NMDA recep<strong>to</strong>rantagonist (CGP-43487, 5 mg/kg 2 h after an injectionof BrdU, 200 mg/kg) increases hippocampal cellproliferation 5 days later. By 3 wk, despite a decline inthe number of newly born cells, more cells survived inthe treated aged rats compared with saline aged counterparts(391).II) Activating fac<strong>to</strong>rs. The involvement of the neurosteroidPreg-S in the hippocampal neurogenesis of oldrats has been examined. Indeed, 1) hippocampal levels ofPreg-S in the HF of senescent rats correlate with spatialmemory abilities, 2) intrahippocampal infusion of Preg-Sin cognitively impaired aged rats reverses memory disturbances(551), <strong>and</strong> 3) Preg-S stimulates neurogenesis inthe adult DG (see sect. IID2A). We have shown that icvPreg-S infusion increases cell proliferation in the senescentDG (348). This suggests that the age-related decreasein Preg-S levels could lead <strong>to</strong> a loss of stimulation ofhippocampal neurogenesis <strong>and</strong> <strong>to</strong> subsequent cognitivedeficits. Finally, because Preg-S acts as a negative allostericmodula<strong>to</strong>r of GABA A recep<strong>to</strong>rs, our results supportthe inhibi<strong>to</strong>ry hypothesis of neurodegenerative disorderssuch as Alzheimer’s disease <strong>and</strong> age-related brain impairments,which postulates the involvement of abnormallystrong inhibi<strong>to</strong>ry GABAergic transmission (341).Because adult neurogenesis can be enhanced by theadministration of growth fac<strong>to</strong>rs (see sect. IID2C), theresponsiveness of the aged brain has been the subject ofinvestigation. In particular, a stimula<strong>to</strong>ry effect of IGF-Ihas been examined since 1) IGF-I brain levels decreasewith age (90), 2) IGF-I recep<strong>to</strong>r mRNA is upregulated as afunction of aging <strong>and</strong> cognitive decline (522), <strong>and</strong> 3) IGF-Iinfusion improves cognitive function in aged rats (345).The chronic icv administration of IGF-I (1.2 g/day for 14days) in senescent rats elicits an approximately threefoldincrease in BrdU-labeled cells (BrdU, 2 50 mg · kg 1 ·day 1 on days 7–11) in the SGZ <strong>and</strong> the hilus (310). In a4-wk survival study, IGF-I <strong>and</strong> saline-treated animals displayeda similar number of newly born cells (BrdU-labeledon days 7–11) compared with animals killed after ashort survival delay, indicating that IGF-I acts on cellproliferation rather than on cell survival. Finally, in contrast<strong>to</strong> what was observed in adult rats (1), IGF-I did notmodify cell fate in old animals, with 20% of newly borncells differentiating in<strong>to</strong> neurons (inferred from NeuNexpression). Thus the threefold increase in BrdU-positivecells elicited by IGF-I translated in<strong>to</strong> a threefold increasein adult-born neurons.The influences of FGF-2 <strong>and</strong> HB-EGF were also examinedin 20-mo-old mice (icv infusion of 10 g/ml for 3days concurrent with 50 mg · kg 1 · day 1 BrdU). Oneweek after the cessation of the treatments, the number ofnewly born cells was higher in both the SGZ <strong>and</strong> the SVZ;these cells expressed DCX <strong>and</strong> occasionally GFAP (238).More speculatively, it may be hypothesized thatproinflamma<strong>to</strong>ry cy<strong>to</strong>kines such as interleukin 6 (IL-6)also play an important role in age-related decline in neurogenesis.Indeed, 1) it is involved in the appearance ofage-related cognitive disorders in humans (59, 120, 328,355, 466, 565), 2) chronic stress increases its productionin aged patients (272), 3) IL-6 expression is increased inthe HF of senescence-accelerated mice (539), <strong>and</strong> 4) hippocampalneurogenesis is reduced in adult transgenicmice with chronic IL-6 production (552) or after administrationof bacterial lipopolysaccharide, a potent activa<strong>to</strong>rof IL-6 (138, 372).In summary, precursors that are able <strong>to</strong> enter the cellcycle are still present in the aged brain, <strong>and</strong> low levels ofneurogenesis are linked <strong>to</strong> the accumulation of inhibi<strong>to</strong>rysubstances <strong>and</strong>/or the decrement of stimula<strong>to</strong>ry fac<strong>to</strong>rs.The mechanisms of action of these modula<strong>to</strong>rs are stillunknown <strong>and</strong>, in particular, it remains <strong>to</strong> be determinedwhether they act on the length of the cell cycle or on thenumber of dividing cells. Whatever the underlying mechanism,these results point <strong>to</strong> possible pharmacologicalactions for preventing or treating age-related cognitiveimpairments.Downloaded from physrev.physiology.org on November 25, 2005Physiol Rev • VOL 85 • APRIL 2005 • www.prv.org


NEUROGENESIS IN THE ADULT BRAIN 5472. Functional implications of age-related modulationof neurogenesis in memoryThe hypothesis that the decline in neurogenesis maybe responsible for age-related alteration in cognitive functionhas been tested by examining the influence of anenriched environment, <strong>and</strong> by taking advantage of theexistence of interindividual differences in age-related cognitivefunctions. The influence of enriched environmentson spatial memory <strong>and</strong> on neurogenesis (cell proliferation,cell survival, <strong>and</strong> neuronal differentiation) has beenstudied in 18-mo-old mice (269). Although the effects arenot as strong as those observed in young animals, after 68days of enrichment, neuronal differentiation is enhancedwhereas cell proliferation <strong>and</strong> cell survival remain unchangedcompared with nonenriched aged animals; performancesin spatial memory are slightly improved. Asimilar conclusion was reached when middle-aged femalemice were raised for 10 mo in an enriched environment(264), which indicates that as was found in younger subjects(see sect. IIIA1A), long-term exposure <strong>to</strong> an enrichedenvironment in old subjects does not afford any furtherbeneficial effect compared with short-term exposure asneither cell proliferation nor cell survival are increased. Inthis study, the improvement in learning parameters observedafter enrichment was suggested <strong>to</strong> be aspecific,resulting from better adaptive skills in exploration,greater locomo<strong>to</strong>r aptitude, <strong>and</strong> endurance following enrichment,leading the authors <strong>to</strong> suggest that the contributionof adult hippocampal neurogenesis <strong>to</strong> hippocampalplasticity might require a physically active pursuit asopposed <strong>to</strong> a passive sensory stimulation (264).Large individual differences exist in cognitive abilities,<strong>and</strong> all aged rats do not exhibit cognitive disorders;some rats perform as well as young controls, whereasothers present spatial memory impairments (457). Theexistence of such individual differences has been used asa naturalistic strategy <strong>to</strong> study the neurobiological correlatesof cognitive aging, <strong>and</strong> it has been shown that theextent of memory dysfunction is related <strong>to</strong> the magnitudeof changes occurring in the HF. Following a characterizationof the cognitive abilities of aged rats in the watermaze, we have shown (Fig. 6) that neurogenesis (i.e., cellproliferation, cell determination, <strong>and</strong> cell survival) ishigher in cognitively unimpaired rats than in impairedones (130).Because age-impaired <strong>and</strong> -unimpaired rats did notdiffer in granule cell number, as previously described(458), it seems that the rejuvenation of granule cellsrather than a change in their absolute number is the mainfac<strong>to</strong>r sustaining memory formation. In other words, successfulaging, i.e., preserved cognitive functions, is associatedwith the maintenance of a relatively high neurogenesislevel, whereas pathological aging, i.e., memorydeficits, is linked <strong>to</strong> exhaustion of neurogenesis. Thesedifferences in hippocampal neurogenesis may be due <strong>to</strong>differences in stimula<strong>to</strong>ry <strong>and</strong>/or inhibi<strong>to</strong>ry substances,which may respectively prevent or accelerate pathologicalaging. We propose that the age-related increase incorticosterone levels, resulting from repeated stressfulevents, may lead, through life-span inhibition of neurogenesis,<strong>to</strong> defects in hippocampal networks that wouldbe responsible for cognitive dysfunctions. The observationthat lowering corticosterone levels in middle-agedanimals significantly increases hippocampal neurogenesis<strong>and</strong> postpones the onset of spatial memory deficits supportsthis hypothesis (M. F. Montaron, E. Drapeau, C.Aurousseau, M. Le Moal, P. V. Piazza, <strong>and</strong> D. N. Abrous,unpublished data).During pathological aging, alterations in hippocampalneurogenesis most probably mirror a more generalfailure in the mechanisms underlying neuronal plasticity(364). Similarly, it has been shown that lipofuscin deposits,an index of cell suffering, are higher in aged rats withimpaired cognitive function (64) <strong>and</strong> are decreased inaged mice housed in an enriched environment (264). According<strong>to</strong> the hypothesis of the “Red Queen Theory” (5),during aging there is competition for the available plasticity<strong>to</strong> compensate for neuronal degeneration or <strong>to</strong> s<strong>to</strong>renew information. Thus, in pathological aging, hippocampalplasticity is exhausted, <strong>and</strong> the capacity of the oldDownloaded from physrev.physiology.org on November 25, 2005FIG. 6. Hippocampal neurogenesis depends onthe cognitive status of the aged rats. To demonstratea quantitative correlation between neurogenesis <strong>and</strong>performance in a hippocampal-dependent test, we<strong>to</strong>ok advantage of the well-established presence ofindividual differences in spatial memory abilitieswithin a population of old rats. Indeed, in the watermaze, some old individuals show clear impairment inspatial reference memory while others are not impaired<strong>and</strong> exhibit cognitive capacities similar <strong>to</strong>those of younger individuals. We found that cell proliferation(A), the survival of adult-born cells (B), <strong>and</strong>the number of adult-born neurons (C) are higher inthe dentate gyrus of aged unimpaired (AU) than agedimpaired rats (AI). *P 0.05, **P 0.01 comparedwith AU. [Modified from Drapeau et al. (130).]Physiol Rev • VOL 85 • APRIL 2005 • www.prv.org


548 ABROUS, KOEHL, AND LE MOALbrain <strong>to</strong> acquire new information <strong>and</strong> <strong>to</strong> compensate forongoing degenerating processes is impeded.D. Involvement of <strong>Neurogenesis</strong> in Pathologies1. Pathological conditions associated with anupregulation of neurogenesisA) EPILEPSY. Human temporal lobe epilepsy (TLE) isassociated with atrophy of the HF, an effect known sincethe beginning of the 19th century, <strong>and</strong> a loss of hippocampal<strong>and</strong> DG neurons (57, 384). In this pathology, thedentate granule cells give rise <strong>to</strong> abnormal axonal projectionin<strong>to</strong> the supragranular inner molecular layer of theDG <strong>and</strong> <strong>to</strong> an aberrant reorganization of the mossy fibers(MF) in the CA3 subfield of Ammon’s horn (529). It hasbeen suggested that the sprouting of MF is relevant in thehyperexcitability of the TLE (426). This has led <strong>to</strong> investigationin<strong>to</strong> whether newly born cells are important inthe genesis of dentate MF sprouting (MFS) <strong>and</strong> participatein the reorganization of the network.Kindling, an animal model of TLE, is characterized bya permanent epileptic state produced by a series of seizuresinduced by electrical stimulations made in variousbrain areas such as the hippocampus (42), the perforantpath (396), <strong>and</strong> the amygdala (425, 492). Single <strong>and</strong> repeatedkindling stimulations in the ventral CA1 regioninduce a facilitation of mi<strong>to</strong>tic activity 2 wk later (42).Stimulation in the perforant pathway leads <strong>to</strong> a markedincrease in the number of BrdU-labeled cells as early as 3days after the last stimulation, whereas repeated kindlingdoes not evoke further stimulation of cell proliferation(396). Cell division is in fact suppressed, suggesting adiminished efficiency of repeated or prolonged stimulation<strong>to</strong> elicit a neurogenic response. In the amygdalakindling model, it has been reported that during the earlyphases of kindling, when focal seizures are present, DGcell proliferation remains unchanged, whereas it is upregulatedduring the later phase of kindling, when mo<strong>to</strong>rseizures are present (425, 492).In various chemoconvulsant (pilocarpine or kainate)models of human TLE, the induction of seizures is accompaniedby a dramatic increase in neurogenesis in the SGZof the DG (430). This increase is observed after a latentperiod of a few days following the status epilepticus (SE)<strong>and</strong> persists during the first week, before levels of neurogenesisreturn <strong>to</strong> baseline over the following weeks (396,430). The recruitment of proliferative cells after kainatetreatment occurs preferentially in GFAP cells, suggestingan increase in proliferative glial-like astrocytes (type B)(223). The expression patterns of multiple bHLH transcriptionfac<strong>to</strong>rs have been studied following epilep<strong>to</strong>genesis;some were found <strong>to</strong> be increased (Mash1, Id2), somedecreased (Hes5), <strong>and</strong> others unchanged (NeuroD, NeuroD2,Id3, <strong>and</strong> Math2), suggesting that molecules controllingcell-fate decisions in the developing dentate gyrus arealso operative during seizure-induced neurogenesis (140).The newly born cells induced by seizures, which survivefor up <strong>to</strong> 4 wk <strong>and</strong> differentiate in<strong>to</strong> neurons (430), migrate<strong>to</strong> abnormal locations such as the CA3 cell layer, thedentate hilus <strong>and</strong> inner molecular layer (108, 430, 487).These ec<strong>to</strong>pic granulelike neurons retain many, but notall, of their granule cell intrinsic properties (487). Incidentally,ec<strong>to</strong>pic locations of SVZ-derived neuroblasts havealso been observed in the striatum <strong>and</strong> the cortex followingpilocarpine treatment (428).Seizure-induced neurogenesis appears <strong>to</strong> precedeMFS, suggesting that newly born cells could be importantin the genesis of the ec<strong>to</strong>pic innervation of Ammon’s hornby dentate fibers. This hypothesis has been tested bycoadministrating kainate or pilocarpine with cycloheximide(CHX), a protein synthesis inhibi<strong>to</strong>r known <strong>to</strong> abolishMFS (40). Contrary <strong>to</strong> what was expected, CHX didnot influence chemoconvulsant-induced neurogenesis, indicatingthat its effect on MFS is independent of cellproliferation (97). Similarly, a single session of X-raytreatment, which attenuates pilocarpine-induced neurogenesis,fails <strong>to</strong> prevent MFS (427), indicating that neurogenesisupregulation is not sufficient <strong>to</strong> generate MFS.Thus the participation of the newly born cells in networkreorganization <strong>and</strong> in the recurrence of epileptic seizure isstill open <strong>to</strong> debate.The mechanisms by which seizure activity stimulatesneurogenesis are unknown. They may involve electricalactivation since 1) physiological activity such as LTPelicitedmossy fiber stimulation is sufficient <strong>to</strong> increaseneurogenesis in the DG (118), <strong>and</strong> 2) pure electrical activation,consisting in a continuous stimulation of the perforantpath, elicits focal hippocampal seizure discharges<strong>and</strong> increases BrdU cell number 6 days later (430). Seizure-inducedupregulation of neurogenesis may also involve1) an alteration in excita<strong>to</strong>ry amino acid transmissiondue <strong>to</strong> degeneration of the en<strong>to</strong>rhinal afferences <strong>to</strong>the granule neurons of the DG (430); 2) activation by thedying cells (42, 50); 3) a change in bFGF, as kainateinducedupregulation of neurogenesis is blocked in micelacking bFGF, an effect reversed by gene transfer (582);4) changes in other neurotrophic fac<strong>to</strong>rs (145, 164, 172,583); <strong>and</strong> 5) activation of the 5-HT 1A recep<strong>to</strong>r as infusionsof a 5-HT 1A recep<strong>to</strong>r antagonist (WAY-100,635) impedethe increase in pilocarpine-induced neurogenesis whereascell death, MF sprouting, <strong>and</strong> the onset of spontaneouslyrecurring seizure are not prevented (451).In summary, neurogenesis is altered by epileptiformstatus, <strong>and</strong> surprisingly maintenance of the newly borncells (in homotypic <strong>and</strong> ec<strong>to</strong>pic locations) is systematicallyobserved in the different models of epilepsy. Theabnormal circuits may contribute <strong>to</strong> the unstable hippocampalcircuitry <strong>and</strong> the development of chronic TLE.As proposed by Gray <strong>and</strong> Sundstrom (188), the DG couldDownloaded from physrev.physiology.org on November 25, 2005Physiol Rev • VOL 85 • APRIL 2005 • www.prv.org


NEUROGENESIS IN THE ADULT BRAIN 549“regulate as a gate with respect <strong>to</strong> controlling the propagationof seizure with granule cells controlling thethroughput of epileptiform activity transition through theHF by virtue of a feed forward inhibi<strong>to</strong>ry pathway.” Thesealterations in neurogenesis may participate in the memorydysfunction associated with epilepsy (530). Althoughunverified, this latter finding suggests that the presence ofmore new neurons is not necessarily associated with agood memory <strong>and</strong> that neurogenesis <strong>and</strong> memory functionmay follow an inverted U-shaped curve.B) ISCHEMIA. Until recently it was thought that the functionalrecovery that occurs in some cases following braindamage (such as stroke <strong>and</strong> trauma, see sect. IIID1D) wasrelated <strong>to</strong> the remodeling of neuronal pathways in nondamagedareas. The discovery of adult neurogenesisprompted researchers <strong>to</strong> investigate the role played bynew neurons in recovery of lost functions.Transient global ischemia, induced in gerbils <strong>and</strong> ratsby bilateral common carotid artery occlusions, increasescell proliferation in the DG in a dose-dependent manner(260, 315, 575, 576). A peak in cell proliferation occurs 1–2wk after ischemia, <strong>and</strong> a quarter of the newly generatedcells die during the first month (315). During the first 2 wkafter their birth, the newly born cells transiently expressNeuroD <strong>and</strong> DCX (258). Those cells that survive differentiatein<strong>to</strong> granule neurons expressing NeuN or MAP2 at 1mo of age (258, 315). They acquire functional features ofmature neurons as NMDA stimulation induces the phosphorylationof ERK in 34 <strong>and</strong> 61% of BrdU-NeuN labeledcells of 1 <strong>and</strong> 2 mo of age, respectively (258).After focal cerebral ischemia, induced by unilateralmiddle cerebral artery occlusion (MCAO) (24, 237, 589,590), cell genesis is increased <strong>to</strong> various degrees in theipsilateral DG (as well as in the SVZ, OB, <strong>and</strong> cortex) overa period of several weeks. Increased BrdU incorporationin the DG (as in the others structures) contralateral <strong>to</strong> theinfarct, occurring following a lapse of a few days, suggestsa role for diffusible or humoral fac<strong>to</strong>rs (237, 534). Thenewly born cells differentiate in<strong>to</strong> granule neurons withoutany apparent degeneration (590). The enhancement ofBrdU-labeled cells in the OB <strong>and</strong> the cortex has beenattributed <strong>to</strong> the migration of neuronal precursors fromthe SVZ <strong>to</strong>wards their normal target or <strong>to</strong>wards the infarct,respectively (588). The presence of cell clusters incortical areas, in the vicinity of blood vessels, also suggeststhe recruitment of resident quiescent stemlike cellsor the infiltration of blood-borne cells (589). New cellsarising from the SVZ also colonize the striatum afterischemia, 20% of which survive (representing 0.2% of thecell loss) <strong>and</strong> assume the phenotype of cells lost due <strong>to</strong>the lesion (24, 239, 429).<strong>Neurogenesis</strong> upregulation in the DG has been attributed<strong>to</strong> cell death in the en<strong>to</strong>rhinal cortex or the CA1region (46), a hypothesis not always verified (25, 315). Ithas been proposed that the neuronal damage that followsischemia involves the release of glutamate <strong>and</strong> overstimulationof glutamate recep<strong>to</strong>rs, which in turn upregulatesneurogenesis. Indeed, the blockade of NMDA or AMPA/kainate glutamate recep<strong>to</strong>rs by specific antagonists(MK801; NBQX) inhibits the stroke-induced increase inneurogenesis (25, 46); however, these effects are surprisingin light of the stimula<strong>to</strong>ry influence of glutamate recep<strong>to</strong>rblockade on neurogenesis in control brains (seesect. IID2B).Mi<strong>to</strong>tic fac<strong>to</strong>rs such as bFGF are certainly involvedas ischemia-induced upregulation of neurogenesis isblocked in mice genetically deficient in bFGF (582) <strong>and</strong> isincreased following adenovirally mediated transfer ofbFGF (347). Chronic icv infusions of bFGF <strong>and</strong> EGFgreatly enhance the potential of endogenous progeni<strong>to</strong>rs<strong>to</strong> proliferate in response <strong>to</strong> ischemia <strong>and</strong> the cells thenregenerate CA1 pyramidal neurons. The regenerated neuronsare integrated in<strong>to</strong> the neural circuitry as they receiveafferences, project on<strong>to</strong> their normal target, thesubiculum, <strong>and</strong> reverse the ischemia-induced deficits incognitive functions (399). This constitutes the most impressiveexample of network reconstruction leading <strong>to</strong>behavioral recovery that takes advantage of adult endogenousneurogenesis. Infusion of VEGF (0.54 g/day ondays 1–3 of reperfusion concurrent with 2 50 mg ·kg 1 · day 1 BrdU) increases the number of adult-borncells 1 mo (<strong>and</strong> not 3 days) after labeling, beyond theincrease caused by ischemia (526). Ischemia-induced cellproliferation, measured within 3 wk post-BrdU pulse (2 50mg · kg 1 · day 1 BrdU injected for 6 days starting 1 dayafter MCAO), is also increased by icv infusion of IGF-I <strong>and</strong>glial-derived neurotrophic fac<strong>to</strong>r (117).Modulation of cell birth by ischemia also involves theproduction of several classes of inflamma<strong>to</strong>ry moleculemetabolites. Administration of inhibi<strong>to</strong>rs of cyclooxygenase(Cox; i.e., acetylsalicylic acid, indomethacin, NS398),the rate-limiting enzyme for prostagl<strong>and</strong>in synthesis, curtailsischemia-induced proliferation after transient globalischemia in adult gerbils or rats (293, 485, 547). Mutationof one of the Cox isoenzymes, Cox-2, suppresses theischemia-induced increase in hippocampal cell proliferation(485). Cox-2 may affect neurogenesis through theproduction of prostagl<strong>and</strong>in E 2 (547), which may act directlyvia prostagl<strong>and</strong>in EP3 recep<strong>to</strong>rs expressed in theGCL (397) or indirectly through bFGF (477). Alternatively,the production of NO, a key fac<strong>to</strong>r in inflammation,may be at play as administration of a NO donor (DETA/NONOate) <strong>to</strong> rats subjected <strong>to</strong> MCAO significantly increasescell proliferation in the SVZ <strong>and</strong> the DG, <strong>and</strong>significantly improves neurological recovery (587). Thisenhanced neurogenesis is certainly associated with theactivation of inducible NOS since its inhibition (by aminoguanidine)prevents ischemia-induced neurogenesis.Furthermore, stroke-induced neurogenesis is abolished inmice lacking the iNOS gene (590). The participation ofDownloaded from physrev.physiology.org on November 25, 2005Physiol Rev • VOL 85 • APRIL 2005 • www.prv.org


550 ABROUS, KOEHL, AND LE MOALleukotrienes, another key class of inflamma<strong>to</strong>ry molecules,has been postulated in cell proliferation induced bystroke (340). Finally, fac<strong>to</strong>rs produced as part of theischemia-hypoxic response such as the cy<strong>to</strong>kine erythropoietin(507) <strong>and</strong> the recently discovered “stem cell fac<strong>to</strong>r”(SCF) acting through the c-kit recep<strong>to</strong>r tyrosine kinase(235), may also modulate neurogenesis afterischemia.In summary, enhancement of neurogenesis in the DG<strong>and</strong> other brain regions may compensate for disconnectedcircuits that occur after ischemia <strong>and</strong> contribute <strong>to</strong>the improvement of functional outcome. Exploiting thisplasticity potential of the ischemic brain may provide apossible strategy for enhancing recovery in patients sufferingfrom this injury.C) HUNTINGTON’S DISEASE. Hunting<strong>to</strong>n’s disease (HD) is aneurodegenerative disease that leads <strong>to</strong> neuronal loss inthe caudate-putamen. The discovery of progeni<strong>to</strong>r cells inthe human brain (144) has raised the possibility thatprogeni<strong>to</strong>rs from the SVZ may provide a source of replacement.Postmortem analysis of control <strong>and</strong> HD humanbrains has revealed that cell proliferation, evaluatedwith PCNA, is increased in the subependymal layer in HDbrains as a function of the severity of the illness (103).The newly generated cells expressed neuronal <strong>and</strong> glialmarkers (Tuj1 <strong>and</strong> GFAP, respectively).These pioneer data indicate that the diseased adultbrain is still capable of neuronal regeneration, whichopens new avenues in the treatment of neurodegenerativediseases.D) OTHER TYPES OF CEREBRAL INJURY. Traumatic brain injury(TBI), the most common cause of death in developedcountries, increases cell proliferation in the SGZ (as wellas in the SVZ <strong>and</strong> the cortex; Refs. 107, 319, 459). Thenumber of newly born cells increases as early as 24 hpost-TBI; the cells accumulate over time <strong>and</strong> expressTOAD-64 <strong>and</strong> NeuN (65%) 9 <strong>and</strong> 21 days, respectively,after the last BrdU injection (200 mg/kg for 9 days; Ref.107). Similarly <strong>to</strong> what can be observed after ischemia,DETA/NONOate administration (for 7 days starting 1 dayafter TBI) <strong>to</strong> rats subjected <strong>to</strong> TBI significantly increasesproliferation, survival, migration, <strong>and</strong> differentiation ofneural progeni<strong>to</strong>rs in the DG (as well as other brainstructures) <strong>and</strong> significantly improves neurological functionaloutcome (319).2. Pathologies associated with a downregulationof neurogenesisA) AFFECTIVE DISORDERS. In unipolar major depression,bipolar mood disorders, <strong>and</strong> other chronic diseases associatedwith affective disorders [posttraumatic stress disorder(PTSD), Cushing’s disease], brain imaging studieshave consistently described hippocampal atrophy (60,195, 329, 504, 518, 519, 550). Although a role for neurogenesisin mood disorders is speculative, it has beensuggested that a fall in neurogenesis in the DG contributes,<strong>to</strong>gether with atrophy <strong>and</strong> death of hippocampalneurons, <strong>to</strong> hippocampal attrition.I) Antidepressants. It has been suggested that biogenicamine deficiency underlies mood disorders, <strong>and</strong>indeed, treatments increasing extracellular levels of sero<strong>to</strong>ninor norepinephrine are effective in the remediationof depressive symp<strong>to</strong>ms. Because sero<strong>to</strong>nin <strong>and</strong> norepinephrinestimulate cell genesis (see sect. IID2B <strong>and</strong> Table1), <strong>and</strong> given that it takes weeks for antidepressants <strong>to</strong>take effect, it has been proposed that they may exert theiraction via a remodeling of neuronal networks by promotingeither neurogenesis or the survival of hippocampalneurons believed <strong>to</strong> be endangered (131). Chronic (<strong>and</strong>not acute) treatments with different classes of antidepressantssuch as a selective sero<strong>to</strong>nin reuptake inhibi<strong>to</strong>r(fluoxetine, 5 or 10 mg · kg 1 · day 1 for 11–28 days), aselective norepinephrine reuptake inhibi<strong>to</strong>r (reboxetine,mg·kg 1 · day 1 for 21 days), a monoamine oxidaseinhibi<strong>to</strong>r (tranylcypromine, 7.5 mg · kg 1 · day 1 the firstweek <strong>and</strong> 10 mg · kg 1 · day 1 the second week), <strong>and</strong>tricyclic antidepressants (TCAs, imipramine <strong>and</strong> desipramine:20 mg · kg 1 · day 1 for 28 days) increase hippocampalcell proliferation evaluated 2 or 24 h after thelast BrdU pulse (1 75 mg/kg or 4 75 mg/kg every 2 h)performed on the final day or 4 days after the treatment(335, 483). Three weeks after labeling, most of the newlyborn cells express NeuN (70%), independently of theclasses of antidepressants (335, 483). It was furthershown that a fluoxetine- but not an imipramine-inducedincrease in cell proliferation is mediated by 5HT 1A recep<strong>to</strong>rs,since it is abolished in mice lacking 5HT 1A recep<strong>to</strong>rs(483).To determine effects of fluoxetine on cell survival,BrdU (4 75 mg/kg spaced by 2 h) was administeredbefore intitiation of the chronic treatment (5 mg/kg for 14days). Neither survival nor maturation of the cells, evaluated28 days after the last BrdU injection, was influencedby fluoxetine treatment (335). The preclinical contributionof hippocampal neurogenesis has been examined inthe learned helplessness paradigm, a classical animalmodel for depression, <strong>and</strong> it was reported that inescapablestress induced a decrease in cell proliferation thatwas reversed, <strong>to</strong>gether with behavioral despair, by a 7-daycourse of fluoxetine (10 mg/kg) (334). A strongest linkbetween “depression” <strong>and</strong> neurogenesis was evidencedfollowing X-irradiation that blocked the effects of fluoxetine<strong>and</strong> imipramine on both novelty-suppressed feeding(another test used <strong>to</strong> assess chronic antidepressant effects)<strong>and</strong> neurogenesis (483).Although the etiologies of mood disorders are numerous,ranging from various environmental fac<strong>to</strong>rs <strong>to</strong> geneticvulnerability, several lines of evidence suggest thatstress-induced cellular changes in the adult HF, mediatedDownloaded from physrev.physiology.org on November 25, 2005Physiol Rev • VOL 85 • APRIL 2005 • www.prv.org


NEUROGENESIS IN THE ADULT BRAIN 551by an upregulation of the HPA axis, contribute <strong>to</strong> thepathophysiology of these disorders, at least in animalmodels. The impact of antidepressants on neurogenesiswas therefore tested in various stress models. In adultmale tree shrews exposed <strong>to</strong> chronic psychosocial stress(see sect. IIIB1), chronic treatment with tianeptine (50mg·kg 1 · day 1 for 28 days), a modified TCA, reversedthe stress-induced decline in hippocampal volume, hippocampalactivity, <strong>and</strong> hippocampal neurogenesis (104).In rat pups, fluoxetine treatment (5 mg/kg for 7 daysconcurrent with 50 mg/kg BrdU) counteracted the downregulationof cell proliferation observed in the DG of2-wk-old maternally separated pups (300). In a chronicmild stress model (CMS; see sect. IIIB1), chronic treatmentwith the antidepressant fluoxetine (10 mg/kg for 28days starting 3 wk after the beginning of the CMS) reversedthe stress-induced reduction of cell proliferation<strong>and</strong> prevented the stress-induced reduction of neurogenesis<strong>and</strong> hippocampal atrophy (10).II) Electroconvulsive therapy. Electroconvulsivetherapy (ECT) is one of the most effective treatments formajor depression, especially in subjects who do not respond<strong>to</strong> antidepressants, but its mechanisms of actionremain largely unknown. Electroconvulsive seizure (ECS)differs from kindling <strong>and</strong> chemoconvulsant treatments inthat it does not lead <strong>to</strong> an epileptiform state <strong>and</strong> does notcause cell death. ECS (one daily for 10 days) enhances (2or 24 h after a BrdU pulse) cell proliferation in the DG,<strong>and</strong> the newly born cells survive for at least 3 mo, a timepoint when most of them express NeuN (75%) <strong>and</strong> fewGFAP (13%) (331, 335). This proliferative effect is dosedependentas a series of seizures further increases neurogenesis(331). The neurogenic efficacy of ECS has beeninvestigated in conditions mimicking stress-induced depression,i.e., in animals with high corticosterone levels.The downregulation of neurogenesis (75%) in the GCL ofrats chronically treated with corticosterone was eliminatedby ECS performed at the end of the treatment (212).Because ECS upregulates several fac<strong>to</strong>rs described asstimulating neurogenesis, such as bFGF, neuropeptide Y,VEGF, BDNF, <strong>and</strong> Cox-2 (404, 406), these fac<strong>to</strong>rs arelikely media<strong>to</strong>rs of the proliferative effect of ECS. Amongthem, BDNF deserves special mention as ECS-inducedMFS is diminished in BDNF knockout mice (549).III) Mood stabilizers. Chronic treatment with lithiumcan reverse the hippocampal attrition in sufferers of manicdepressiveillness (MDI) <strong>and</strong> enhance the levels of N-acetylaspartate, a putative marker of neuronal viability(375, 376, 544). Chronic treatment of mice with lithium(2.4 g/kg during 3–4 wk) increases the number of BrdUlabeledcells by 25% 1 day after BrdU treatment (1 50mg/kg over 12 days), two-thirds of which coexpressNeuN. Interestingly, the expression of bcl-2, an antiapop<strong>to</strong>ticregula<strong>to</strong>r, is also increased suggesting that survivalof the newly born neurons might be enhanced (84).IV) Summary. These results suggest that antidepressants,ECT, <strong>and</strong> mood stabilizers may exert some of theirtherapeutic effects on mood disorders by stimulating neurogenesis.They also suggest that alternative strategies forstimulating neurogenesis may provide new avenues forthe treatment of mood disorders. Since a fall in neurogenesishas been associated with an alteration in cognitivefunctions, further studies are needed <strong>to</strong> clarify the involvemen<strong>to</strong>f neurogenesis, <strong>and</strong> hippocampal function, inthe development of cognitive deficits observed in depressionin young <strong>and</strong> elderly patients.B) SCHIZOPHRENIA AND NEUROLEPTICS. Developmental dysfunctionof the HF is thought <strong>to</strong> play a major role in thepathogenesis of schizophrenia (206, 488), <strong>and</strong> defectssuch as a reduction in hippocampal volume (53, 400, 558),hippocampal shape deformations (101), abnormalities inthe GCL (357), the MF pathway (170), the hippocampalcell density (41, 150, 233) <strong>and</strong> orientation (93, 288), <strong>and</strong> inseveral cellular markers (203, 313) have been reported.These changes are believed <strong>to</strong> underlie the progressivedeficit in cognition that is a hallmark of schizophrenia.Interestingly, models of aberrant neurogenesis (prenatalstress, X-rays, or MAM manipulations) are thought <strong>to</strong>reproduce some of the behavioral characteristics associatedwith schizophrenia (313).The influence on neurogenesis of chronic treatmentwith the typical neuroleptic haloperidol, a mainstay in thetreatment of schizophrenia, has been evaluated, leading<strong>to</strong> controversial results. Indeed, haloperidol has beenshown <strong>to</strong> increase (110) or <strong>to</strong> have no effect (199, 335,483, 562) on neurogenesis in the rodent DG. Differences indosage, time course of drug treatment, species <strong>and</strong> age ofthe animals, <strong>and</strong> BrdU labeling pro<strong>to</strong>col could account forthe observed discrepancy. On the other h<strong>and</strong>, chronictreatment with atypical neuroleptics such as risperidone(0.5 mg · kg 1 · day 1 for 21 days) or olanzapine (2 mg ·kg 1 · day 1 for 21 days) increases cell proliferation (24 hafter 1 100 mg/kg BrdU on the 20th day of neuroleptictreatment) in the SVZ but not in the DG (562). A low doseof clozapine (0.5 mg · kg 1 · day 1 for 4 wk before a singleinjection of BrdU at 200 mg/kg) enhances the number ofBrdU-IR cells in the DG; however, this effect is transientas the newly born cells do not survive 3 wk after labeling.Although these data clearly show that further work isrequired <strong>to</strong> sort out discrepancies in results, they alsosuggest that schizophrenia may be associated with reducedneurogenesis in the DG, where normal levels couldbe reestablished by neuroleptic treatment, a hypothesisthat awaits confirmation.C) ADDICTION TO DRUGS OF ABUSE. Long-term neuroadaptivechanges in the mesolimbic dopaminergic system haveclassically been observed in drug abuse (283, 402). Apotential role of the HF in addiction has been suggestedbased on the observation that both a reduction in hippocampalvolume <strong>and</strong> structural abnormalities are ob-Downloaded from physrev.physiology.org on November 25, 2005Physiol Rev • VOL 85 • APRIL 2005 • www.prv.org


552 ABROUS, KOEHL, AND LE MOALserved in patients with chronic alcoholism (4, 112, 357,524) <strong>and</strong> in psychostimulant addicts (33). In animals,chronic treatment with different drugs of abuse leads <strong>to</strong>structural changes in the HF (308, 432, 461, 463), <strong>to</strong> cellloss (563), <strong>and</strong> <strong>to</strong> LTP alterations (109, 158, 191, 200, 202,449). The possible involvement of the HF in addiction issupported by the observation that 1) inhibiting the activityof the calcium/calmodulin-dependent protein kinase IIimpairs drug conditioning (321, 535) <strong>and</strong> attenuates thedependence on morphine <strong>and</strong> relapse (321), 2) dorsalhippocampal lesions disrupt acquisition of cocaine conditioning(365), 3) electrical stimulation of the glutamatergicefferent pathway of the HF <strong>to</strong> the nucleus accumbensreinstates cocaine-seeking behavior (561) <strong>and</strong> D-amphetamine self-administration behavior (533), <strong>and</strong> 4)inactivation of the ventral subiculum decreases reinstatemen<strong>to</strong>f cocaine-seeking behavior (525). These resultssuggest that memory traces of the association between aspecific context <strong>and</strong> the availability of the drug could bes<strong>to</strong>red in the HF, <strong>and</strong> the hypothesis that drug addiction isan aberrant form of learning mediated by maladaptiverecruitments of hippocampal-dependent memory systemhas recently gained interest (45, 147, 401, 462, 571). In thiscontext, the involvement of neurogenesis in addiction hasbeen studied by examining the influence of several drugsof abuse on adult hippocampal neurogenesis <strong>and</strong> by comparingneurogenesis in animals that differ in their vulnerability<strong>to</strong> developing drug abuse.I) Opiates. Among the multiple actions of opiates inthe brain, their influence on cognition <strong>and</strong> reward is withouta doubt the most relevant parameter with regard <strong>to</strong>opiate addiction. Eisch et al. (137) were the first <strong>to</strong> demonstratethat the exogenous chronic (daily implantationof subcutaneous pellets containing 75 mg morphine during5 days) <strong>and</strong> not acute (10 mg/kg) administration ofmorphine decreases cell proliferation in the GCL by 28%(measured 2 h after an injection of BrdU, 100 mg/kg, onday 6). When cells are allowed <strong>to</strong> mature for 4 wk,survival of BrdU-labeled cells is halved <strong>and</strong> 90% of thenewly born cells differentiate in<strong>to</strong> neurons. More relevant<strong>to</strong> addiction, the influence of heroin self-administrationhas been investigated. After 26 days of heroin intake (60g ·kg 1 · injection 1 ), a downregulation of cell proliferation(30%) is observed in both the GCL <strong>and</strong> the hiluswhile cell death is unaffected. The downregulation of cellproliferation is independent of corticosterone secretion(137), a hormone known <strong>to</strong> have a major influence inaddiction (343). Finally, it was recently reported that invitro reduced signaling through - <strong>and</strong> -opioid recep<strong>to</strong>rspresent on adult hippocampal progeni<strong>to</strong>rs, although decreasingcell proliferation, leads <strong>to</strong> a net increase in thenumber of newly generated neurons (437).II) Nicotine. The neuroactive compound nicotine isconsidered <strong>to</strong> be responsible for the development <strong>and</strong> themaintenance of <strong>to</strong>bacco addiction. To analyze the effectsof nicotine abuse on hippocampal neurogenesis, rats weretrained <strong>to</strong> self-administer nicotine (0.02, 0.04, <strong>and</strong> 0.08mg·kg 1 · infusion 1 ) for 42 days. A profound decreasein hippocampal cell proliferation was observed for thetwo highest doses of nicotine 1 day after BrdU pulse (4 50 mg/kg on days 39–41). This effect is not simply due <strong>to</strong>a difference in blood flow leading <strong>to</strong> a decrease in BrdUavailability, since nicotine intake does not affect cell proliferationin the SVZ. Half of the newly born cells in theDG were found <strong>to</strong> express NeuN. In parallel with thedecrease in cell proliferation, cell death, measured by thenumber of pyknotic cells, was increased by the two highestnicotine doses (3). Similar results were obtained followingimposed administration of nicotine (1 mg · kg 1 ·day 1 for 3 days concurrent with 50 mg/kg BrdU) <strong>to</strong>adolescent rats (230). Although the mechanisms involvedin the effects of nicotine have not yet been investigated,nicotine could act directly on nicotinic acetylcholine recep<strong>to</strong>rsthat are present on immortalized hippocampalprogeni<strong>to</strong>r cells (44), or indirectly through an upregulationof the HPA axis (67) <strong>and</strong>/or a downregulation of thesero<strong>to</strong>ninergic system (43).III) Alcohol. The possibility that alcohol brings aboutdamage in the adult brain by disrupting neurogenesis hasbeen examined. Thus both acute (gavage with 5 g/kgethanol) <strong>and</strong> chronic (ethanol infusion 3 times/day over 4days for a mean dose of 9.3g·kg 1 · day 1 ) binge alcoholtreatments halve cell proliferation in the SGZ of the DGwithin 5 h (409). Furthermore, after 28 days, chronicbinge treatment decreases the survival rate of the newlyborn cells (409). In contrast, in animals fed for 6 wk witha moderate dose of ethanol (6.4% vol/vol), the number ofnewly born cells (labeled by 7 BrdU injections at 2-hintervals) is not reduced when animals are killed 1 h afterthe last BrdU injection (218). In fact, the number ofnascent cells decreases only after 2 wk suggesting that, inthis experimental model, cell survival, rather than cellproliferation, is affected by ethanol exposure, a hypothesisthat was confirmed by showing that cell death wasdramatically increased in the DG. Finally, these effectsare specific <strong>to</strong> the DG as bulbar neurogenesis is notinfluenced by chronic alcoholism. Similar results wereobtained with imposed injections of alcohol <strong>to</strong> adolescentrats (1 mg · kg 1 · day 1 for 3 days concurrent with 50mg·kg 1 · day 1 BrdU), which led <strong>to</strong> a downregulation ofhippocampal cell proliferation <strong>and</strong> increased cell death(230). Furthermore, coadministration of alcohol with nicotinereduces cell proliferation <strong>and</strong> cell death more potentlythan treatment with either one of the agents (230).Although these effects of ethanol could be mediated byNMDA or GABA A recep<strong>to</strong>rs (224), oxidative stress mightalso play a role since the antioxidant ebselen, used in thetreatment of cognitive disorders of alcoholic patients,prevents ethanol effects on hippocampal neurogenesis(218).Downloaded from physrev.physiology.org on November 25, 2005Physiol Rev • VOL 85 • APRIL 2005 • www.prv.org


NEUROGENESIS IN THE ADULT BRAIN 553IV) Cannabinoid. Derivatives of cannabis sativasuch as marijuana are acknowledged <strong>to</strong> be the most commonlyused illicit drugs (336). The structure of their activecomponents, the cannabinoids, the identification in thebrain of the cannabinoid-1 (CB1) recep<strong>to</strong>rs, <strong>and</strong> the isolationof an endogenous substance acting on these recep<strong>to</strong>rs,the endocannabinoid an<strong>and</strong>amide, have been reportedonly recently. It has been suggested that the CB1recep<strong>to</strong>rs that are expressed in the HF by the GABAergicinterneurons are important in learning <strong>and</strong> memory (109).Chronic treatment with an<strong>and</strong>amide (5 mg · kg 1 · day 1for 4 days) inhibits neurogenesis in the DG by decreasingthe number of newly born cells that differentiate in<strong>to</strong>granule neurons 12 days after the cessation of the treatment(100 mg/kg BrdU on day 2). Conversely, blockingthe endogenous cannabinoid <strong>to</strong>ne with SR14716 (1 mg ·kg 1 · day 1 <strong>to</strong>gether with an<strong>and</strong>amide), an antagonist ofthe CB1 recep<strong>to</strong>rs, increases hippocampal neurogenesisby favoring neuronal differentiation (476). These effectsare in accordance with a potentiality <strong>to</strong> modulate neuralcell fate (197) in particular through the ERK signalingpathway (476). In addition <strong>to</strong> showing that endocannabinoidsconstitute a physiological system regulating neurogenesis,these data raise the possibility that cannabinoidaddiction could be associated with an alteration in hippocampalneurogenesis.V) <strong>Neurogenesis</strong> as a substrate for vulnerability <strong>to</strong>addiction. The high responder rats (see sect. IIIB2) <strong>and</strong>the prenatally stressed rats (see sect. IIIB3) characterizedby curtailed neurogenesis share some similarities that arerelevant <strong>to</strong> drug addiction. Indeed, the HRs spontaneouslydevelop amphetamine (443, 445) or nicotine (528) selfadministrationbehavior, <strong>and</strong> prenatally stressed rats exhibitgreater sensitivity <strong>to</strong> the psychomo<strong>to</strong>r-stimulant effectsof nicotine (279) <strong>and</strong> amphetamine (215) <strong>and</strong> aremore vulnerable <strong>to</strong> drug addictive effects as they developintravenous amphetamine self-administration (116). Thussubjects starting off with the lowest neurogenesis may bemost vulnerable <strong>to</strong> the development of addictive behavior.It is noteworthy that there is high comorbidity betweendrug abuse <strong>and</strong> many psychiatric illness, amongwhich depression <strong>and</strong> schizophrenia (34, 152). Becausehippocampal neurogenesis is reduced in preclinical modelsof these pathologies, it may be hypothesized thatimpaired hippocampal neurogenesis may constitute a potentialsubstrate for vulnerability <strong>to</strong> drug abuse, depression,<strong>and</strong> schizophrenia (see also Fig. 8).VI) Conclusions. The downregulation of hippocampalneurogenesis, <strong>to</strong>gether with other maladaptive plasticityprocesses, i.e., dendritic remodeling <strong>and</strong> synapticplasticity modifications, are associated with permanentfunctional alterations in behavioral inhibition <strong>and</strong> learning,hallmarks of addiction. An alteration in hippocampalplasticity could be responsible for the cognitive deficitsthat appear in the course of the “illness” (88, 109, 133, 194)or during drug withdrawal (211). Because drug-seeking<strong>and</strong> -taking behaviors are sensitive <strong>to</strong> the presence ofdrug-associated stimuli (contextual cues), initially lowhippocampal plasticity <strong>and</strong> a drug-induced decrease inhippocampal plasticity may be involved in the vulnerability<strong>and</strong> the maintenance of drug abuse. Changes in hippocampalactivity could lead <strong>to</strong> a dysregulation of thedopaminergic mesoaccumbens reward system (283, 402)through the hippocampal glutamatergic output that gatesinformation in the nucleus accumbens (173) <strong>and</strong>/orthrough the hippocampal glutamatergic output <strong>to</strong> themesencephalic dopaminergic cell bodies. Indeed, stimulationof the ventral subiculum increases the firing ofdopaminergic neurons (51) <strong>and</strong> enhances dopamine releasewithin the nucleus accumbens (302). Finally, drugtakingin subjects vulnerable <strong>to</strong> drug addiction most probablyfurther increases corticosterone secretion, stimulatesthe dopaminergic reward system, <strong>and</strong> decreaseshippocampal neurogenesis, thereby constituting a pathophysiologicalloop.IV. CONCLUSIONThe discovery of neoneurogenesis in discrete areasof the adult brain has opened up an extremely interestingfield of research in itself. Indeed, although it was initiallyconfronted with the universal belief that after a certainperiod of development neither neural cell genesis nordivisions were any longer possible, it now constitutes apro<strong>to</strong>typic model of developmental neuroscience. Thestudy of the mechanisms that preside over cell proliferation,migration, differentiation, <strong>and</strong> survival is essentiallyperformed in the SVZ-OB paradigm as these phenomenaoccur in distinct compartments (cell proliferation in theSVZ, migration in the RMS, differentiation in the OB) thusrendering their analysis easier. However, the evaluationof their behavioral consequences is most frequently examinedin relation <strong>to</strong> the HF (with the exception of Ref.464).To conclude, the main issues raised by neurogenesisare as follows.1) <strong>From</strong> a cellular point of view, a true characterization,<strong>and</strong> thus definition of stemlike <strong>and</strong> progeni<strong>to</strong>r cellsin the adult brain is still lacking. Although immenseprogress has been achieved in the description of differentcell types (<strong>and</strong> their properties) in the SVZ, no consensushas been reached. Furthermore, although it has beensuggested that adult neural stemlike cells could behavesimilarly <strong>to</strong> embryonic stem cells as far as the generationof neurons <strong>and</strong> differentiation in<strong>to</strong> any neuronal type areconcerned, little is known regarding their intrinsic properties<strong>and</strong> the nature of the signals that lead <strong>to</strong> the generationof neurons in the adult brain. It is expected thatfunctional genomic technologies may help in defining theDownloaded from physrev.physiology.org on November 25, 2005Physiol Rev • VOL 85 • APRIL 2005 • www.prv.org


554 ABROUS, KOEHL, AND LE MOALtranscriptional program of neural progeni<strong>to</strong>rs <strong>and</strong> thechanges in gene expression in progeni<strong>to</strong>rs over time (105,323).2) Future studies will have <strong>to</strong> better characterize theearly development of adult-born neurons <strong>to</strong> dissect thesteps that are influenced by experience <strong>and</strong> vulnerable <strong>to</strong>traumatic conditions (stress, drug, epilepsy. . .). Thisraises the problem of tracking the fate of adult-born cellsusing BrdU, [ 3 H]dT, or a retrovirus, which may alter thefunctioning of the adult-born neurons under investigation.3) Undoubtedly one of the most interesting sets ofdata accumulated concerns the fac<strong>to</strong>rs that powerfullyregulate neurogenesis. However, the mechanisms bywhich these regulations are exerted in the normal <strong>and</strong>pathological brain remain obscure.4) Although most work is focused on the generationof glutamatergic excita<strong>to</strong>ry granule neurons in the adultDG, the recent discovery that GABAergic inhibi<strong>to</strong>ry neuronsare also produced raises the important possibilitythat external fac<strong>to</strong>rs (from molecules <strong>to</strong> modifications inanimals’ environments) might diversely affect the genesisof either type of neurons <strong>and</strong> thus have distinct functionalconsequences.5) Functional studies are <strong>to</strong>o often restricted <strong>to</strong> theevaluation of a single parameter; they should be extended<strong>to</strong> the different components of neurogenesis (basal neurogenesis<strong>and</strong> experimentally induced cell proliferation,cell death, <strong>and</strong> cell survival) <strong>and</strong> <strong>to</strong> other parametersrelated <strong>to</strong> the ana<strong>to</strong>mical (e.g., the presence of synapticcontacts) <strong>and</strong> functional (electrophysiological properties,ERK phosphorylation, immediate early gene activation. . .) integration of new neurons in<strong>to</strong> the preexistingnetworks. Finally, the contribution of newly born neurons<strong>to</strong> the functioning of the HF <strong>and</strong> its output should beevaluated as well as their influence on the activity of otherrelated structures (e.g., the frontal cortex).6) Functional evaluation of hippocampal neurogenesishas, in fact, been the object of relatively few studiesthat have been restricted <strong>to</strong> a limited number of behavioraltasks (mainly the Morris water maze). A more completediversified behavioral analysis is therefore necessary<strong>to</strong> underst<strong>and</strong> the contribution of neurogenesis <strong>to</strong>hippocampal functioning.7) Except for the elegant work of Shors et al. (509),which relies on the use of MAM which is unfortunately ahighly <strong>to</strong>xic compound, most studies correlate changes inrates of neuron addition with behavioral changes, in particularcognitive functions. Thus it is not clear whetherthese changes are coincidental or coregulated. A moredirect approach <strong>to</strong> address the existence of a causal relationshipbetween behavioral changes <strong>and</strong> neurogenesiswill probably depend on the development of inducibleDownloaded from physrev.physiology.org on November 25, 2005FIG. 7. Variation of hippocampal functioning as afunction of hippocampal neurogenesis <strong>and</strong> associatedpathological states.Physiol Rev • VOL 85 • APRIL 2005 • www.prv.org


NEUROGENESIS IN THE ADULT BRAIN 555transgenic models allowing a structure-specific blockadeof neurogenesis at a given time during behavioral testing.However, this approach is a real challenge since it isbased on the invalidation of the crucial genes controllingneurogenesis site specifically, which have not yet beenidentified.8) <strong>From</strong> a phylogenetic point of view, the apparentreduction in neuropoiesis, when moving from rodents <strong>to</strong>primates, raises the question of the associated evolutionaryadvantage.9) <strong>From</strong> studies that have correlated changes in ratesof neuron addition with hippocampal functioning, an invertedU curve shape can be proposed (see Fig. 7). Thusadult neurogenesis may be beneficial for adult brain functioningonly within a physiological adaptive range. Beyonda critical level of neurogenesis, which is over passedby stressful life events, aging, <strong>and</strong> drug abuse, dysfunctionappears. On the other h<strong>and</strong>, the addition of adult-bornneurons above a critical set point (i.e., following epileptiformstatus) may lead <strong>to</strong> pathological states <strong>and</strong> behavioralimpairments most probably by introducing noise inthe networks, as a consequence of ec<strong>to</strong>pic connectivity.10) Within this framework, it is not known whetheran alteration in neurogenesis is the cause or the result ofpsychiatric illness (Fig. 8). Indeed, in preclinical studies ithas been shown that phenotypes vulnerable <strong>to</strong> the developmen<strong>to</strong>f affective or cognitive disorders are characterizedby lower hippocampal neurogenesis. It may be hypothesizedthat these subjects who start off with impairedneurogenesis may be predisposed <strong>to</strong> the development ofaffective disorders, addictive behavior, <strong>and</strong> age-relatedcognitive disorders. Alternatively, the onset of pathologycould produce changes in brain plasticity by actingthrough several mechanisms (for example, stress <strong>and</strong>HPA activity are important media<strong>to</strong>rs in the genesis ofcognitive impairment, depression, addiction). Repeated“traumatic” episodes, <strong>to</strong>gether with a genetic predisposition,may lead <strong>to</strong> reduced neurogenesis <strong>and</strong> participate inthe worsening of the symp<strong>to</strong>ms. Although these propositionsare highly speculative, they highlight the fact thattreatments that increase neurogenesis may help <strong>to</strong> cure orprevent the development of affective <strong>and</strong>/or cognitivedisorders.Address for reprint requests <strong>and</strong> other correspondence: D. N.Abrous, INSERM U588, Univ. of Bordeaux 2, Institut FrançoisMagendie, 146 rue Léo Saignat, 33077 Bordeaux Cedex,France (E-mail: abrous@bordeaux.inserm.fr).REFERENCESFIG. 8. Is impaired neurogenesis acause or a consequence of affective <strong>and</strong>cognitive disorders? Based on the reciprocalinfluences of neurogenesis on affective<strong>and</strong> cognitive states, two modelscan be proposed. In the first model, impairmentin neurogenesis processes resultingfrom both genetic <strong>and</strong> environmentalinfluences would predispose theindividual (vulnerable phenotype) <strong>to</strong> developingaffective <strong>and</strong> cognitive disordersin response <strong>to</strong> environmental challenges.In the second model, genetic <strong>and</strong>environmental influences may orientbrain functions <strong>to</strong>ward deleterious developmentsresulting in affective <strong>and</strong> cognitivedisorders that would, in turn, impairthe normal course of neurogenesis.1. Aberg MA, Aberg ND, Hedbacker H, Oscarsson J, <strong>and</strong> ErikssonPS. Peripheral infusion of IGF-I selectively induces neurogenesisin the adult rat hippocampus. J Neurosci 20: 2896–2903,2000.2. Aberg MA, Aberg ND, Palmer TD, Alborn AM, Carlsson-Skwirut C, Bang P, Rosengren LE, Olsson T, Gage FH, <strong>and</strong>Eriksson PS. IGF-I has a direct proliferative effect in adult hippocampalprogeni<strong>to</strong>r cells. Mol Cell Neurosci 24: 23–40, 2003.3. Abrous DN, Adriani W, Montaron MF, Aurousseau C, RougonG, Le Moal M, <strong>and</strong> Piazza PV. Nicotine self-administration impairshippocampal plasticity. J Neurosci 22: 3656–3662, 2002.4. Agartz I, Momenan R, Rawlings RR, Kerich MJ, <strong>and</strong> HommerDW. Hippocampal volume in patients with alcohol dependence.Arch Gen Psychiatry 56: 356–363, 1999.Downloaded from physrev.physiology.org on November 25, 2005Physiol Rev • VOL 85 • APRIL 2005 • www.prv.org


556 ABROUS, KOEHL, AND LE MOAL5. Agnati LF, Zoli M, Biagini G, <strong>and</strong> Fuxe K. Neuronal plasticity<strong>and</strong> ageing processes in the frame of the “Red Queen Theory.” ActaPhysiol Sc<strong>and</strong> 145: 301–309, 1992.6. Ahmed S, Reynolds BA, <strong>and</strong> Weiss S. BDNF enhances the differentiationbut not the survival of CNS stem cell-derived neuronalprecursors. J Neurosci 15: 5765–5778, 1995.7. Akamatsu W, Okano HJ, Osumi N, Inoue T, Nakamura S,Sakakibara Si Miura M, Matsuo N, Darnell RB, <strong>and</strong> Okano H.Mammalian ELAV-like neuronal RNA-binding proteins HuB <strong>and</strong>HuC promote neuronal development in both the central <strong>and</strong> theperipheral nervous systems. Proc Natl Acad Sci USA 96: 9885–9890,1999.8. Alder<strong>to</strong>n WK, Cooper CE, <strong>and</strong> Knowles RG. Nitric oxide synthases:structure, function <strong>and</strong> inhibition. Biochem J 357: 593–615,2001.9. Alonso G, Prie<strong>to</strong> M, <strong>and</strong> Chauvet N. Tangential migration ofyoung neurons arising from the subventricular zone of adult rats isimpaired by surgical lesions passing through their natural migra<strong>to</strong>rypathway. J Comp Neurol 405: 508–528, 1999.10. Alonso R, Griebel G, Pavone G, Stemmelin J, Le Fur G, <strong>and</strong>Soubrie P. Blockade of CRF1 or V1B recep<strong>to</strong>rs reverses stressinducedsuppression of neurogenesis in a mouse model of depression.Mol Psychiatry 9: 224, 2004.11. Altman J. Are new neurones formed in the brains of adult mammals?Science 135: 1127–1128, 1962.12. Altman J. Au<strong>to</strong>radiographic investigation of cell proliferation inthe brains of rats <strong>and</strong> cats. Anat Rec 145: 573–592, 1963.13. Altman J <strong>and</strong> Das GD. Au<strong>to</strong>radiographic <strong>and</strong> his<strong>to</strong>logical evidenceof postnatal hippocampal neurogenesis in rats. J CompNeurol 124: 319–336, 1965.14. Altman J <strong>and</strong> Das GD. Au<strong>to</strong>radiographic <strong>and</strong> his<strong>to</strong>logical studiesof postnatal neurogenesis. I. A longitudinal investigation of thekinetics, migration <strong>and</strong> transformation of cells incorporating tritiatedthymidine in neonate rats, with special reference <strong>to</strong> postnatalneurogenesis in some brain regions. J Comp Neurol 126: 337–389,1966.15. Alvarez-Buylla A, Seri B, <strong>and</strong> Doetsch F. Identification of neuralstem cells in the adult vertebrate brain. Brain Res Bull 57: 751–758,2002.16. Amaral DG <strong>and</strong> Witter MP. The three-dimensional organizationof the hippocampal formation: a review of ana<strong>to</strong>mical data. Neuroscience31: 571–591, 1989.17. Ambrogini P, Orsini L, Mancini C, Ferri P, Barbanti I, <strong>and</strong>Cuppini R. Persistently high corticosterone levels but not normalcircadian fluctuations of the hormone affect cell proliferation in theadult rat dentate gyrus. Neuroendocrinology 76: 366–372, 2002.18. Ambrogini P, Orsini L, Mancini C, Ferri P, Ciaroni S, <strong>and</strong>Cuppini R. Learning may reduce neurogenesis in adult rat dentategyrus. Neurosci Lett 359: 13–16, 2004.19. Anagnostaras SG, Maren S, <strong>and</strong> Fanselow MS. Temporallygraded retrograde amnesia of contextual fear after hippocampaldamage in rats: within-subjects examination. J Neurosci 19: 1106–1114, 1999.20. Anderson MF, Aberg MAI, Nilsson M, <strong>and</strong> Eriksson PS. Insulin-likegrowth fac<strong>to</strong>r-I <strong>and</strong> neurogenesis in the adult mammalianbrain. Dev Brain Res 134: 115–122, 2002.21. An<strong>to</strong>nopoulos J, Pappas IS, <strong>and</strong> Parnavelas JG. Activation ofthe GABA A recep<strong>to</strong>r inhibits the proliferative effects of bFGF incortical progeni<strong>to</strong>r cells. Eur J Neurosci 9: 291–298, 1997.22. Arsenijevic Y <strong>and</strong> Weiss S. Insulin-like growth fac<strong>to</strong>r-I is a differentiationfac<strong>to</strong>r for postmi<strong>to</strong>tic CNS stem cell-derived neuronalprecursors: distinct actions from those of brain-derived neurotrophicfac<strong>to</strong>r. J Neurosci 18: 2118–2128, 1998.23. Artavanis-Tsakonas S, R<strong>and</strong> MD, <strong>and</strong> Lake RJ. Notch signaling:cell fate control <strong>and</strong> signal integration in development. Science 284:770–776, 1999.24. Arvidsson A, Collin T, Kirik D, Kokaia Z, <strong>and</strong> Lindvall O.Neuronal replacement from endogenous precursors in the adultbrain after stroke. Nat Med 8: 963–970, 2002.25. Arvidsson A, Kokaia Z, <strong>and</strong> Lindvall O. N-methyl-D-aspartaterecep<strong>to</strong>r-mediated increase of neurogenesis in adult rat dentategyrus following stroke. Eur J Neurosci 14: 10–18, 2001.26. Azmitia EC. Modern views on an ancient chemical: sero<strong>to</strong>nineffects on cell proliferation, maturation, <strong>and</strong> apop<strong>to</strong>sis. Brain ResBull 56: 413–424, 2001.27. Bai F, Bergeron M, <strong>and</strong> Nelson DL. Chronic AMPA recep<strong>to</strong>rpotentia<strong>to</strong>r (LY451646) treatment increases cell proliferation inadult rat hippocampus. Neuropharmacology 44: 1013–1021, 2003.28. Bailey CH <strong>and</strong> K<strong>and</strong>el ER. Structural changes accompanyingmemory s<strong>to</strong>rage. Annu Rev Physiol 55: 397–426, 1993.29. Banasr M, Hery M, Brezun JM, <strong>and</strong> Daszuta A. Sero<strong>to</strong>ninmediates oestrogen stimulation of cell proliferation in the adultdentate gyrus. Eur J Neurosci 14: 1417–1424, 2001.30. Banasr M, Hery M, Printemps R, <strong>and</strong> Daszuta A. Sero<strong>to</strong>nininducedincreases in adult cell proliferation <strong>and</strong> neurogenesis aremediated through different <strong>and</strong> common 5-HT recep<strong>to</strong>r subtypes inthe dentate gyrus <strong>and</strong> the subventricular zone. Neuropsychopharmacology29: 450–460, 2004.31. Barami K, Iversen K, Furneaux H, <strong>and</strong> Goldman SA. Hu proteinas an early marker of neuronal phenotypic differentiation bysubependymal zone cells of the adult songbird forebrain. J Neurobiol28: 82–101, 1995.32. Baron M. An overview of the Notch signalling pathway. Semin CellDev Biol 14: 113–119, 2003.33. Bartzokis G, Beckson M, Lu PH, Edwards N, Rapoport R,Wiseman E, <strong>and</strong> Bridge P. Age-related brain volume reductions inamphetamine <strong>and</strong> cocaine addicts <strong>and</strong> normal controls: implicationsfor addiction research. Psychiatry Res 98: 93–102, 2000.34. Batel P. Addiction <strong>and</strong> schizophrenia. Eur Psychiatry 15: 115–122,2000.35. Bauer S, Moyse E, Jourdan F, Colpaert F, Martel JC, <strong>and</strong>Marien M. Effects of the alpha(2)-adrenorecep<strong>to</strong>r antagonist dexefaroxanon neurogenesis in the olfac<strong>to</strong>ry bulb of the adult rat invivo: selective protection against neuronal death. Neuroscience117: 281–291, 2003.36. Baulieu EE <strong>and</strong> Robel P. Neurosteroids: a new brain function. JSteroid Biochem Mol Biol 37: 395–403, 1997.37. Bayer SA, Brunner RL, Hine R, <strong>and</strong> Altman J. Behaviouraleffects of interference with the postnatal acquisition of hippocampalgranule cells. Nature New Biol 242: 222–224, 1973.38. Belluzzi O, Benedusi M, Ackman J, <strong>and</strong> LoTurco JJ. Electrophysiologicaldifferentiation of new neurons in the olfac<strong>to</strong>ry bulb.J Neurosci 23: 10411–10418, 2003.39. Belvindrah R, Rougon G, <strong>and</strong> Chazal G. Increased neurogenesisin adult mCD24-deficient mice. J Neurosci 22: 3594–3607, 2002.40. Bendotti C, Pende M, Guglielmetti F, <strong>and</strong> Samanin R. Cycloheximideinhibits kainic acid-induced GAP-43 mRNA in dentategranule cells in rats. Neuroreport 7: 2539–2542, 1996.41. Benes FM, Kwok EW, Vincent SL, <strong>and</strong> Todtenkopf MS. Areduction of nonpyramidal cells in sec<strong>to</strong>r CA2 of schizophrenics<strong>and</strong> manic depressives. Biol Psychiatry 44: 88–97, 1998.42. Bengzon J, Kokaia Z, Elmer E, Nanobashvili A, Kokaia M, <strong>and</strong>Lindvall O. Apop<strong>to</strong>sis <strong>and</strong> proliferation of dentate gyrus neuronsafter single <strong>and</strong> intermittent limbic seizures. Proc Natl Acad SciUSA 94: 10432–10437, 1997.43. Benwell ME, Balfour DJ, <strong>and</strong> Anderson JM. Smoking-associatedchanges in the sero<strong>to</strong>nergic systems of discrete regions ofhuman brain. Psychopharmacology 102: 68–72, 1990.44. Berger F, Gage FH, <strong>and</strong> Vijayaraghavan S. Nicotinic recep<strong>to</strong>rinducedapop<strong>to</strong>tic cell death of hippocampal progeni<strong>to</strong>r cells.J Neurosci 18: 6871–6881, 1998.45. Berke JD <strong>and</strong> Hyman SE. Addiction, dopamine, <strong>and</strong> the molecularmechanisms of memory. Neuron 25: 515–532, 2000.46. Bernabeu R <strong>and</strong> Sharp FR. NMDA <strong>and</strong> AMPA/kainate glutamaterecep<strong>to</strong>rs modulate dentate neurogenesis <strong>and</strong> CA3 synapsin-I innormal <strong>and</strong> ischemic hippocampus. J Cereb Blood Flow Metab 20:1669–1680, 2000.47. Bernier PJ, Vinet J, Cossette M, <strong>and</strong> Parent A. Characterizationof the subventricular zone of the adult human brain: evidencefor the involvement of Bcl-2. Neurosci Res 37: 67–78, 2000.48. Berridge MJ. Calcium signalling <strong>and</strong> cell proliferation. Bioessays17: 491–500, 1995.49. Bertr<strong>and</strong> N, Castro DS, <strong>and</strong> Guillemot F. Proneural genes <strong>and</strong>the specification of neural cell types. Nat Rev Neurosci 3: 517–530,2002.Downloaded from physrev.physiology.org on November 25, 2005Physiol Rev • VOL 85 • APRIL 2005 • www.prv.org


NEUROGENESIS IN THE ADULT BRAIN 55750. Biebl M, Cooper CM, Winkler J, <strong>and</strong> Kuhn HG. Analysis ofneurogenesis <strong>and</strong> programmed cell death reveals a self-renewingcapacity in the adult rat brain. Neurosci Lett 291: 17–20, 2000.51. Blaha CD, Yang CR, Floresco SB, Barr AM, <strong>and</strong> Phillips AG.Stimulation of the ventral subiculum of the hippocampus evokesglutamate recep<strong>to</strong>r-mediated changes in dopamine efflux in the ratnucleus accumbens. Eur J Neurosci 9: 902–911, 1997.52. Blanchard RJ <strong>and</strong> Blanchard DC. Effects of hippocampal lesionson the rat’s reaction <strong>to</strong> a cat. J Comp Physiol Psychol 78: 77–82,1972.53. Bogerts B, Meertz E, <strong>and</strong> Schonfeldt-Bausch R. Basal ganglia<strong>and</strong> limbic system pathology in schizophrenia. A morphometricstudy of brain volume <strong>and</strong> shrinkage. Arch Gen Psychiatry 42:784–791, 1985.54. Bondolfi L, Ermini F, Long JM, Ingram DK, <strong>and</strong> Jucker M.Impact of age <strong>and</strong> caloric restriction on neurogenesis in the dentategyrus of C57BL/6 mice. Neurobiol Aging 25: 333–340, 2004.55. Bonfanti L <strong>and</strong> Theodosis DT. Expression of polysialylated neuralcell adhesion molecule by proliferating cells in the subependymallayer of the adult rat, in its rostral extension <strong>and</strong> in theolfac<strong>to</strong>ry bulb. Neuroscience 62: 291–305, 1994.56. Bontempi B, Laurent-Demir C, Destrade C, <strong>and</strong> Jaffard R.Time-dependent reorganization of brain circuitry underlying longtermmemory s<strong>to</strong>rage. Nature 400: 671–675, 1999.57. Bouchet C <strong>and</strong> Cazauvieilh G. Epilepsie et l’aliénation mentale.Arch Gen Med 9: 510–542, 1825.58. Br<strong>and</strong>t MD, Jessberger S, Steiner B, Kronenberg G, Reuter K,Bick-S<strong>and</strong>er A, von der BW, <strong>and</strong> Kempermann G. Transientcalretinin expression defines early postmi<strong>to</strong>tic step of neuronaldifferentiation in adult hippocampal neurogenesis of mice. Mol CellNeurosci 24: 603–613, 2003.59. Breitner JC, Gau BA, Welsh KA, Plassman BL, McDonald WM,Helms MJ, <strong>and</strong> Anthony JC. Inverse association of anti-inflamma<strong>to</strong>rytreatments <strong>and</strong> Alzheimer’s disease: initial results of aco-twin control study. Neurology 44: 227–232, 1994.60. Bremner JD, R<strong>and</strong>all P, Scott TM, Bronen RA, Seibyl JP,Southwick SM, Delaney RC, McCarthy G, Charney DS, <strong>and</strong>Innis RB. MRI-based measurement of hippocampal volume inpatients with combat-related posttraumatic stress disorder. Am JPsychiatry 152: 973–981, 1995.61. Brezun JM <strong>and</strong> Daszuta A. Depletion in sero<strong>to</strong>nin decreasesneurogenesis in the dentate gyrus <strong>and</strong> the subventricular zone ofadult rats. Neuroscience 89: 999–1002, 1999.62. Brezun JM <strong>and</strong> Daszuta A. Sero<strong>to</strong>nergic reinnervation reverseslesion-induced decreases in PSA-NCAM labeling <strong>and</strong> proliferationof hippocampal cells in adult rats. Hippocampus 10: 37–46, 2000.63. Brezun JM <strong>and</strong> Daszuta A. Sero<strong>to</strong>nin may stimulate granule cellproliferation in the adult hippocampus, as observed in rats graftedwith foetal raphe neurons. Eur J Neurosci 12: 391–396, 2000.64. Brizze KR <strong>and</strong> Ordy JM. Age pigments, cell loss <strong>and</strong> hippocampalfunction. Mech Ageing Dev 9: 143–162, 1979.65. Brown JP, Couillard-Despres S, Cooper-Kuhn CM, Winkler J,Aigner L, <strong>and</strong> Kuhn HG. Transient expression of doublecortinduring adult neurogenesis. J Comp Neurol 467: 1–10, 2003.66. Brunner RL. A cross-sectional study of behavior at three agesafter neonatal X irradiation of the hippocampus. Behav Biol 22:211–218, 1978.67. Caggiula AR, Donny EC, Epstein LH, Sved AF, Knopf S, RoseC, McAllister CG, Antelman SM, <strong>and</strong> Perkins KA. The role ofcorticosteroids in nicotine’s physiological <strong>and</strong> behavioral effects.Psychoneuroendocrinology 23: 143–159, 1998.68. Cahill L <strong>and</strong> McGaugh JL. Mechanisms of emotional arousal <strong>and</strong>lasting declarative memory. Trends Neurosci 21: 294–299, 1998.69. Callaghan DA, Dong L, Callaghan SM, Hou YX, Dagnino L, <strong>and</strong>Slack RS. Neural precursor cells differentiating in the absence ofRb exhibit delayed terminal mi<strong>to</strong>sis <strong>and</strong> deregulated E2F 1 <strong>and</strong> 3activity. Dev Biol 207: 257–270, 1999.70. Calza L, Giardino L, Pozza M, Bettelli C, Micera A, <strong>and</strong> AloeL. Proliferation <strong>and</strong> phenotype regulation in the subventricularzone during experimental allergic encephalomyelitis: in vivo evidenceof a role for nerve growth fac<strong>to</strong>r. Proc Natl Acad Sci USA 95:3209–3214, 1998.71. Cameron HA <strong>and</strong> Gould E. <strong>Adult</strong> neurogenesis is regulated byadrenal steroids in the dentate gyrus. Neuroscience 61: 203–209,1994.72. Cameron HA <strong>and</strong> Gould E. Distinct populations of cells in theadult dentate gyrus undergo mi<strong>to</strong>sis or apop<strong>to</strong>sis in response <strong>to</strong>adrenalec<strong>to</strong>my. J Comp Neurol 369: 56–63, 1996.73. Cameron HA, McEwen BS, <strong>and</strong> Gould E. Regulation of adultneurogenesis by excita<strong>to</strong>ry input <strong>and</strong> NMDA recep<strong>to</strong>r activation inthe dentate gyrus. J Neurosci 15: 4687–4692, 1995.74. Cameron HA <strong>and</strong> McKay RD. Res<strong>to</strong>ring production of hippocampalneurons in old age. Nat Neurosci 2: 894–897, 1999.75. Cameron HA <strong>and</strong> McKay RD. <strong>Adult</strong> neurogenesis produces alarge pool of new granule cells in the dentate gyrus. J Comp Neurol435: 406–417, 2001.76. Cameron HA, Tanapat P, <strong>and</strong> Gould E. Adrenal steroids <strong>and</strong>N-methyl-D-aspartate recep<strong>to</strong>r activation regulate neurogenesis inthe dentate gyrus of adult rats through a common pathway. Neuroscience82: 349–354, 1997.77. Cameron HA, Woolley CS, <strong>and</strong> Gould E. Adrenal steroid recep<strong>to</strong>rimmunoreactivity in cells born in the adult rat dentate gyrus.Brain Res 611: 342–346, 1993.78. Cameron HA, Woolley CS, McEwen BS, <strong>and</strong> Gould E. Differentiationof newly born neurons <strong>and</strong> glia in the dentate gyrus of theadult rat. Neuroscience 56: 337–344, 1993.79. Capela A <strong>and</strong> Temple S. LeX/ssea-1 is expressed by adult mouseCNS stem cells, identifying them as nonependymal. Neuron 35:865–875, 2002.80. Carlen M, Cassidy RM, Brismar H, Smith GA, Enquist LW, <strong>and</strong>Frisen J. Functional integration of adult-born neurons. Curr Biol12: 606–608, 2002.81. Carle<strong>to</strong>n A, Petreanu LT, Lansford R, Alvarez-Buylla A, <strong>and</strong>Lledo PM. Becoming a new neuron in the adult olfac<strong>to</strong>ry bulb. NatNeurosci 6: 507–518, 2003.82. Cavallaro S, D’Agata V, Manickam P, Dufour F, <strong>and</strong> Alkon DL.Memory-specific temporal profiles of gene expression in the hippocampus.Proc Natl Acad Sci USA 99: 16279–16284, 2002.83. Cecchini T, Ciaroni S, Ferri P, Ambrogini P, Cuppini R, SantiS, <strong>and</strong> Del Gr<strong>and</strong>e P. Alpha-<strong>to</strong>copherol, an exogenous fac<strong>to</strong>r ofadult hippocampal neurogenesis regulation. J Neurosci Res 73:447–455, 2003.84. Chen G, Rajkowska G, Du F, Seraji-Bozorgzad N, <strong>and</strong> ManjiHK. Enhancement of hippocampal neurogenesis by lithium. J Neurochem75: 1729–1734, 2000.85. Chiasson BJ, Tropepe V, Morshead CM, <strong>and</strong> van der Kooy D.<strong>Adult</strong> mammalian forebrain ependymal <strong>and</strong> subependymal cellsdemonstrate proliferative potential, but only subependymal cellshave neural stem cell characteristics. J Neurosci 19: 4462–4471,1999.86. Ciaroni S, Cecchini T, Ferri P, Cuppini R, Ambrogini P, SantiS, Benedetti S, Del Gr<strong>and</strong>e P, <strong>and</strong> Papa S. Neural precursorproliferation <strong>and</strong> newborn cell survival in the adult rat dentategyrus are affected by vitamin E deficiency. Neurosci Res 44: 369–377, 2002.87. Ciaroni S, Cuppini R, Cecchini T, Ferri P, Ambrogini P, CuppiniC, <strong>and</strong> Del Gr<strong>and</strong>e P. <strong>Neurogenesis</strong> in the adult rat dentategyrus is enhanced by vitamin E deficiency. J Comp Neurol 411:495–502, 1999.88. Ciopolli C <strong>and</strong> Galliani L. Addiction time <strong>and</strong> intellectual impairmentin heroin users. Psychol Rep 60: 1099–1105, 1987.89. Clarke AR, Ma<strong>and</strong>ag ER, van Roon M, van der Lugt NM, v<strong>and</strong>er Valk M, Hooper ML, Berns A, <strong>and</strong> te Riele H. Requirementfor a functional Rb-1 gene in murine development. Nature 359:328–330, 1992.90. Cohen P, Ocrant I, Fielder PJ, Neely EK, Gargosky SE, DealCI, Ceda GP, Youngman O, Pham H, Lamson G, Giudice LC,<strong>and</strong> Rosenfeld RG. Insulin-like growth fac<strong>to</strong>rs (IGFs): implicationsfor aging. Psychoneuroendocrinology 17: 335–342, 1992.91. Coleman P. How old is old? Neurobiol Aging 25: 1, 2004.92. Coleman P, Finch C, <strong>and</strong> Joseph J. The need for multiple timepoints in aging studies. Neurobiol Aging 25: 3–4, 2004.93. Conrad AJ, Abebe T, Austin R, Forsythe S, <strong>and</strong> Scheibel AB.Hippocampal pyramidal cell disarray in schizophrenia as a bilateralphenomenon. Arch Gen Psychiatry 48: 413–417, 1991.Downloaded from physrev.physiology.org on November 25, 2005Physiol Rev • VOL 85 • APRIL 2005 • www.prv.org


558 ABROUS, KOEHL, AND LE MOAL94. Cooper-Kuhn CM, Vroemen M, Brown J, Ye H, Thompson MA,Winkler J, <strong>and</strong> Kuhn HG. Impaired adult neurogenesis in micelacking the transcription fac<strong>to</strong>r E2F1. Mol Cell Neurosci 21: 312–323, 2002.95. Cooper-Kuhn CM <strong>and</strong> Georg Kuhn H. Is it all DNA repair?Methodological considerations for detecting neurogenesis in theadult brain. Dev Brain Res 134: 13–21, 2002.96. Corot<strong>to</strong> FS, Henegar JR, <strong>and</strong> Maruniak JA. Odor deprivationleads <strong>to</strong> reduced neurogenesis <strong>and</strong> reduced neuronal survival in theolfac<strong>to</strong>ry bulb of the adult mouse. Neuroscience 61: 739–744, 1994.97. Covolan L, Ribeiro LT, Longo BM, <strong>and</strong> Mello LE. Cell damage<strong>and</strong> neurogenesis in the dentate granule cell layer of adult rats afterpilocarpine- or kainate-induced status epilepticus. Hippocampus10: 169–180, 2000.98. Craig CG, Tropepe V, Morshead CM, Reynolds BA, Weiss S,<strong>and</strong> van der Kooy D. In vivo growth fac<strong>to</strong>r expansion of endogeneoussubependymal neural precursor cell populations in the adultmouse. J Neurosci 16: 2649–2658, 1996.99. Cr<strong>and</strong>all J, Sakai Y, Zhang J, Koul O, Mineur Y, Crusio WE,<strong>and</strong> McCaffery P. 13-Cis-retinoic acid suppresses hippocampalcell division <strong>and</strong> hippocampal-dependent learning in mice. ProcNatl Acad Sci USA 101: 5111–5116, 2004.100. Cremer H, Lange R, Chris<strong>to</strong>ph A, Plomann M, Vopper G, RoesJ, Brown R, Baldwin S, Kraemer P, Scheff S, Barthels D,Rajewsky K, <strong>and</strong> Wille W. Inactivation of the N-CAM gene in miceresults in size reduction of the olfac<strong>to</strong>ry bulb <strong>and</strong> deficits in spatiallearning. Nature 367: 455–459, 1994.101. Csernansky JG, Joshi S, Wang L, Haller JW, Gado M, MillerJP, Gren<strong>and</strong>er U, <strong>and</strong> Miller MI. Hippocampal morphometry inschizophrenia by high dimensional brain mapping. Proc Natl AcadSci USA 95: 11406–11411, 1998.102. Cuppini R, Ciaroni S, Cecchini T, Ambrogini P, Ferri P, CuppiniC, Ninfali P, <strong>and</strong> Del Gr<strong>and</strong>e P. Tocopherols enhanceneurogenesis in dentate gyrus of adult rats. Int J Vitam Nutr Res72: 170–176, 2002.103. Curtis MA, Penney EB, Pearson AG, Roon-Mom WM, ButterworthNJ, Dragunow M, Connor B, <strong>and</strong> Faull RL. Increased cellproliferation <strong>and</strong> neurogenesis in the adult human Hunting<strong>to</strong>n’sdisease brain. Proc Natl Acad Sci USA 100: 9023–9027, 2003.104. Czeh B, Michaelis T, Watanabe T, Frahm J, de Biurrun G, vanKampen M, Bar<strong>to</strong>lomucci A, <strong>and</strong> Fuchs E. Stress-inducedchanges in cerebral metabolites, hippocampal volume, <strong>and</strong> cellproliferation are prevented by antidepressant treatment withtianeptine. Proc Natl Acad Sci USA 98: 12796–12801, 2001.105. D’Amour KA <strong>and</strong> Gage FH. Genetic <strong>and</strong> functional differencesbetween multipotent neural <strong>and</strong> pluripotent embryonic stem cells.Proc Natl Acad Sci USA 100 Suppl: 11866–11872, 2003.106. Dash PK, Blum S, <strong>and</strong> Moore AN. Caspase activity plays anessential role in long-term memory. Neuroreport 11: 2811–2814,2000.107. Dash PK, Mach SA, <strong>and</strong> Moore AN. Enhanced neurogenesis inthe rodent hippocampus following traumatic brain injury. J NeurosciRes 63: 313–319, 2001.108. Dashtipour K, Tran PH, Okazaki MM, Nadler JV, <strong>and</strong> RibakCE. Ultrastructural features <strong>and</strong> synaptic connections of hilarec<strong>to</strong>pic granule cells in the rat dentate gyrus are different fromthose of granule cells in the granule cell layer. Brain Res 890:261–271, 2001.109. Davies SN, Pertwee RG, <strong>and</strong> Riedel G. Functions of cannabinoidrecep<strong>to</strong>rs in the hippocampus. Neuropharmacology 42: 993–1007,2002.110. Dawirs RR, Hildebr<strong>and</strong>t K, <strong>and</strong> Teuchert-Noodt G. <strong>Adult</strong> treatmentwith haloperidol increases dentate granule cell proliferationin the gerbil hippocampus. J Neural Transm 105: 317–327, 1998.111. Dayer AG, Ford AA, Cleaver KM, Yassaee M, <strong>and</strong> CameronHA. Short-term <strong>and</strong> long-term survival of new neurons in the ratdentate gyrus. J Comp Neurol 460: 563–572, 2003.112. De Bellis MD, Clark DB, Beers SR, Soloff PH, Boring AM, HallJ, Kersh A, <strong>and</strong> Keshavan MS. Hippocampal volume in adolescent-onsetalcohol use disorders. Am J Psychiatry 157: 737–744,2000.113. Decker L, Avellana-Adalid V, Nait-Oumesmar B, Durbec P,<strong>and</strong> Baron-Van Evercooren A. Oligodendrocyte precursor migration<strong>and</strong> differentiation: combined effects of PSA residues, growthfac<strong>to</strong>rs, <strong>and</strong> substrates. Mol Cell Neurosci 16: 422–439, 2000.114. Deisseroth K, Singla S, Toda H, Monje M, Palmer TD, <strong>and</strong>Malenka RC. Excitation-neurogenesis coupling in adult neuralstem/progeni<strong>to</strong>r cells. Neuron 42: 535–552, 2004.115. Dellu F, Mayo W, Vallee M, Maccari S, Piazza PV, Le Moal M,<strong>and</strong> Simon H. Behavioral reactivity <strong>to</strong> novelty during youth as apredictive fac<strong>to</strong>r of stress-induced corticosterone secretion in theelderly: a life-span study in rats. Psychoneuroendocrinology 21:441–453, 1996.116. Deminière JM, Piazza PV, Guegan G, Abrous N, Maccari S, LeMoal M, <strong>and</strong> Simon H. Increased locomo<strong>to</strong>r response <strong>to</strong> novelty<strong>and</strong> propensity <strong>to</strong> intravenous amphetamine self-administration inadult offspring of stressed mothers. Brain Res 586: 135–139, 1992.117. Dempsey RJ, Sailor KA, Bowen KK, Tureyen K, <strong>and</strong> VemugantiR. Stroke-induced progeni<strong>to</strong>r cell proliferation in adult spontaneouslyhypertensive rat brain: effect of exogenous IGF-I <strong>and</strong>GDNF. J Neurochem 87: 586–597, 2003.118. Derrick BE, York AD, <strong>and</strong> Martinez JL Jr. Increased granulecell neurogenesis in the adult dentate gyrus following mossy fiberstimulation sufficient <strong>to</strong> induce long-term potentiation. Brain Res857: 300–307, 2000.119. Diaz-Granados JL, Greene PL, <strong>and</strong> Amsel A. Memory-basedlearning in preweanling <strong>and</strong> adult rats after infantile x-irradiationinducedhippocampal granule cell hypoplasia. Behav Neurosci 106:940–946, 1992.120. Dickson DW. Neuropathology of Alzheimer’s disease <strong>and</strong> otherdementias. Clin Geriatr Med 17: 209–228, 2001.121. Döbrössy MDE, Aurousseau C, Le Moal M, Piazza PV <strong>and</strong>Abrous DN. Differential effects of learning on neurogenesis: learningincreases or decreases the number of newly born cells dependingon their birth date. Mol Psychiatry 8: 974–982, 2003.122. Doetsch F <strong>and</strong> Alvarez-Buylla A. <strong>Network</strong> of tangential pathwaysfor neuronal migration in adult mammalian brain. Proc NatlAcad Sci USA 93: 14895–14900, 1996.123. Doetsch F, Caille I, Lim DA, Garcia-Verdugo JM, <strong>and</strong> Alvarez-Buylla A. Subventricular zone astrocytes are neural stem cells inthe adult mammalian brain. Cell 97: 703–716, 1999.124. Doetsch F, Garcia-Verdugo JM, <strong>and</strong> Alvarez-Buylla A. Cellularcomposition <strong>and</strong> three-dimensional organization of the subventriculargerminal zone in the adult mammalian brain. J Neurosci 17:5046–5061, 1997.125. Doetsch F, Garcia-Verdugo JM, <strong>and</strong> Alvarez-Buylla A. Regenerationof a germinal layer in the adult mammalian brain. Proc NatlAcad Sci USA 96: 11619–11624, 1999.126. Doetsch F, Petreanu L, Caille I, Garcia-Verdugo JM, <strong>and</strong>Alvarez-Buylla A. EGF converts transit-amplifying neurogenicprecursors in the adult brain in<strong>to</strong> multipotent stem cells. Neuron36: 1021–1034, 2002.127. Doetsch F, Verdugo JMG, Caille I, Alvarez-Buylla A, ChaoMV, <strong>and</strong> Casaccia-Bonnefil P. Lack of the cell-cycle inhibi<strong>to</strong>rp27Kip1 results in selective increase of transit-amplifying cells foradult neurogenesis. J Neurosci 22: 2255–2264, 2002.128. Dong H, Csernansky CA, Goico B, <strong>and</strong> Csernansky JG. Hippocampalneurogenesis follows kainic acid-induced apop<strong>to</strong>sis inneonatal rats. J Neurosci 23: 1742–1749, 2003.129. Drapeau E, Döbrössy M, Aurousseau C, Le Moal M, Piazza PV,<strong>and</strong> Abrous DN. Learning in rats induces cell death <strong>and</strong> cellproliferation in the hippocampus. Proc Soc Neurosci New OrleansLA 2003.130. Drapeau E, Mayo W, Aurousseau C, Le Moal M, Piazza PV, <strong>and</strong>Abrous DN. Spatial memory performances of aged rats in thewater maze predict levels of hipppocampal neurogenesis. Proc NatlAcad Sci USA 100: 14385–14390, 2003.131. Duman RS, Nakagawa S, <strong>and</strong> Malberg J. Regulation of adultneurogenesis by antidepressant treatment. Neuropsychopharmacology25: 836–844, 2001.132. Dunaevsky A <strong>and</strong> Mason CA. Spine motility: a means <strong>to</strong>wards anend? Trends Neurosci 26: 155–160, 2003.133. Eckardt MJ <strong>and</strong> Martin PR. Clinical assessment of cognition inalcoholism. Alcohol Clin Exp Res 10: 123–127, 1986.134. Ehninger D <strong>and</strong> Kempermann G. Regional effects of wheel running<strong>and</strong> environmental enrichment on cell genesis <strong>and</strong> microgliaDownloaded from physrev.physiology.org on November 25, 2005Physiol Rev • VOL 85 • APRIL 2005 • www.prv.org


NEUROGENESIS IN THE ADULT BRAIN 559proliferation in the adult murine neocortex. Cereb Cortex 13: 845–851, 2003.135. Eichenbaum H. A cortical-hippocampal system for declarativememory. Nat Rev Neurosci 1: 41–50, 2000.136. Eichenbaum H. The hippocampus <strong>and</strong> declarative memory: cognitivemechanisms <strong>and</strong> neural codes. Behav Brain Res 127: 199–207, 2001.137. Eisch AJ, Barrot M, Schad CA, Self DW, <strong>and</strong> Nestler EJ.Opiates inhibit neurogenesis in the adult rat hippocampus. ProcNatl Acad Sci USA 97: 7579–7584, 2000.138. Ekdahl CT, Claasen JH, Bonde S, Kokaia Z, <strong>and</strong> Lindvall O.Inflammation is detrimental for neurogenesis in adult brain. ProcNatl Acad Sci USA 100: 13632–13637, 2003.139. Ekstrom AD, Kahana MJ, Caplan JB, Fields TA, Isham EA,Newman EL, <strong>and</strong> Fried I. Cellular networks underlying humanspatial navigation. Nature 425: 184–188, 2003.140. Elliott RC, Khademi S, Pleasure SJ, Parent JM, <strong>and</strong> LowensteinDH. Differential regulation of basic helix-loop-helix mRNAsin the dentate gyrus following status epilepticus. Neuroscience 106:79–88, 2001.141. Emsley JG <strong>and</strong> Hagg T. Endogenous <strong>and</strong> exogenous ciliary neurotrophicfac<strong>to</strong>r enhances forebrain neurogenesis in adult mice.Exp Neurol 183: 298–310, 2003.142. Endl E, Hollmann C, <strong>and</strong> Gerdes J. Antibodies against the Ki-67protein: assessment of the growth fraction <strong>and</strong> <strong>to</strong>ols for cell cycleanalysis. In: Methods in Cell Biology. Orl<strong>and</strong>o, FL: Academic, 2001,p. 399–418.143. Engert F <strong>and</strong> Bonhoeffer T. Dendritic spine changes associatedwith hippocampal long-term synaptic plasticity. Nature 399: 66–70,1999.144. Eriksson PS, Perfilieva E, Bjork-Eriksson T, Alborn AM, NordborgC, Peterson DA, <strong>and</strong> Gage FH. <strong>Neurogenesis</strong> in the adulthuman hippocampus. Nat Med 4: 1313–1317, 1998.145. Ernfors P, Bengzon J, Kokaia Z, Persson H, <strong>and</strong> Lindvall O.Increased levels of messenger RNAs for neurotrophic fac<strong>to</strong>rs in thebrain during kindling epilep<strong>to</strong>genesis. Neuron 7: 165–176, 1991.146. Ettlin RA, Perentes E, Kolopp M, Lardelli P, Arnold J,Karami<strong>to</strong>poulou E, <strong>and</strong> Oberholzer M. Overview of quantitativemethods in <strong>to</strong>xicologic pathology. In Vivo 7: 315–324, 1993.147. Everitt BJ <strong>and</strong> Wolfe ME. Psychomo<strong>to</strong>r stimulant addiction: aneural systems perspective. J Neurosci 22: 3312–3320, 2002.148. Fabel K, Fabel K, Tam B, Kaufer D, Baiker A, Simmons N, KuoCJ, <strong>and</strong> Palmer TD. VEGF is necessary for exercise-induced adulthippocampal neurogenesis. Eur J Neurosci 18: 2803–2812, 2003.149. Falconer EM <strong>and</strong> Galea LA. Sex differences in cell proliferation,cell death <strong>and</strong> defensive behavior following acute preda<strong>to</strong>r odorstress in adult rats. Brain Res 975: 22–36, 2003.150. Falkai P <strong>and</strong> Bogerts B. Cell loss in the hippocampus of schizophrenics.Eur Arch Psychiatry Neurol Sci 236: 154–161, 1986.151. Falkenberg T, Mohammed AK, Henriksson B, Persson H, WinbladB, <strong>and</strong> Lindefors N. Increased expression of brain-derivedneurotrophic fac<strong>to</strong>r mRNA in rat hippocampus is associated withimproved spatial memory <strong>and</strong> enriched environment. Neurosci Lett138: 153–156, 1992.152. Farrell M, Howes S, Bebbing<strong>to</strong>n P, Brugha T, Jenkins R,Lewis G, Marsden J, Taylor C, <strong>and</strong> Meltzer H. Nicotine, alcohol<strong>and</strong> drug dependence <strong>and</strong> psychiatric comorbidity: results of anational household survey. Br J Psychiatry 179: 432–437, 2001.153. Feng R, Rampon C, Tang YP, Shrom D, Jin J, Kyin M, SopherB, Martin GM, Kim SH, <strong>and</strong> Langdon RB. Deficient neurogenesisin forebrain-specific presenilin-1 knockout mice is associated withreduced clearance of hippocampal memory traces. Neuron 32:911–926, 2001.154. Filippov V, Kronenberg G, Pivneva T, Reuter K, Steiner B,Wang LP, Yamaguchi M, Kettenmann H, <strong>and</strong> Kempermann G.Subpopulation of nestin-expressing progeni<strong>to</strong>r cells in the adultmurine hippocampus shows electrophysiological <strong>and</strong> morphologicalcharacteristics of astrocytes. Mol Cell Neurosci 23: 373–382,2003.155. Francis F, Koulakoff A, Boucher D, Chafey P, Schaar B, VinetMC, Friocourt G, McDonnell N, Reiner O, Kahn A, McConnellSK, Berwald-Netter Y, Denoulet P, <strong>and</strong> Chelly J. Doublecortinis a developmentally regulated, microtubule-associated protein expressedin migrating <strong>and</strong> differentiating neurons. Neuron 23: 247–256, 1999.156. Frederiksen K <strong>and</strong> McKay RD. Proliferation <strong>and</strong> differentiationof rat neuroepithelial precursor cells in vivo. J Neurosci 8: 1144–1151, 1988.157. Freed CR, Greene PE, Breeze RE, Tsai WY, DuMouchel W,Kao R, Dillon S, Winfield H, Culver S, Trojanowski JQ, EidelbergD, <strong>and</strong> Fahn S. Transplantation of embryonic dopamineneurons for severe Parkinson’s disease. N Engl J Med 344: 710–719,2001.158. Fujii S, Ji Z, Morita N, <strong>and</strong> Sumikawa K. Acute <strong>and</strong> chronicnicotine exposure differentially facilitate the induction of LTP.Brain Res 846: 137–143, 1999.159. Fukuda S, Ka<strong>to</strong> F, Tozuka Y, Yamaguchi M, Miyamo<strong>to</strong> Y, <strong>and</strong>Hisatsune T. Two distinct subpopulations of nestin-positive cellsin adult mouse dentate gyrus. J Neurosci 23: 9357–9366, 2003.160. Gage FH. Stem cells of the central nervous system. Curr OpinNeurobiol 8: 671–676, 1998.161. Gaiano N <strong>and</strong> Fishell G. The role of notch in promoting glial <strong>and</strong>neural stem cell fates. Annu Rev Neurosci 25: 471–490, 2002.162. Gal<strong>and</strong> P <strong>and</strong> Degraef C. Cyclin/PCNA immunostaining as analternative <strong>to</strong> tritiated thymidine pulse labelling for marking Sphase cells in paraffin sections from animal <strong>and</strong> human tissues. CellTissue Kinet 22: 383–392, 1989.163. Galea LA <strong>and</strong> McEwen BS. Sex <strong>and</strong> seasonal differences in therate of cell proliferation in the dentate gyrus of adult wild meadowvoles. Neuroscience 89: 955–964, 1999.164. Gall CM <strong>and</strong> Isackson PJ. Limbic seizures increase neuronalproduction of messenger RNA for nerve growth fac<strong>to</strong>r. Science 245:758–761, 1989.165. Gallo V, Pende M, Scherer S, Molne M, <strong>and</strong> Wright P. Expression<strong>and</strong> regulation of kainate <strong>and</strong> AMPA recep<strong>to</strong>rs in uncommitted<strong>and</strong> committed neural progeni<strong>to</strong>rs. Neurochem Res 20: 549–560,1995.166. Garcia-Verdugo JM, Doetsch F, Wichterle H, Lim DA, <strong>and</strong>Alvarez-Buylla A. Architecture <strong>and</strong> cell types of the adult subventricularzone: in search of the stem cells. J Neurobiol 36: 234–248,1998.167. Geuna S, Borrione P, Fornaro M, <strong>and</strong> Giacobini-Robecchi MG.<strong>Adult</strong> stem cells <strong>and</strong> neurogenesis: his<strong>to</strong>rical roots <strong>and</strong> state of theart. Anat Rec 265: 132–141, 2001.168. Gleeson JG, Lin PT, Flanagan LA, <strong>and</strong> Walsh CA. Doublecortinis a microtubule-associated protein <strong>and</strong> is expressed widely bymigrating neurons. Neuron 23: 257–271, 1999.169. Goldman SA, Kirschenbaum B, Harrison-Restelli C, <strong>and</strong> ThalerHT. Neuronal precursors of the adult rat subependymal zonepersist in<strong>to</strong> senescence, with no decline in spatial extent or response<strong>to</strong> BDNF. J Neurobiol 32: 554–566, 1997.170. Goldsmith SK <strong>and</strong> Joyce JN. Alterations in hippocampal mossyfiber pathway in schizophrenia <strong>and</strong> Alzheimer’s disease. Biol Psychiatry37: 122–126, 1995.171. Gomez-Pinilla F, So V, <strong>and</strong> Kesslak JP. Spatial learning <strong>and</strong>physical activity contribute <strong>to</strong> the induction of fibroblast growthfac<strong>to</strong>r: neural substrates for increased cognition associated withexercise. Neuroscience 85: 53–61, 1998.172. Gomez-Pinilla F, van der Wal EA, <strong>and</strong> Cotman CW. Possiblecoordinated gene expressions for FGF recep<strong>to</strong>r, FGF-5, <strong>and</strong> FGF-2following seizures. Exp Neurol 133: 164–174, 1995.173. Go<strong>to</strong> Y <strong>and</strong> O’Donnell P. Synchronous activity in the hippocampus<strong>and</strong> nucleus accumbens in vivo. J Neurosci 21: RC131, 2001.174. Gotz M. Glial cells generate neurons—master control within CNSregions: developmental perspectives on neural stem cells. Neuroscientist9: 379–397, 2003.175. Gould E, Beylin A, Tanapat P, Reeves A, <strong>and</strong> Shors TJ. Learningenhances adult neurogenesis in the hippocampal formation.Nat Neurosci 2: 260–265, 1999.176. Gould E, Cameron HA, Daniels DC, Woolley CS, <strong>and</strong> McEwenBS. Adrenal hormones suppress cell division in the adult rat dentategyrus. J Neurosci 12: 3642–3650, 1992.177. Gould E, McEwen BS, Tanapat P, Galea LA, <strong>and</strong> Fuchs E.<strong>Neurogenesis</strong> in the dentate gyrus of the adult tree shrew is regulatedby psychosocial stress <strong>and</strong> NMDA recep<strong>to</strong>r activation. J Neurosci17: 2492–2498, 1997.Downloaded from physrev.physiology.org on November 25, 2005Physiol Rev • VOL 85 • APRIL 2005 • www.prv.org


560 ABROUS, KOEHL, AND LE MOAL178. Gould E, Reeves AJ, Fallah M, Tanapat P, Gross CG, <strong>and</strong>Fuchs E. Hippocampal neurogenesis in adult Old World primates.Proc Natl Acad Sci USA 96: 5263–5267, 1999.179. Gould E, Reeves AJ, Graziano MS, <strong>and</strong> Gross CG. <strong>Neurogenesis</strong>in the neocortex of adult primates. Science 286: 548–552, 1999.180. Gould E <strong>and</strong> Tanapat P. Lesion-induced proliferation of neuronalprogeni<strong>to</strong>rs in the dentate gyrus of the adult rats. Neuroscience 80:427–436, 1997.181. Gould E, Tanapat P, McEwen BS, Flugge G, <strong>and</strong> Fuchs E.Proliferation of granule cell precursors in the dentate gyrus of adultmonkeys is diminished by stress. Proc Natl Acad Sci USA 95:3168–3171, 1998.182. Gould E, Vail N, Wagers M, <strong>and</strong> Gross CG. <strong>Adult</strong>-generatedhippocampal <strong>and</strong> neocortical neurons in macaques have a transientexistence. Proc Natl Acad Sci USA 98: 10910–10917, 2001.183. Gould E, Woolley CS, <strong>and</strong> McEwen BS. Adrenal steroids regulatepostnatal development of the rat dentate gyrus. I. Effects ofglucocorticoids on cell death. J Comp Neurol 313: 479–485, 1991.184. Gould MN <strong>and</strong> Yatvin MB. The effects of x-irradiation on theearly stages of the memory system in rats. Physiol Behav 11:177–179, 1973.185. Graham YP, Heim C, Goodman SH, Miller AH, <strong>and</strong> NemeroffCB. The effects of neonatal stress on brain development: implicationsfor psychopathology. Dev Psychopathol 11: 545–565, 1999.186. Gr<strong>and</strong>barbe L, Bouissac J, R<strong>and</strong> M, Hrabe DA, Artavanis-Tsakonas S, <strong>and</strong> Mohier E. Delta-Notch signaling controls thegeneration of neurons/glia from neural stem cells in a stepwiseprocess. Development 130: 1391–1402, 2003.187. Gray JA <strong>and</strong> McNaugh<strong>to</strong>n N. Comparison between the behavioraleffects of septal <strong>and</strong> hippocampal lesions: a review. NeurosciBiobehav Rev 7: 119–188, 1982.188. Gray WP <strong>and</strong> Sundstrom LE. Kainic acid increases the proliferationof granule cell progeni<strong>to</strong>rs in the dentate gyrus of the adultrat. Brain Res 790: 52–59, 1998.189. Gross CG. <strong>Neurogenesis</strong> in the adult brain: death of a dogma. NatRev Neurosci 1: 67–73, 2000.190. Gross RE, Mehler MF, Mabie PC, Zang Z, Santschi L, <strong>and</strong>Kessler JA. Bone morphogenetic proteins promote astroglial lineagecommitment by mammalian subventricular zone progeni<strong>to</strong>rcells. Neuron 17: 595–606, 1996.191. Grover CA <strong>and</strong> Frye GD. Ethanol effects on synaptic neurotransmission<strong>and</strong> tetanus-induced synaptic plasticity in hippocampalslices of chronic in vivo lead-exposed adult rats. Brain Res 734:61–71, 1996.192. Grutzendler J, Kasthuri N, <strong>and</strong> Gan WB. Long-term dendriticspine stability in the adult cortex. Nature 420: 812–816, 2002.193. Gu W, Brannstrom T, <strong>and</strong> Wester P. Cortical neurogenesis inadult rats after reversible pho<strong>to</strong>thrombotic stroke. J Cereb BloodFlow Metab 20: 1166–1173, 2000.194. Guerra D, Sole A, Cami J, <strong>and</strong> Tobena A. Neuropsychologicalperformance in opiate addicts after rapid de<strong>to</strong>xification. Drug AlcoholDependence 20: 261–270, 1987.195. Gurvits TV, Shen<strong>to</strong>n ME, Hokama H, Ohta H, Lasko NB,Gilbertson MW, Orr SP, Kikinis R, Jolesz FA, McCarley RW,<strong>and</strong> Pitman RK. Magnetic resonance imaging study of hippocampalvolume in chronic, combat-related posttraumatic stress disorder.Biol Psychiatry 40: 1091–1099, 1996.196. Gutiérrez R. The GABAergic phenotype of the “glutamatergic”granule cells of the dentate gyrus. Prog Neurobiol 71: 337–358,2003.197. Guzman M, Galve-Roperh I, <strong>and</strong> Sanchez C. Ceramide: a newsecond messenger of cannabinoid action. Trends Pharmacol Sci22: 19–22, 2001.198. Hack I, Bancila M, Loulier K, Carroll P, <strong>and</strong> Cremer H. Reelinis a detachment signal in tangential chain-migration during postnatalneurogenesis. Nat Neurosci 5: 939–945, 2002.199. Halim ND, Weickert CS, McClin<strong>to</strong>ck BW, Weinberger DR, <strong>and</strong>Lipska BK. Effects of chronic haloperidol <strong>and</strong> clozapine treatmen<strong>to</strong>n neurogenesis in the adult rat hippocampus. Neuropsychopharmacology29: 1063–1069, 2004.200. Hamid S, Dawe GS, Gray JA, <strong>and</strong> Stephenson JD. Nicotineinduces long-lasting potentiation in the dentate gyrus of nicotineprimedrats. Neurosci Res 29: 81–85, 1997.201. Hampson RE, Simeral JD, <strong>and</strong> Deadwyler SA. Distribution ofspatial <strong>and</strong> nonspatial information in dorsal hippocampus. Nature402: 610–614, 1999.202. Harrison JM, Allen RG, Pellegrino MJ, Williams JT, <strong>and</strong> ManzoniOJ. Chronic morphine treatment alters endogenous opioidcontrol of hippocampal mossy fiber synaptic transmission. J Neurophysiol87: 2464–2470, 2002.203. Harrison PJ. The neuropathology of schizophrenia. A criticalreview of the data <strong>and</strong> their interpretation. Brain 122: 593–624,1999.204. Hastings NB <strong>and</strong> Gould E. Rapid extension of axons in<strong>to</strong> the CA3region by adult-generated granule cells. J Comp Neurol 413: 146–154, 1999.205. Heale VR, V<strong>and</strong>erwolf CH, <strong>and</strong> Kavaliers M. Components ofweasel <strong>and</strong> fox odors elicit fast wave bursts in the dentate gyrus ofrats. Behav Brain Res 63: 159–165, 1994.206. Heckers S. Neuroimaging studies of the hippocampus in schizophrenia.Hippocampus 11: 520–528, 2001.207. Heim C <strong>and</strong> Nemeroff CB. The role of childhood trauma in theneurobiology of mood <strong>and</strong> anxiety disorders: preclinical <strong>and</strong> clinicalstudies. Biol Psychiatry 49: 1023–1039, 2001.208. Heine VM, Maslam S, Joels M, <strong>and</strong> Lucassen PJ. Prominentdecline of newborn cell proliferation, differentiation, <strong>and</strong> apop<strong>to</strong>sisin the aging dentate gyrus, in absence of an age-related hypothalamus-pituitary-adrenalaxis activation. Neurobiol Aging 25: 361–375, 2004.209. Heine VM, Maslam S, Zareno J, Joels M, <strong>and</strong> Lucassen PJ.Suppressed proliferation <strong>and</strong> apop<strong>to</strong>tic changes in the rat dentategyrus after acute <strong>and</strong> chronic stress are reversible. Eur J Neurosci19: 131–144, 2004.210. Heins N, Malatesta P, Cecconi F, Nakafuku M, Tucker KL,Hack MA, Chapou<strong>to</strong>n P, Barde YA, <strong>and</strong> Gotz M. Glial cellsgenerate neurons: the role of the transcription fac<strong>to</strong>r Pax6. NatNeurosci 5: 308–315, 2002.211. Heishman SJTRC <strong>and</strong> Henningfiled JE. Nicotine <strong>and</strong> smoking:a review of effects on human performance. Exp Clin Psychopharmacol2: 345–395, 1994.212. Hellsten J, Wennström M, Mohapel P, Ekdahl CT, Bengzon J,<strong>and</strong> Tingström A. Electroconvulsive seizures increase hippocampalneurogenesis after chronic corticosterone treatment. EurJ Neurosci 16: 283–290, 2002.213. Henderson VW, Paganini-Hill A, Miller BL, Elble RJ, ReyesPF, Shoupe D, McCleary CA, Klein RA, Hake AM, <strong>and</strong> FarlowMR. Estrogen for Alzheimer’s disease in women: r<strong>and</strong>omized, double-blind,placebo-controlled trial. Neurology 54: 295–301, 2000.214. Hendzel MJ, Wei Y, Mancini MA, Van Hooser A, Ranalli T,Brinkley BR, Bazett-Jones DP, <strong>and</strong> Allis CD. Mi<strong>to</strong>sis-specificphosphorylation of his<strong>to</strong>ne H3 initiates primarily within pericentromericheterochromatin during G 2 <strong>and</strong> spreads in an orderedfashion coincident with mi<strong>to</strong>tic chromosome condensation. Chromosoma106: 348–360, 1997.215. Henry C, Guegant G, Cador M, Arnauld E, Arsaut J, Le MoalM, <strong>and</strong> Demotes-Mainard J. Prenatal stress in rats facilitatesamphetamine-induced sensitization <strong>and</strong> induces long-lastingchanges in dopamine recep<strong>to</strong>rs in the nucleus accumbens. BrainRes 685: 179–186, 1995.216. Hering H <strong>and</strong> Sheng M. Dendritic spines: structure, dynamics <strong>and</strong>regulation. Nat Rev Neurosci 2: 880–888, 2001.217. Herman JP <strong>and</strong> Abrous DN. Dopaminergic neural grafts afterfifteen years: results <strong>and</strong> perspectives. Prog Neurobiol 44: 1–35,1994.218. Herrera DG, Yague AG, Johnsen-Soriano S, Bosch-Morell F,Collado-Morente L, Muriach M, Romero FJ, <strong>and</strong> Garcia-VerdugoJM. Selective impairment of hippocampal neurogenesis bychronic alcoholism: protective effects of an antioxidant. Proc NatlAcad Sci USA 100: 7919–7924, 2003.219. Holmes MM <strong>and</strong> Galea LA. Defensive behavior <strong>and</strong> hippocampalcell proliferation: differential modulation by naltrexone duringstress. Behav Neurosci 116: 160–168, 2002.220. Hu H, Tomasiewicz H, Magnuson T, <strong>and</strong> Rutishauser U. Therole of polysialic acid in migration of olfac<strong>to</strong>ry bulb interneuronprecursors in the subventricular zone. Neuron 16: 735–743, 1996.Downloaded from physrev.physiology.org on November 25, 2005Physiol Rev • VOL 85 • APRIL 2005 • www.prv.org


NEUROGENESIS IN THE ADULT BRAIN 561221. Huang L <strong>and</strong> Bittman EL. Olfac<strong>to</strong>ry bulb cells generated in adultmale golden hamsters are specifically activated by exposure <strong>to</strong>estrous females. Horm Behav 41: 343–350, 2002.222. Hughes P <strong>and</strong> Dragunow M. Induction of immediate-early genes<strong>and</strong> the control of neurotransmitter-regulated gene expressionwithin the nervous system. Pharmacol Rev 47: 133–178, 1995.223. Huttmann K, Sadgrove M, Wallraff A, Hinterkeuser S, KirchhoffF, Steinhauser C, <strong>and</strong> Gray WP. Seizures preferentiallystimulate proliferation of radial glia-like astrocytes in the adultdentate gyrus: functional <strong>and</strong> immunocy<strong>to</strong>chemical analysis. EurJ Neurosci 18: 2769–2778, 2003.224. Ikonomidou C, Bittigau P, Ishimaru MJ, Wozniak DF, Koch C,Genz K, Price MT, Stefovska V, Horster F, Tenkova T, DikranianK, <strong>and</strong> Olney JW. Ethanol-induced apop<strong>to</strong>tic neurodegeneration<strong>and</strong> fetal alcohol syndrome. Science 287: 1056–1060, 2000.225. Imura T, Kornblum HI, <strong>and</strong> Sofroniew MV. The predominantneural stem cell isolated from postnatal <strong>and</strong> adult forebrain but notearly embryonic forebrain expresses GFAP. J Neurosci 23: 2824–2832, 2003.226. Iso T, Kedes L, <strong>and</strong> Hamamori Y. HES <strong>and</strong> HERP families:multiple effec<strong>to</strong>rs of the Notch signaling pathway. J Cell Physiol194: 237–255, 2003.227. Izquierdo I <strong>and</strong> Medina JH. Memory formation: the sequence ofbiochemical events in the hippocampus <strong>and</strong> its connection <strong>to</strong> activityin other brain structures. Neurobiol Learn Memory 68: 285–316, 1997.228. Jacks T, Fazeli A, Schmitt EM, Bronson RT, Goodell MA, <strong>and</strong>Weinberg RA. Effects of an Rb mutation in the mouse. Nature 359:295–300, 1992.229. Jacobs LF <strong>and</strong> Schenk F. Unpacking the cognitive map: theparallel map theory of hippocampal function. Psychol Rev 110:285–315, 2003.230. Jang MH, Shin MC, Jung SB, Lee TH, Bahn GH, Kwon YK, KimEH, <strong>and</strong> Kim CJ. Alcohol <strong>and</strong> nicotine reduce cell proliferation<strong>and</strong> enhance apop<strong>to</strong>sis in dentate gyrus. Neuroreport 13: 1509–1513, 2002.231. Jankovski A, Garcia C, Soriano E, <strong>and</strong> Sotelo C. Proliferation,migration <strong>and</strong> differentiation of neuronal progeni<strong>to</strong>r cells in theadult mouse subventricular zone surgically separated from its olfac<strong>to</strong>rybulb. Eur J Neurosci 10: 3853–3868, 1998.232. Jessberger S <strong>and</strong> Kempermann G. <strong>Adult</strong>-born hippocampal neuronsmature in<strong>to</strong> activity-dependent responsiveness. Eur J Neurosci18: 2707–2712, 2003.233. Jeste DV <strong>and</strong> Lohr JB. Hippocampal pathologic findings inschizophrenia. A morphometric study. Arch Gen Psychiatry 46:1019–1024, 1989.234. Jiang W, Wang JC, Zhang Z, Sheerin AH, <strong>and</strong> Zhang X. Responseof seizure-induced newborn neurons in the dentate gyrus ofadult rats <strong>to</strong> second episode of seizures. Brain Res 1006: 248–252,2004.235. Jin K, Mao XO, Sun Y, Xie L, <strong>and</strong> Greenberg DA. Stem cellfac<strong>to</strong>r stimulates neurogenesis in vitro <strong>and</strong> in vivo. J Clin Invest110: 311–319, 2003.236. Jin K, Mao XO, Sun Y, Xie L, Jin L, Nishi E, Klagsbrun M, <strong>and</strong>Greenberg DA. Heparin-binding epidermal growth fac<strong>to</strong>r-likegrowth fac<strong>to</strong>r: hypoxia-inducible expression in vitro <strong>and</strong> stimulationof neurogenesis in vitro <strong>and</strong> in vivo. J Neurosci 22: 5365–5373,2002.237. Jin K, Minami M, Lan JQ, Mao XO, Batteur S, Simon RP, <strong>and</strong>Greenberg DA. <strong>Neurogenesis</strong> in dentate subgranular zone <strong>and</strong>rostral subventricular zone after focal cerebral ischemia in the rat.Proc Natl Acad Sci USA 98: 4710–4715, 2001.238. Jin K, Sun Y, Xie L, Batteur S, Mao XO, Smelick C, LogvinovaA, <strong>and</strong> Greenberg DA. <strong>Neurogenesis</strong> <strong>and</strong> aging: FGF-2 <strong>and</strong> HB-EGF res<strong>to</strong>re neurogenesis in hippocampus <strong>and</strong> subventricular zoneof aged mice. Aging Cell 2: 175–183, 2003.239. Jin K, Sun Y, Xie L, Peel A, Mao XO, Batteur S, <strong>and</strong> GreenbergDA. Directed migration of neuronal precursors in<strong>to</strong> the ischemiccerebral cortex <strong>and</strong> striatum. Mol Cell Neurosci 24: 171–189, 2003.240. Jin K, Xie L, Childs J, Sun Y, Mao XO, Logvinova A, <strong>and</strong>Greenberg DA. Cerebral neurogenesis is induced by intranasaladministration of growth fac<strong>to</strong>rs. Ann Neurol 53: 405–409, 2003.241. Jin K, Zhu Y, Sun Y, Mao XO, Xie L, <strong>and</strong> Greenberg DA.Vascular endothelial growth fac<strong>to</strong>r (VEGF) stimulates neurogenesisin vitro <strong>and</strong> in vivo. Proc Natl Acad Sci USA 99: 11946–11950,2002.242. Johansson CB, Svensson M, Wallstedt L, Janson AM, <strong>and</strong>Frisen J. Neural stem cells in the adult human brain. Exp Cell Res253: 733–736, 1999.243. Junier MP. What role(s) for TGFalpha in the central nervoussystem? Prog Neurobiol 62: 443–473, 2000.244. Justice NJ <strong>and</strong> Jan YN. Variations on the Notch pathway inneural development. Curr Opin Neurobiol 12: 64–70, 2002.245. Kamiguchi H, Yoshida K, Wakamo<strong>to</strong> H, Inaba M, Sasaki H,Otani M, <strong>and</strong> Toya S. Cy<strong>to</strong>kine-induced selective increase ofhigh-molecular-weight bFGF isoforms <strong>and</strong> their subcellular kineticsin cultured rat hippocampal astrocytes. Neurochem Res 21:701–706, 1996.246. Kaneko Y, Sakakibara S, Imai T, Suzuki A, Nakamura Y,Sawamo<strong>to</strong> K, Ogawa Y, Toyama Y, Miyata T, <strong>and</strong> Okano H.Musashi1: an evolutionally conserved marker for CNS progeni<strong>to</strong>rcells including neural stem cells. Dev Neurosci 22: 139–153, 2000.247. Kaplan MS. <strong>Neurogenesis</strong> in the 3-month-old rat visual cortex.J Comp Neurol 195: 323–338, 1981.248. Kaplan MS. Formation <strong>and</strong> turnover of neurons in young <strong>and</strong>senescent animals: an electronmicroscopic <strong>and</strong> morphometricanalysis. Ann NY Acad Sci 457: 173–192, 1985.249. Kaplan MS. Environment complexity stimulates visual cortex neurogenesis:death of a dogma <strong>and</strong> a research career. Trends Neurosci24: 617–620, 2001.250. Kaplan MS <strong>and</strong> Bell DH. Neuronal proliferation in the 9-montholdrodent-radioau<strong>to</strong>graphic study of granule cells in the hippocampus.Exp Brain Res 52: 1–5, 1983.251. Kaplan MS <strong>and</strong> Bell DH. Mi<strong>to</strong>tic neuroblasts in the 9-day-old <strong>and</strong>11-month-old rodent hippocampus. J Neurosci 4: 1429–1441, 1984.252. Kaplan MS <strong>and</strong> Hinds JW. <strong>Neurogenesis</strong> in the adult rat: electronmicroscopic analysis of light radioau<strong>to</strong>graphs. Science 197: 1092–1094, 1977.253. Kaplan MS, McNelly NA, <strong>and</strong> Hinds JW. Population dynamics ofadult-formed granule neurons of the rat olfac<strong>to</strong>ry bulb. J CompNeurol 239: 117–125, 1985.254. Karishma KK <strong>and</strong> Herbert J. Dehydroepi<strong>and</strong>rosterone (DHEA)stimulates neurogenesis in the hippocampus of the rat, promotessurvival of newly formed neurons <strong>and</strong> prevents corticosteroneinducedsuppression. Eur J Neurosci 16: 445–453, 2002.255. Kasai H, Matsuzaki M, Noguchi J, Yasumatsu N, <strong>and</strong> NakaharaH. Structure-stability-function relationships of dendriticspines. Trends Neurosci 26: 360–368, 2003.256. Ka<strong>to</strong> T, Yokouchi K, Fukushima N, Kawagishi K, Li Z, <strong>and</strong>Moriizumi T. Continual replacement of newly-generated olfac<strong>to</strong>ryneurons in adult rats. Neurosci Lett 307: 17–20, 2001.257. Ka<strong>to</strong>h-Semba R, Asano T, Ueda H, Morishita R, Takeuchi IK,Inaguma Y, <strong>and</strong> Ka<strong>to</strong> K. Riluzole enhances expression of brainderivedneurotrophic fac<strong>to</strong>r with consequent proliferation of granuleprecursor cells in the rat hippocampus. FASEB J 16: 1328–1330,2002.258. Kawai T, Takagi N, Miyake-Takagi K, Okuyama N, MochizukiN, <strong>and</strong> Takeo S. Characterization of BrdU-positive neurons inducedby transient global ischemia in adult hippocampus. J CerebBlood Flow Metab 24: 548–555, 2004.259. Kee N, Sivalingam S, Boonstra R, <strong>and</strong> Woj<strong>to</strong>wicz JM. Theutility of Ki-67 <strong>and</strong> BrdU as proliferative markers of adult neurogenesis.J Neurosci Methods 115: 97–105, 2002.260. Kee NJ, Pres<strong>to</strong>n E, <strong>and</strong> Woj<strong>to</strong>wicz JM. Enhanced neurogenesisafter transient global ischemia in the dentate gyrus of the rat. ExpBrain Res 136: 313–320, 2001.261. Kempermann G, Br<strong>and</strong>on EP, <strong>and</strong> Gage FH. Environmentalstimulation of 129/SvJ mice causes increased cell proliferation <strong>and</strong>neurogenesis in the adult dentate gyrus. Curr Biol 8: 939–942, 1998.262. Kempermann G <strong>and</strong> Gage FH. Experience-dependent regulationof adult hippocampal neurogenesis: effects of long-term stimulation<strong>and</strong> stimulus withdrawal. Hippocampus 9: 321–332, 1999.263. Kempermann G, Gast D, Kronenberg G, Yamaguchi M, <strong>and</strong>Gage FH. Early determination <strong>and</strong> long-term persistence of adult-Downloaded from physrev.physiology.org on November 25, 2005Physiol Rev • VOL 85 • APRIL 2005 • www.prv.org


562 ABROUS, KOEHL, AND LE MOALgenerated new neurons in the hippocampus of mice. Development130: 391–399, 2003.264. Kempermann G, Gast D, <strong>and</strong> Gage FH. Neuroplasticity in oldage: sustained fivefold induction of hippocampal neurogenesis bylong-term environmental enrichment. Ann Neurol 52: 135–143,2002.265. Kempermann G, Jessberger S, Steiner B, <strong>and</strong> Kronenberg G.Miles<strong>to</strong>nes of neuronal development in the adult hippocampus.Trends Neurosci 27: 447–452, 2004.266. Kempermann G <strong>and</strong> Gage FH. Genetic influence on phenotypicdifferentiation in adult hippocampal neurogenesis. Dev Brain Res134: 1–12, 2002.267. Kempermann G, Kuhn HG, <strong>and</strong> Gage FH. More hippocampalneurons in adult mice living in an enriched environment. Nature386: 493–495, 1997.268. Kempermann G, Kuhn HG, <strong>and</strong> Gage FH. Genetic influence onneurogenesis in the dentate gyrus of adult mice. Proc Natl Acad SciUSA 94: 10409–10414, 1997.269. Kempermann G, Kuhn HG, <strong>and</strong> Gage FH. Experience-inducedneurogenesis in the senescent dentate gyrus. J Neurosci 18: 3206–3212, 1998.270. Kesner RP, Gilbert PE, <strong>and</strong> Wallenstein GV. Testing neuralnetwork models of memory with behavioral experiments. CurrOpin Neurobiol 10: 260–265, 2000.271. Kesslak JP, So V, Choi J, Cotman CW, <strong>and</strong> Gomez-Pinilla F.Learning upregulates brain-derived neurotrophic fac<strong>to</strong>r messengerribonucleic acid: a mechanism <strong>to</strong> facilitate encoding <strong>and</strong> circuitmaintenance? Behav Neurosci 112: 1012–1019, 1998.272. Kiecolt-Glaser JK, Preacher KJ, MacCallum RC, Atkinson C,Malarkey WB, <strong>and</strong> Glaser R. Chronic stress <strong>and</strong> age-relatedincreases in the proinflamma<strong>to</strong>ry cy<strong>to</strong>kine IL-6. Proc Natl Acad SciUSA 100: 9090–9095, 2003.273. Kim JJ, Clark RE, <strong>and</strong> Thompson RF. Hippocampec<strong>to</strong>my impairsthe memory of recently, but not remotely, acquired traceeyeblink conditioned responses. Behav Neurosci 109: 195–203,1995.274. Kim JJ <strong>and</strong> Fanselow MS. Modality-specific retrograde amnesiaof fear. Science 256: 675–677, 1992.275. Kirschenbaum B, Doetsch F, Lois C, <strong>and</strong> Alvarez-Buylla A.<strong>Adult</strong> subventricular zone neuronal precursors continue <strong>to</strong> proliferate<strong>and</strong> migrate in the absence of the olfac<strong>to</strong>ry bulb. J Neurosci19: 2171–2180, 1999.276. Kirschenbaum B <strong>and</strong> Goldman SA. Brain-derived neurotrophicfac<strong>to</strong>r promotes the survival of neurons arising from the adult ratforebrain subependymal zone. Proc Natl Acad Sci USA 92: 210–214,1995.277. Kiss JZ <strong>and</strong> Rougon G. Cell biology of polysialic acid. Curr OpinNeurobiol 7: 640–646, 1997.278. Kitamura T, Mishina M, <strong>and</strong> Sugiyama H. Enhancement ofneurogenesis by running wheel exercises is suppressed in micelacking NMDA recep<strong>to</strong>r epsilon 1 subunit. Neurosci Res 47: 55–63,2003.279. Koehl M, Bjijou Y, Le Moal M, <strong>and</strong> Cador M. Nicotine-inducedlocomo<strong>to</strong>r activity is increased by preexposure of rats <strong>to</strong> prenatalstress. Brain Res 882: 196–200, 2000.280. Koehl M, Lemaire V, Mayo W, Abrous DN, Maccari S, PiazzaPV, Le Moal M, <strong>and</strong> Vallee M. Individual vulnerability <strong>to</strong> substanceabuse <strong>and</strong> affective disorders: role of early environmentalinfluences. Neuro<strong>to</strong>xicity Res 4: 281–296, 2002.281. Koketsu D, Mikami A, Miyamo<strong>to</strong> Y, <strong>and</strong> Hisatsune T. Nonrenewalof neurons in the cerebral neocortex of adult macaquemonkeys. J Neurosci 23: 937–942, 2003.282. Kolb B, Pedersen B, Ballermann M, Gibb R, <strong>and</strong> Whishaw IQ.Embryonic <strong>and</strong> postnatal injections of bromodeoxyuridine produceage-dependent morphological <strong>and</strong> behavioral abnormalities.J Neurosci 19: 2337–2346, 1999.283. Koob GF <strong>and</strong> Le Moal M. Drug addiction, dysregulation of reward,<strong>and</strong> allostasis. Neuropsychopharmacology 24: 97–129, 2001.284. Kornack DR <strong>and</strong> Rakic P. Continuation of neurogenesis in thehippocampus of the adult macaque monkey. Proc Natl Acad SciUSA 96: 5768–5773, 1999.285. Kornack DR <strong>and</strong> Rakic P. Cell proliferation without neurogenesisin adult primate neocortex. Science 294: 2127–2130, 2001.286. Kornack DR <strong>and</strong> Rakic P. The generation, migration, <strong>and</strong> differentiationof olfac<strong>to</strong>ry neurons in the adult primate brain. Proc NatlAcad Sci USA 98: 4752–4757, 2001.287. Kostyuk P. Plasticity in Nerve Cell Function. Oxford, UK: Clarendon,1998.288. Kovelman JA <strong>and</strong> Scheibel AB. A neurohis<strong>to</strong>logical correlate ofschizophrenia. Biol Psychiatry 19: 1601–1621, 1984.289. Kronenberg G, Reuter K, Steiner B, Br<strong>and</strong>t MD, Jessberger S,Yamaguchi M, <strong>and</strong> Kempermann G. Subpopulations of proliferatingcells of the adult hippocampus respond differently <strong>to</strong> physiologicneurogenic stimuli. J Comp Neurol 467: 455–463, 2003.290. Kuhn HG, Dickinson-Anson H, <strong>and</strong> Gage FH. <strong>Neurogenesis</strong> inthe dentate gyrus of the adult rat: age-related decrease of neuronalprogeni<strong>to</strong>r proliferation. J Neurosci 16: 2027–2033, 1996.291. Kuhn HG, Winkler J, Kempermann G, Thal LJ, <strong>and</strong> Gage FH.Epidermal growth fac<strong>to</strong>r <strong>and</strong> fibroblast growth fac<strong>to</strong>r-2 have differenteffects on neural progeni<strong>to</strong>rs in the adult rat brain. J Neurosci17: 5820–5829, 1997.292. Kulkarni VA, Jha S, <strong>and</strong> Vaidya VA. Depletion of norepinephrinedecreases the proliferation, but does not influence the survival <strong>and</strong>differentiation, of granule cell progeni<strong>to</strong>rs in the adult rat hippocampus.Eur J Neurosci 16: 2008–2012, 2002.293. Kumihashi K, Uchida K, Miyazaki H, Kobayashi J, TsushimaT, <strong>and</strong> Machida T. Acetylsalicylic acid reduces ischemia-inducedproliferation of dentate cells in gerbils. Neuroreport 12: 915–917,2001.294. Kurki P, V<strong>and</strong>erlaan M, Dolbeare F, Gray J, <strong>and</strong> Tan EM.Expression of proliferating cell nuclear antigen (PCNA)/cyclin duringthe cell cycle. Exp Cell Res 166: 209–219, 1986.295. Lai K, Kaspar BK, Gage FH, <strong>and</strong> Schaffer DV. Sonic hedgehogregulates adult neural progeni<strong>to</strong>r proliferation in vitro <strong>and</strong> in vivo.Nat Neurosci 6: 21–27, 2003.296. Laming PR, Kimelberg H, Robinson S, Salm A, Hawrylak N,Muller C, Roots B, <strong>and</strong> Ng K. Neuronal-glial interactions <strong>and</strong>behaviour. Neurosci Biobehav Rev 24: 295–340, 2000.297. Lamprecht R <strong>and</strong> LeDoux J. Structural plasticity <strong>and</strong> memory.Nat Rev Neurosci 5: 45–54, 2004.298. Lauder JM <strong>and</strong> Krebs H. Sero<strong>to</strong>nin as a differentiation signal inearly neurogenesis. Dev Neurosci 1: 15–30, 1978.299. Lee EY, Chang CY, Hu N, Wang YC, Lai CC, Herrup K, Lee WH,<strong>and</strong> Bradley A. Mice deficient for Rb are nonviable <strong>and</strong> showdefects in neurogenesis <strong>and</strong> haema<strong>to</strong>poiesis. Nature 359: 288–294,1992.300. Lee HJ, Kim JW, Yim SV, Kim MJ, Kim SA, Kim YJ, Kim CJ,<strong>and</strong> Chung JH. Fluoxetine enhances cell proliferation <strong>and</strong> preventsapop<strong>to</strong>sis in dentate gyrus of maternally separated rats. MolPsychiatry 6: 610–728, 2001.301. Lee J, Duan W, <strong>and</strong> Mattson MP. Evidence that brain-derivedneurotrophic fac<strong>to</strong>r is required for basal neurogenesis <strong>and</strong> mediates,in part, the enhancement of neurogenesis by dietary restrictionin the hippocampus of adult mice. J Neurochem 82: 1367–1375,2002.302. Legault M, Rompre PP, <strong>and</strong> Wise RA. Chemical stimulation ofthe ventral hippocampus elevates nucleus accumbens dopamine byactivating dopaminergic neurons of the ventral tegmental area.J Neurosci 20: 1635–1642, 2000.303. Legrier ME, Ducray A, Propper A, Chao M, <strong>and</strong> Kastner A.Cell cycle regulation during mouse olfac<strong>to</strong>ry neurogenesis. CellGrowth Differ 12: 591–601, 2001.304. Lemaire V <strong>and</strong> Agmo A. High <strong>and</strong> low reactivity <strong>to</strong> novelty <strong>and</strong>sexual behavior in male rats. Psychobiology 25: 241–245, 1997.305. Lemaire V, Aurousseau C, Le Moal M, <strong>and</strong> Abrous DN. Behaviouraltrait of reactivity <strong>to</strong> novelty is related <strong>to</strong> hippocampal neurogenesis.Eur J Neurosci 11: 4006–4014, 1999.306. Lemaire V, Koehl M, Le Moal M, <strong>and</strong> Abrous DN. Prenatalstress produces learning deficits associated with an inhibition ofneurogenesis in the hippocampus. Proc Natl Acad Sci USA 97:11032–11037, 2000.307. Lendahl U, Zimmerman LB, <strong>and</strong> McKay RD. CNS stem cellsexpress a new class of intermediate filament protein. Cell 60:585–595, 1990.308. Lescaudron L, Jaffard R, <strong>and</strong> Verna A. Modifications in number<strong>and</strong> morphology of dendritic spines resulting from chronic ethanolDownloaded from physrev.physiology.org on November 25, 2005Physiol Rev • VOL 85 • APRIL 2005 • www.prv.org


NEUROGENESIS IN THE ADULT BRAIN 563consumption <strong>and</strong> withdrawal: a Golgi study in the mouse anterior<strong>and</strong> posterior hippocampus. Exp Neurol 106: 156–161, 1989.309. Leuner B, Mendolia-Loffredo S, Kozorovitskiy Y, Samburg D,Gould E, <strong>and</strong> Shors TJ. Learning enhances the survival of newneurons beyond the time when the hippocampus is required formemory. J Neurosci 24: 7477–7481, 2004.310. Lichtenwalner RJ, Forbes ME, Bennett SA, Lynch CD,Sonntag WE, <strong>and</strong> Riddle DR. Intracerebroventricular infusion ofinsulin-like growth fac<strong>to</strong>r-I ameliorates the age-related decline inhippocampal neurogenesis. Neuroscience 107: 603–613, 2001.311. Lim DA, Tramontin AD, Trevejo JM, Herrera DG, Garcia-Verdugo JM, <strong>and</strong> Alvarez-Buylla A. Noggin antagonizes BMPsignaling <strong>to</strong> create a niche for adult neurogenesis. Neuron 28:713–726, 2000.312. Linnarsson S, Willson CA, <strong>and</strong> Ernfors P. Cell death in regeneratingpopulations of neurons in BDNF mutant mice. Brain Res75: 61–69, 2000.313. Lipska BK <strong>and</strong> Weinberger DR. To model a psychiatric disorderin animals: schizophrenia as a reality test. Neuropsychopharmacology23: 223–239, 2000.314. Liu G <strong>and</strong> Rao Y. Neuronal migration from the forebrain <strong>to</strong> theolfac<strong>to</strong>ry bulb requires a new attractant persistent in the olfac<strong>to</strong>rybulb. J Neurosci 23: 6651–6659, 2003.315. Liu J, Solway K, Messing RO, <strong>and</strong> Sharp FR. Increased neurogenesisin the dentate gyrus after transient global ischemia ingerbils. J Neurosci 18: 7768–7778, 1998.316. Liu S, Wang J, Zhu D, Fu Y, Lukowiak K, <strong>and</strong> Lu YM. Generationof functional inhibi<strong>to</strong>ry neurons in the adult rat hippocampus.J Neurosci 23: 732–736, 2003.317. Lois C <strong>and</strong> Alvarez-Buylla A. Long-distance neuronal migrationin the adult mammalian brain. Science 264: 1145–1148, 1994.318. LoTurco JJ, Owens DF, Heath MJ, Davis MB, <strong>and</strong> KriegsteinAR. GABA <strong>and</strong> glutamate depolarize cortical progeni<strong>to</strong>r cells <strong>and</strong>inhibit DNA synthesis. Neuron 15: 1287–1298, 1995.319. Lu D, Mahmood A, Zhang R, <strong>and</strong> Copp M. Upregulation ofneurogenesis <strong>and</strong> reduction in functional deficits following administrationof DEtA/NONOate, a nitric oxide donor, after traumaticbrain injury in rats. J Neurosurg 99: 351–361, 2003.320. Lu L, Bao G, Chen H, Xia P, Fan X, Zhang J, Pei G, <strong>and</strong> Ma L.Modification of hippocampal neurogenesis <strong>and</strong> neuroplasticity bysocial environments. Exp Neurol 183: 600–609, 2003.321. Lu L, Zeng S, Liu D, <strong>and</strong> Ceng X. Inhibition of the amygdala <strong>and</strong>hippocampal calcium/calmodulin-dependent protein kinase II attenuatesthe dependence <strong>and</strong> relapse <strong>to</strong> morphine differently inrats. Neurosci Lett 291: 191–195, 2000.322. Lund RD. Developement <strong>and</strong> Plasticity of the Brain. An Introduction.New York: Oxford Univ. Press, 1978.323. Luo Y, Cai J, Liu Y, Xue H, Chrest FJ, Wers<strong>to</strong> RP, <strong>and</strong> Rao M.Microarray analysis of selected genes in neural stem <strong>and</strong> progeni<strong>to</strong>rcells. J Neurochem 83: 1481–1497, 2002.324. Lupien SJ <strong>and</strong> McEwen BS. The acute effects of corticosteroidson cognition: integration of animal <strong>and</strong> human model studies.Brain Res Rev 24: 1–27, 1997.325. Luskin MB. Restricted proliferation <strong>and</strong> migration of postnatallygenerated neurons derived from the forebrain subventricular zone.Neuron 11: 173–189, 1993.326. Luskin MB. Neuroblasts of the postnatal mammalian forebrain:their phenotype <strong>and</strong> fate. J Neurobiol 36: 221–233, 1998.327. Ma W, Liu QY, Maric D, Sathanoori R, Chang YH, <strong>and</strong> BarkerJL. Basic FGF-responsive telencephalic precursor cells expressfunctional GABA(A) recep<strong>to</strong>r/Cl-channels in vitro. J Neurobiol 35:277–286, 1998.328. Mackenzie IR. Activated microglia in dementia with Lewy bodies.Neurology 55: 132–134, 2000.329. MacQueen GM, Campbell S, McEwen BS, Macdonald K,Amano S, Joffe RT, Nahmias C, <strong>and</strong> Young LT. Course ofillness, hippocampal function, <strong>and</strong> hippocampal volume in majordepression. Proc Natl Acad Sci USA 100: 1387–1392, 2003.330. Madsen TM, Kristjansen PE, Bolwig TG, <strong>and</strong> Wortwein G.Arrested neuronal proliferation <strong>and</strong> impaired hippocampal functionfollowing fractionated brain irradiation in the adult rat. Neuroscience119: 635–642, 2003.331. Madsen TM, Treschow A, Bengzon J, Bolwig TG, Lindvall O,<strong>and</strong> Tingstrom A. Increased neurogenesis in a model of electroconvulsivetherapy. Biol Psychiatry 47: 1043–1049, 2000.332. Magavi SS, Leavitt BR, <strong>and</strong> Macklis JD. Induction of neurogenesisin the neocortex of adult mice. Nature 405: 951–955, 2000.333. Majewska MD. Neurosteroids: endogenous bimodal modula<strong>to</strong>rsof the GABA A recep<strong>to</strong>r. Mechanism of action <strong>and</strong> physiologicalsignificance. Prog Neurobiol 38: 379–395, 1992.334. Malberg JE <strong>and</strong> Duman RS. Cell proliferation in adult hippocampusis decreased by inescapable stress: reversal by fluoxetine treatment.Neuropsychopharmacology 28: 1562–1571, 2003.335. Malberg JE, Eisch AJ, Nestler EJ, <strong>and</strong> Duman RS. Chronicantidepressant treatment increases neurogenesis in adult rat hippocampus.J Neurosci 20: 9104–9110, 2000.336. Maldonado R. Study of cannabinoid dependence in animals. PharmacolTher 95: 153–164, 2002.337. Malenka RC <strong>and</strong> Nicoll RA. Long-term potentiation—a decade ofprogress? Science 285: 1870–1874, 1999.338. Maletic-Savatic M, Malinow R, <strong>and</strong> Svoboda K. Rapid dendriticmorphogenesis in CA1 hippocampal dendrites induced by synapticactivity. Science 283: 1923–1927, 1999.339. Manev H, Uz T, <strong>and</strong> Manev R. Glia as a putative target forantidepressant treatments. J Affective Disorders. In press.340. Manev H, Uz T, Manev R, <strong>and</strong> Zhang Z. <strong>Neurogenesis</strong> <strong>and</strong>neuroprotection in the adult brain. A putative role for 5-lipoxygenase?Ann NY Acad Sci 939: 45–51, 2001.341. Marczynski TJ. GABAergic deafferentation hypothesis of brainaging <strong>and</strong> Alzheimer’s disease revisited. Brain Res Bull 45: 341–379, 1998.342. Maren S, Aharonov G, <strong>and</strong> Fanselow MS. Neuro<strong>to</strong>xic lesions ofthe dorsal hippocampus <strong>and</strong> Pavlovian fear conditioning in rats.Behav Brain Res 88: 261–274, 1997.343. Marinelli M <strong>and</strong> Piazza PV. Interaction between glucocorticoidhormones, stress <strong>and</strong> psychostimulant drugs. Eur J Neurosci 16:387–394, 2002.344. Markakis EA <strong>and</strong> Gage FH. <strong>Adult</strong>-generated neurons in the dentategyrus send axonal projections <strong>to</strong> field CA3 <strong>and</strong> are surroundedby synaptic vesicles. J Comp Neurol 406: 449–460, 1999.345. Markowska AL, Mooney M, <strong>and</strong> Sonntag WE. Insulin-likegrowth fac<strong>to</strong>r-1 ameliorates age-related behavioral deficits. Neuroscience87: 559–569, 1998.346. Marti E <strong>and</strong> Bovolenta P. Sonic hedgehog in CNS development:one signal, multiple outputs. Trends Neurosci 25: 89–96, 2002.347. Matsuoka N, Nozaki K, Takagi Y, Nishimura M, Hayashi J,Miyatake S, <strong>and</strong> Hashimo<strong>to</strong> N. Adenovirus-mediated gene transferof fibroblast growth fac<strong>to</strong>r-2 increases BrdU-positive cells afterforebrain ischemia in gerbils. Stroke 34: 1519–1525, 2003.348. Mayo W, Lemaire V, Malaterre J, Rodriguez JJ, Cayre M,Stewart MG, Kharouby M, Rougon G, Le Moal M, Piazza PV,<strong>and</strong> Abrous DN. The neurosteroid pregnenolone sulfate enhancesneurogenesis in the young <strong>and</strong> senescent dentate gyrus. NeurobiolAging 26: 103–114, 2005.349. Mayo W, Vallée M, Darnaudéry M, <strong>and</strong> Le Moal M. Neurosteroids:Behavioral Studies. Clif<strong>to</strong>n, NJ: Humana, 1999.350. McEchron MD <strong>and</strong> Disterhoft JF. Hippocampal encoding ofnon-spatial trace conditioning. Hippocampus 9: 385–396, 1999.351. McEwen BS. Plasticity of the hippocampus: adaptation <strong>to</strong> chronicstress <strong>and</strong> allostatic load. Ann NY Acad Sci 933: 265–277, 2001.352. McEwen BS. Sex, stress <strong>and</strong> the hippocampus: allostasis, allostaticload <strong>and</strong> the aging process. Neurobiol Aging 23: 921–939,2002.353. McEwen BS. Early life influences on life-long patterns of behavior<strong>and</strong> health. Ment Retard Dev Disabil Res Rev 9: 149–154, 2003.354. McEwen BS, Cameron H, Chao HM, Gould E, Magarinos AM,Watanabe Y, <strong>and</strong> Woolley CS. Adrenal steroids <strong>and</strong> plasticity ofhippocampal neurons: <strong>to</strong>ward an underst<strong>and</strong>ing of underlying cellular<strong>and</strong> molecular mechanisms. Cell Mol Neurobiol 13: 457–482,1993.355. McGeer PL, Schulzer M, <strong>and</strong> McGeer EG. Arthritis <strong>and</strong> antiinflamma<strong>to</strong>ryagents as possible protective fac<strong>to</strong>rs for Alzheimer’sdisease: a review of 17 epidemiologic studies. Neurology 47: 425–432, 1996.Downloaded from physrev.physiology.org on November 25, 2005Physiol Rev • VOL 85 • APRIL 2005 • www.prv.org


564 ABROUS, KOEHL, AND LE MOAL356. McKay R. Stem cells in the central nervous system. Science 276:66–71, 1997.357. McLardy T <strong>and</strong> Path FRC. Hippocampal zinc <strong>and</strong> structuraldeficit in brains from chronic alcoholics <strong>and</strong> some schizophrenics.J Orthomol Psychiatry 4: 32–36, 1975.358. Mehler MF. Mechanisms regulating lineage diversity during mammaliancerebral cortical neurogenesis <strong>and</strong> gliogenesis. ResultsProbl Cell Differ 39: 27–52, 2002.359. Meirer R, Gurunluoglu R, <strong>and</strong> Siemionow M. Neurogenic perspectiveon vascular endothelial growth fac<strong>to</strong>r: review of the literature.J Reconstr Microsurg 17: 625–630, 2001.360. Memberg SP <strong>and</strong> Hall AK. Dividing neuron precursors expressneuron-specific tubulin. J Neurobiol 27: 26–43, 1995.361. Menezes JR <strong>and</strong> Luskin MB. Expression of neuron-specific tubulindefines a novel population in the proliferative layers of thedeveloping telencephalon. J Neurosci 14: 5399–5416, 1994.362. Mercer A, Ronnholm H, Holmberg J, Lundh H, Heidrich J,Zachrisson O, Ossoinak A, Frisen J, <strong>and</strong> Patrone C. PACAPpromotes neural stem cell proliferation in adult mouse brain.J Neurosci Res 76: 205–215, 2004.363. Mercier F, Kitasako JT, <strong>and</strong> Hat<strong>to</strong>n GI. Ana<strong>to</strong>my of the brainneurogenic zones revisited: frac<strong>to</strong>nes <strong>and</strong> the fibroblast/macrophagenetwork. J Comp Neurol 451: 170–188, 2002.364. Mesulam MM. Neuroplasticity failure in Alzheimer’s disease:bridging the gap between plaques <strong>and</strong> tangles. Neuron 24: 521–529,1999.365. Meyers RA, Zavala AR, <strong>and</strong> Neisew<strong>and</strong>er JL. Dorsal, but notventral, hippocampal lesions disrupt cocaine place conditioning.Neuroreport 14: 2127–2131, 2003.366. Minturn JE, Geschwind DH, Fryer HJ, <strong>and</strong> Hockfield S. Earlypostmi<strong>to</strong>tic neurons transiently express TOAD-64, a neural specificprotein. J Comp Neurol 355: 369–379, 1995.367. Mirescu C, Peters JD, <strong>and</strong> Gould E. Early life experience altersresponse of adult neurogenesis <strong>to</strong> stress. Nat Neurosci. In press.368. Mizrahi A <strong>and</strong> Katz LC. Dendritic stability in the adult olfac<strong>to</strong>rybulb. Nat Neurosci 6: 1201–1207, 2003.369. Mizumatsu S, Monje ML, Morhardt DR, Rola R, Palmer TD,<strong>and</strong> Fike JR. Extreme sensitivity of adult neurogenesis <strong>to</strong> lowdoses of X-irradiation. Cancer Res 63: 4021–4027, 2003.370. Mohammed AH, Henriksson BG, Soderstrom S, Ebendal T,Olsson T, <strong>and</strong> Seckl JR. Environmental influences on the centralnervous system <strong>and</strong> their implications for the aging rat. BehavBrain Res 57: 183–191, 1993.371. Monje ML, Mizumatsu S, Fike JR, <strong>and</strong> Palmer TD. Irradiationinduces neural precursor-cell dysfunction. Nat Med 8: 955–962,2002.372. Monje ML, Toda H, <strong>and</strong> Palmer TD. Inflamma<strong>to</strong>ry blockaderes<strong>to</strong>res adult hippocampal neurogenesis. Science 302: 1760–1765,2003.373. Montaron MF, Petry KG, Rodriguez JJ, Marinelli M, AurousseauC, Rougon G, Le Moal M, <strong>and</strong> Abrous DN. Adrenalec<strong>to</strong>myincreases neurogenesis but not PSA-NCAM expression in ageddentate gyrus. Eur J Neurosci 11: 1479–1485, 1999.374. Montaron MF, Piazza PV, Aurousseau C, Urani A, Le Moal M,<strong>and</strong> Abrous DN. Implication of corticosteroid recep<strong>to</strong>rs in theregulation of hippocampal structural plasticity. Eur J Neurosci 18:3105–3111, 2003.375. Moore GJ, Bebchuk JM, Hasanat K, Chen G, Seraji-BozorgzadN, Wilds IB, Faulk MW, Koch S, Glitz DA, Jolkovsky L,<strong>and</strong> Manji HK. Lithium increases N-acetyl-aspartate in the humanbrain: in vivo evidence in support of bcl-2’s neurotrophic effects?Biol Psychiatry 48: 1–8, 2000.376. Moore GJ, Bebchuk JM, Wilds IB, Chen G, Manji HK, <strong>and</strong>Menji HK. Lithium-induced increase in human brain grey matter.Lancet 356: 1241–1242, 2000.377. Moreno-Lopez B, Noval JA, Gonzalez-Bonet LG, <strong>and</strong> EstradaC. Morphological bases for a role of nitric oxide in adult neurogenesis.Brain Res 869: 244–250, 2000.378. Morris RG, Garrud P, Rawlins JN, <strong>and</strong> O’Keefe J. Place navigationimpaired in rats with hippocampal lesions. Nature 297:681–683, 1982.379. Morrison SJ. Neuronal potential <strong>and</strong> lineage determination byneural stem cells. Curr Opin Cell Biol 13: 666–672, 2001.380. Morshead CM, Garcia AD, Sofroniew MV, <strong>and</strong> van der Kooy D.The ablation of glial fibrillary acidic protein-positive cells from theadult central nervous system results in the loss of forebrain neuralstem cells but not retinal stem cells. Eur J Neurosci 18: 76–84,2003.381. Morshead CM, Reynolds BA, Craig CG, McBurney MW,Staines WA, Morassutti D, Weiss S, <strong>and</strong> van der Kooy D.Neural stem cells in the adult mammalian forebrain: a relativelyquiescent subpopulation of subependymal cells. Neuron 13: 1071–1082, 1994.382. Morshead CM <strong>and</strong> van der Kooy D. Postmi<strong>to</strong>tic death is the fateof constitutively proliferating cells in the subependymal layer ofthe adult mouse brain. J Neurosci 12: 249–256, 1992.383. Moser MB. Making more synapses: a way <strong>to</strong> s<strong>to</strong>re information?Cell Mol Life Sci 55: 593–600, 1999.384. Mouritzen Dam A. The density of neurons in the human hippocampus.Neuropathol Appl Neurobiol 5: 249–264, 1979.385. Moyer JR Jr, Deyo RA, <strong>and</strong> Disterhoft JF. Hippocampec<strong>to</strong>mydisrupts trace eye-blink conditioning in rabbits. Behav Neurosci104: 243–252, 1990.386. Mullen RJ, Buck CR, <strong>and</strong> Smith AM. NeuN, a neuronal specificnuclear protein in vertebrates. Development 116: 201–211, 1992.387. Mumby DG, Astur RS, Weisend MP, <strong>and</strong> Sutherl<strong>and</strong> RJ. Retrogradeamnesia <strong>and</strong> selective damage <strong>to</strong> the hippocampal formation:memory for places <strong>and</strong> object discriminations. Behav BrainRes 106: 97–107, 1999.388. Murray EA <strong>and</strong> Bussey TJ. Consolidation <strong>and</strong> the medial temporallobe revisited: methodological considerations. Hippocampus11: 1–7, 2001.389. Nacher J, Crespo C, <strong>and</strong> McEwen BS. Doublecortin expressionin the adult rat telencephalon. Eur J Neurosci 14: 629–644, 2001.390. Nacher J, Rosell DR, Alonso-Llosa G, <strong>and</strong> McEwen BS. NMDArecep<strong>to</strong>r antagonist treatment induces a long-lasting increase in thenumber of proliferating cells, PSA-NCAM-immunoreactive granuleneurons <strong>and</strong> radial glia in the adult rat dentate gyrus. Eur J Neurosci13: 512–520, 2001.391. Nacher J, Alonso-Llosa G, Rosell DR, <strong>and</strong> McEwen BS. NMDArecep<strong>to</strong>r antagonist treatment increases the production of newneurons in the aged rat hippocampus. Neurobiol Aging 24: 273–284,2003.392. Nacher J, Rosell DR, <strong>and</strong> McEwen BS. Widespread expressionof rat collapsin response-mediated protein 4 in the telencephalon<strong>and</strong> other areas of the adult rat central nervous system. J CompNeurol 424: 628–639, 2000.393. Nadel L <strong>and</strong> Moscovitch M. Memory consolidation, retrogradeamnesia <strong>and</strong> the hippocampal complex. Curr Opin Neurobiol 7:217–227, 1997.394. Nadel L, Pran L, Hayes SM, Gilboa A, <strong>and</strong> Moscovitch M. Therole of the hippocampal complex in long-term episodic memory.Int Cong Ser 1250: 215–234, 2003.395. Nadel L, Samsonovich A, Ryan L, <strong>and</strong> Moscovitch M. Multipletrace theory of human memory: computational, neuroimaging, <strong>and</strong>neuropsychological results. Hippocampus 10: 352–368, 2000.396. Nakagawa E, Aimi Y, Yasuhara O, Tooyama I, Shimada M,McGeer PL, <strong>and</strong> Kimura H. Enhancement of progeni<strong>to</strong>r celldivision in the dentate gyrus triggered by initial limbic seizures inrat models of epilepsy. Epilepsia 41: 10–18, 2000.397. Nakamura K, Kaneko T, Yamashita Y, Hasegawa H, Ka<strong>to</strong>h H,<strong>and</strong> Negishi M. Immunohis<strong>to</strong>chemical localization of prostagl<strong>and</strong>inEP3 recep<strong>to</strong>r in the rat nervous system. J Comp Neurol 421:543–569, 2000.398. Nakamura Y, Sakakibara S, Miyata T, Ogawa M, Shimazaki T,Weiss S, Kageyama R, <strong>and</strong> Okano H. The bHLH gene hes1 as arepressor of the neuronal commitment of CNS stem cells. J Neurosci20: 283–293, 2000.399. Naka<strong>to</strong>mi H, Kuriu T, Okabe S, Yamamo<strong>to</strong> SI, Hatano O,Kawahara N, Tamura A, Kirino T, <strong>and</strong> Nakafuku M. Regenerationof hippocampal pyramidal neurons after ischemic brain injuryby recruitment of endogenous neural progeni<strong>to</strong>rs. Cell 110:429–441, 2002.400. Nelson MD, Saykin AJ, Flashman LA, <strong>and</strong> Riordan HJ. Hippocampalvolume reduction in schizophrenia as assessed by mag-Downloaded from physrev.physiology.org on November 25, 2005Physiol Rev • VOL 85 • APRIL 2005 • www.prv.org


NEUROGENESIS IN THE ADULT BRAIN 565netic resonance imaging: a meta-analytic study. Arch Gen Psychiatry55: 433–440, 1998.401. Nestler EJ. Molecular neurobiology of addiction. Am J Addict 10:201–217, 2001.402. Nestler EJ <strong>and</strong> Aghajanian GK. Molecular <strong>and</strong> cellular basis ofaddiction. Science 278: 58–63, 1997.403. Nevins JR. E2F: a link between the Rb tumor suppressor protein<strong>and</strong> viral oncoproteins. Science 258: 424–429, 1992.404. New<strong>to</strong>n SS, Collier EF, Hunsberger J, Adams D, TerwilligerR, Selvanayagam E, <strong>and</strong> Duman RS. Gene profile of electroconvulsiveseizures: induction of neurotrophic <strong>and</strong> angiogenic fac<strong>to</strong>rs.J Neurosci 23: 10841–10851, 2003.405. Niblock MM, Brunso-Bech<strong>to</strong>ld JK, <strong>and</strong> Riddle DR. Insulin-likegrowth fac<strong>to</strong>r I stimulates dendritic growth in primary soma<strong>to</strong>sensorycortex. J Neurosci 20: 4165–4176, 2000.406. Nibuya M, Morinobu S, <strong>and</strong> Duman RS. Regulation of BDNF <strong>and</strong>trkB mRNA in rat brain by chronic electroconvulsive seizure <strong>and</strong>antidepressant drug treatments. J Neurosci 15: 7539–7547, 1995.407. Nilsson M, Perfilieva E, Johansson U, Orwar O, <strong>and</strong> ErikssonPS. Enriched environment increases neurogenesis in the adult ratdentate gyrus <strong>and</strong> improves spatial memory. J Neurobiol 39: 569–578, 1999.408. Nimchinsky EA, Sabatini BL, <strong>and</strong> Svoboda K. Structure <strong>and</strong>function of dendritic spines. Annu Rev Physiol 64: 313–353, 2002.409. Nixon K <strong>and</strong> Crews FT. Binge ethanol exposure decreases neurogenesisin the adult rat hippocampus. J Neurochem 83: 1087–1093, 2002.410. Nottebohm F. <strong>From</strong> bird song <strong>to</strong> neurogenesis. Sci Am 260: 74–79,1989.411. Nowakowski RS <strong>and</strong> Hayes NL. New neurons: extraordinaryevidence or extraordinary conclusion? Science 288: 771, 2000.412. Nowakowski RS, Lewin SB, <strong>and</strong> Miller MW. Bromodeoxyuridineimmunohis<strong>to</strong>chemical determination of the lengths of the cellcycle <strong>and</strong> the DNA-synthetic phase for an ana<strong>to</strong>mically definedpopulation. J Neurocy<strong>to</strong>l 18: 311–318, 1989.413. Ohtsuka T, Sakamo<strong>to</strong> M, Guillemot F, <strong>and</strong> Kageyama R. Rolesof the basic helix-loop-helix genes Hes1 <strong>and</strong> Hes5 in expansion ofneural stem cells of the developing brain. J Biol Chem 276: 30467–30474, 2001.414. Okano HJ <strong>and</strong> Darnell RB. A hierarchy of Hu RNA bindingproteins in developing <strong>and</strong> adult neurons. J Neurosci 17: 3024–3037, 1997.415. Okano HJ, Pfaff DW, <strong>and</strong> Gibbs RB. RB <strong>and</strong> Cdc2 expression inbrain: correlations with 3 H-thymidine incorporation <strong>and</strong> neurogenesis.J Neurosci 13: 2930–2938, 1993.416. Okano HJ, Pfaff DW, <strong>and</strong> Gibbs RB. Expression of EGFR-,p75NGFR-, <strong>and</strong> PSTAIR (cdc2)-like immunoreactivity by proliferatingcells in the adult rat hippocampal formation <strong>and</strong> forebrain.Dev Neurosci 18: 199–209, 1996.417. Okuyama N, Takagi N, Kawai T, Miyake-Takagi K, <strong>and</strong> TakeoS. Phosphorylation of extracellular-regulating kinase in NMDArecep<strong>to</strong>r antagonist-induced newly generated neurons in the adultrat dentate gyrus. J Neurochem 88: 717–725, 2004.418. Ono K, Tomasiewicz H, Magnuson T, <strong>and</strong> Rutishauser U. N-CAM mutation inhibits tangential neuronal migration <strong>and</strong> is phenocopiedby enzymatic removal of polysialic acid. Neuron 13:595–609, 1994.419. Oppenheim RW. Cell death during development of the nervoussystem. Annu Rev Neurosci 14: 453–501, 1991.420. Ormerod BK, Falconer EM, <strong>and</strong> Galea LA. N-methyl-D-aspartaterecep<strong>to</strong>r activity <strong>and</strong> estradiol: separate regulation of cell proliferationin the dentate gyrus of adult female meadow vole. J Endocrinol179: 155–163, 2003.421. Ormerod BK <strong>and</strong> Galea LAM. Reproductive status influences cellproliferation <strong>and</strong> cell survival in the dentate gyrus of adult femalemeadow voles: a possible regula<strong>to</strong>ry role for estradiol. Neuroscience102: 369–379, 2001.422. Ormerod BK, Lee TT, <strong>and</strong> Galea LA. Estradiol initially enhancesbut subsequently suppresses (via adrenal steroids) granule cellproliferation in the dentate gyrus of adult female rats. J Neurobiol55: 247–260, 2003.423. Oumesmar BN, Vignais L, Duhamel-Clerin E, Avellana-AdalidV, Rougon G, <strong>and</strong> Baron-Van Evercooren A. Expression of thehighly polysialylated neural cell adhesion molecule during postnatalmyelination <strong>and</strong> following chemically induced demyelination ofthe adult mouse spinal cord. Eur J Neurosci 7: 480–491, 1995.424. Palmer TD, Willhoite AR, <strong>and</strong> Gage FH. Vascular niche for adulthippocampal neurogenesis. J Comp Neurol 425: 479–494, 2000.425. Parent JM, Janumpalli S, McNamara JO, <strong>and</strong> Lowenstein DH.Increased dentate granule cell neurogenesis following amygdalakindling in the adult rat. Neurosci Lett 247: 9–12, 1998.426. Parent JM <strong>and</strong> Lowenstein DH. Mossy fiber reorganization in theepileptic hippocampus. Curr Opin Neurol 10: 103–109, 1997.427. Parent JM, Tada E, Fike JR, <strong>and</strong> Lowenstein DH. Inhibition ofdentate granule cell neurogenesis with brain irradiation does notprevent seizure-induced mossy fiber synaptic reorganization in therat. J Neurosci 19: 4508–4519, 1999.428. Parent JM, Valentin VV, <strong>and</strong> Lowenstein DH. Prolonged seizuresincrease proliferating neuroblasts in the adult rat subventricularzone-olfac<strong>to</strong>ry bulb pathway. J Neurosci 22: 3174–3188,2002.429. Parent JM, Vexler ZS, Gong C, Derugin N, <strong>and</strong> Ferriero DM.Rat forebrain neurogenesis <strong>and</strong> striatal neuron replacement afterfocal stroke. Ann Neurol 52: 802–813, 2002.430. Parent JM, Yu TW, Leibowitz RT, Geschwind DH, Sloviter RS,<strong>and</strong> Lowenstein DH. Dentate granule cell neurogenesis is increasedby seizures <strong>and</strong> contributes <strong>to</strong> aberrant network reorganizationin the adult rat hippocampus. J Neurosci 17: 3727–3738,1997.431. Park HJ, Lim S, Lee HS, Lee HJ, Yoo YM, Lee HJ, Kim SA, YinCS, Seo JC, <strong>and</strong> Chung JH. Acupuncture enhances cell proliferationin dentate gyrus of maternally-separated rats. Neurosci Lett319: 153–156, 2002.432. Paula-Barbosa MM, Br<strong>and</strong>ao F, Madeira MD, <strong>and</strong> Cadete-Leite A. Structural changes in the hippocampal formation afterlong-term alcohol consumption <strong>and</strong> withdrawal in the rat. Addiction88: 237–247, 1993.433. Peissner W, Kocher M, Treuer H, <strong>and</strong> Gillardon F. Ionizingradiation-induced apop<strong>to</strong>sis of proliferating stem cells in the dentategyrus of the adult rat hippocampus. Mol Brain Res 71: 61–68,1999.434. Pencea V, Bingaman KD, Freedman LJ, <strong>and</strong> Luskin MB. <strong>Neurogenesis</strong>in the subventricular zone <strong>and</strong> rostral migra<strong>to</strong>ry streamof the neonatal <strong>and</strong> adult primate forebrain. Exp Neurol 172: 1–16,2001.435. Perez-Martin M, Azcoitia I, Trejo JL, Sierra A, <strong>and</strong> Garcia-Segura LM. An antagonist of estrogen recep<strong>to</strong>rs blocks the inductionof adult neurogenesis by insulin-like growth fac<strong>to</strong>r-I in thedentate gyrus of adult female rat. Eur J Neurosci 18: 923–930, 2003.436. Perfilieva E, Risedal A, Nyberg J, Johansson BB, <strong>and</strong> ErikssonPS. Gender <strong>and</strong> strain influence on neurogenesis in dentategyrus of young rats. J Cereb Blood Flow Metab 21: 211–217, 2001.437. Persson AI, Thorlin T, Bull C, Zarnegar P, Ekman R, TereniusL, <strong>and</strong> Eriksson PS. Mu- <strong>and</strong> delta-opioid recep<strong>to</strong>r antagonistsdecrease proliferation <strong>and</strong> increase neurogenesis in cultures of ratadult hippocampal progeni<strong>to</strong>rs. Eur J Neurosci 17: 1159–1172,2003.438. Pestell RG, Albanese C, Reutens AT, Segall JE, Lee RJ, <strong>and</strong>Arnold A. The cyclins <strong>and</strong> cyclin-dependent kinase inhibi<strong>to</strong>rs inhormonal regulation of proliferation <strong>and</strong> differentiation. EndocrRev 20: 501–534, 1999.439. Petreanu L <strong>and</strong> Alvarez-Buylla A. Maturation <strong>and</strong> death of adultbornolfac<strong>to</strong>ry bulb granule neurons: role of olfaction. J Neurosci22: 6106–6113, 2002.440. Pevny L <strong>and</strong> Rao MS. The stem-cell menagerie. Trends Neurosci26: 351–359, 2003.441. Pham K, Nacher J, Hof PR, <strong>and</strong> McEwen BS. Repeated restraintstress suppresses neurogenesis <strong>and</strong> induces biphasic PSA-NCAMexpression in the adult rat dentate gyrus. Eur J Neurosci 17:879–886, 2003.442. Pham TM, Ickes B, Albeck D, Soderstrom S, Granholm AC,<strong>and</strong> Mohammed AH. Changes in brain nerve growth fac<strong>to</strong>r levels<strong>and</strong> nerve growth fac<strong>to</strong>r recep<strong>to</strong>rs in rats exposed <strong>to</strong> environmentalenrichment for one year. Neuroscience 94: 279–286, 1999.Downloaded from physrev.physiology.org on November 25, 2005Physiol Rev • VOL 85 • APRIL 2005 • www.prv.org


566 ABROUS, KOEHL, AND LE MOAL443. Piazza PV, Deminiere JM, Le Moal M, <strong>and</strong> Simon H. Fac<strong>to</strong>rsthat predict individual vulnerability <strong>to</strong> amphetamine self-administration.Science 245: 1511–1513, 1989.444. Piazza PV, Deroche V, Deminiere JM, Maccari S, Le Moal M,<strong>and</strong> Simon H. Corticosterone in the range of stress-induced levelspossesses reinforcing properties: implications for sensation-seekingbehaviors. Proc Natl Acad Sci USA 90: 11738–11742, 1993.445. Piazza PV, Deroche-Gamonent V, Rouge-Pont F, <strong>and</strong> Le MoalM. Vertical shifts in self-administration dose-response functionspredict a drug-vulnerable phenotype predisposed <strong>to</strong> addiction.J Neurosci 20: 4226–4232, 2000.446. Piazza PV, Maccari S, Deminiere JM, Le Moal M, Mormede P,<strong>and</strong> Simon H. Corticosterone levels determine individual vulnerability<strong>to</strong> amphetamine self-administration. Proc Natl Acad Sci USA88: 2088–2092, 1991.447. Piazza PV, Mittleman G, Deminiere JM, Le Moal M, <strong>and</strong> SimonH. Relationship between schedule-induced polydipsia <strong>and</strong> amphetamineintravenous self-administration. Individual differences <strong>and</strong>role of experience. Behav Brain Res 55: 185–193, 1993.448. Privat A <strong>and</strong> Leblond CP. The subependymal layer <strong>and</strong> neighboringregion in the brain of the young rat. J Comp Neurol 146:277–302, 1972.449. Pu L, Bao GB, Xu NJ, Ma L, <strong>and</strong> Pei G. Hippocampal long-termpotentiation is reduced by chronic opiate treatment <strong>and</strong> can beres<strong>to</strong>red by re-exposure <strong>to</strong> opiates. J Neurosci 22: 1914–1921, 2002.450. Quartz SR <strong>and</strong> Sejnowski TJ. The neural basis of cognitivedevelopment: a constructivist manifes<strong>to</strong>. Behav Brain Sci 20: 537–556, 1997.451. Radley JJ <strong>and</strong> Jacobs BL. 5-HT 1A recep<strong>to</strong>r antagonist administrationdecreases cell proliferation in the dentate gyrus. Brain Res955: 264–267, 2002.452. Rakic P. Limits of neurogenesis in primates. Science 227: 1054–1056, 1985.453. Rakic P. <strong>Adult</strong> neurogenesis in mammals: an identity crisis. J Neurosci22: 614–618, 2002.454. Rakic P. <strong>Neurogenesis</strong> in adult primate neocortex: an evaluation ofthe evidence. Nat Rev Neurosci 3: 65–71, 2002.455. Ramirez-Amaya V, Balderas I, S<strong>and</strong>oval J, Escobar ML, <strong>and</strong>Bermudez-Rat<strong>to</strong>ni F. Spatial long-term memory is related <strong>to</strong>mossy fiber synap<strong>to</strong>genesis. J Neurosci 21: 7340–7348, 2001.456. Rao MS <strong>and</strong> Shetty AK. Efficacy of doublecortin as a marker <strong>to</strong>analyse the absolute number <strong>and</strong> dendritic growth of newly generatedneurons in the adult dentate gyrus. Eur J Neurosci 19: 234–246, 2004.457. Rapp PR <strong>and</strong> Amaral DG. Individual differences in the cognitive<strong>and</strong> neurobiological consequences of normal aging. Trends Neurosci15: 340–345, 1992.458. Rapp PR <strong>and</strong> Gallagher M. Preserved neuron number in thehippocampus of aged rats with spatial learning deficits. Proc NatlAcad Sci USA 93: 9926–9930, 1996.459. Rice AC, Khaldi A, Harvey HB, Salman NJ, White F, FillmoreH, <strong>and</strong> Bullock MR. Proliferation <strong>and</strong> neuronal differentiation ofmi<strong>to</strong>tically active cells following traumatic brain injury. Exp Neurol183: 406–417, 2003.460. Riddle DR, Sonntag WE, <strong>and</strong> Lichtenwalner RJ. Microvascularplasticity in aging. Ageing Res Rev 2: 149–168, 2003.461. Riley JN <strong>and</strong> Walker DW. Morphological alterations in hippocampusafter long-term alcohol consumption in mice. Science 201:646–648, 1978.462. Robbins TW <strong>and</strong> Everitt BJ. Limbic-striatal memory systems <strong>and</strong>drug addiction. Neurobiol Learn Memory 78: 625–636, 2002.463. Robinson TE <strong>and</strong> Kolb B. Persistent structural modifications innucleus accumbens <strong>and</strong> prefrontal cortex neurons produced byprevious experience with amphetamine. J Neurosci 17: 8491–8497,1997.464. Rochefort C, Gheusi G, Vincent JD, <strong>and</strong> Lledo PM. Enrichedodor exposure increases the number of newborn neurons in theadult olfac<strong>to</strong>ry bulb <strong>and</strong> improves odor memory. J Neurosci 22:2679–2689, 2002.465. Rodriguez JJ, Montaron MF, Petry KG, Aurousseau C,Marinelli M, Premier S, Rougon G, Le Moal M, <strong>and</strong> AbrousDN. Complex regulation of the expression of the polysialylatedform of the neuronal cell adhesion molecule by glucocorticoids inthe rat hippocampus. Eur J Neurosci 10: 2994–3006, 1998.466. Rogers J, Kirby LC, Hempelman SR, Berry DL, McGeer PL,Kaszniak AW, Zalinski J, Cofield M, Mansukhani L, <strong>and</strong> WillsonP. Clinical trial of indomethacin in Alzheimer’s disease. Neurology43: 1609–1611, 1993.467. Rogister B, Ben Hur T, <strong>and</strong> Dubois-Dalcq M. <strong>From</strong> neural stemcells <strong>to</strong> myelinating oligodendrocytes. Mol Cell Neurosci 14: 287–300, 1999.468. Rolls ET. A theory of hippocampal function in memory. Hippocampus6: 601–620, 1996.469. Rosenbaum RS, Winocur G, <strong>and</strong> Moscovitch M. New views onold memories: re-evaluating the role of the hippocampal complex.Behav Brain Res 127: 183–197, 2001.470. Rosenzweig MR. Effects of environmental complexity <strong>and</strong> trainingon brain chemistry <strong>and</strong> ana<strong>to</strong>my: a replication <strong>and</strong> extension.J Comp Physiol Psychol 55: 429–437, 1962.471. Rosenzweig MR <strong>and</strong> Bennett E. Experimental Influences onBrain Ana<strong>to</strong>my <strong>and</strong> Brain Chemistry in Rodents. New York:Academic, 1977.472. Rosenzweig MR <strong>and</strong> Bennett EL. Psychobiology of plasticity:effects of training <strong>and</strong> experience on brain <strong>and</strong> behaviour. BehavBrain Res 78: 57–65, 1996.473. Ross SE, Greenberg ME, <strong>and</strong> Stiles CD. Basic helix-loop-helixfac<strong>to</strong>rs in cortical development. Neuron 39: 13–25, 2003.474. Rousselot P, Lois C, <strong>and</strong> Alvarez-Buylla A. Embryonic (PSA)N-CAM reveals chains of migrating neuroblasts between the lateralventricle <strong>and</strong> the olfac<strong>to</strong>ry bulb of adult mice. J Comp Neurol 351:51–61, 1995.475. Roy NS, Wang S, Jiang L, Kang J, Benraiss A, Harrison-Restelli C, Fraser RA, Couldwell WT, Kawaguchi A, Okano H,Nedergaard M, <strong>and</strong> Goldman SA. In vitro neurogenesis by progeni<strong>to</strong>rcells isolated from the adult human hippocampus. Nat Med6: 271–277, 2000.476. Rueda D, Navarros B, martinez-Serrano A, Guzman M, <strong>and</strong>Gave-Roperh I. The endocannabinoid an<strong>and</strong>amide inhibits neuronalprogeni<strong>to</strong>r cell differentiation through attenuation of the Rap1/B-Raf/ERK pathway. J Biol Chem 277: 46645–46650, 2002.477. Sabbieti MG, Marchetti L, Abreu C, Montero A, H<strong>and</strong> AR,Raisz LG, <strong>and</strong> Hurley MM. Prostagl<strong>and</strong>ins regulate the expressionof fibroblast growth fac<strong>to</strong>r-2 in bone. Endocrinology 140:434–444, 1999.478. Saghatelyan A, De Chevigny A, Schachner M, <strong>and</strong> Lledo PM.Tenascin-R mediates activity-dependent recruitment of neuroblastsin the adult mouse forebrain. Nat Neurosci 7: 347–356, 2004.479. Sakakibara S, Imai T, Hamaguchi K, Okabe M, Aruga J, NakajimaK, Yasu<strong>to</strong>mi D, Nagata T, Kurihara Y, Uesugi S, MiyataT, Ogawa M, Mikoshiba K, <strong>and</strong> Okano H. Mouse-Musashi-1, aneural RNA-binding protein highly enriched in the mammalian CNSstem cell. Dev Biol 176: 230–242, 1996.480. Sakakibara SI, Nakamura Y, Sa<strong>to</strong>h H, <strong>and</strong> Okano H. RNAbindingprotein musashi2: developmentally regulated expression inneural precursor cells <strong>and</strong> subpopulations of neurons in mammalianCNS. J Neurosci 21: 8091–8107, 2001.481. Sakamo<strong>to</strong> M, Hirata H, Ohtsuka T, Bessho Y, <strong>and</strong> KageyamaR. The basic helix-loop-helix genes Hesr1/Hey1 <strong>and</strong> Hesr2/Hey2regulate maintenance of neural precursor cells in the brain. J BiolChem 278: 44808–44815, 2003.482. Sanchez MM, Ladd CO, <strong>and</strong> Plotsky PM. Early adverse experienceas a developmental risk fac<strong>to</strong>r for later psychopathology:evidence from rodent <strong>and</strong> primate models. Dev Psychopathol 13:419–449, 2001.483. Santarelli L, Saxe M, Gross C, Surget A, Battaglia F, DulawaS, Weisstaub N, Lee J, Duman R, Arancio O, Belzung C, <strong>and</strong>Hen R. Requirement of hippocampal neurogenesis for the behavioraleffects of antidepressants. Science 301: 805–809, 2003.484. Sapolsky RM. Stress, the Aging Brain, <strong>and</strong> the Mechanisms ofNeuron Death. Cambridge, MA: MIT Press, 1992.485. Sasaki T, Kitagawa K, Sugiura S, Omura-Matsuoka E, TanakaS, Yagita Y, Okano H, Matsumo<strong>to</strong> M, <strong>and</strong> Hori M. Implication ofcyclooxygenase-2 on enhanced proliferation of neural progeni<strong>to</strong>rcells in the adult mouse hippocampus after ischemia. J NeurosciRes 72: 461–471, 2003.Downloaded from physrev.physiology.org on November 25, 2005Physiol Rev • VOL 85 • APRIL 2005 • www.prv.org


NEUROGENESIS IN THE ADULT BRAIN 567486. Scemes E. Components of astrocytic intercellular calcium signaling.Mol Neurobiol 22: 167–179, 2000.487. Scharfman HE, Goodman JH, <strong>and</strong> Sollas AL. Granule-like neuronsat the hilar/CA3 border after status epilepticus <strong>and</strong> theirsynchrony with area CA3 pyramidal cells: functional implicationsof seizure-induced neurogenesis. J Neurosci 20: 6144–6158, 2000.488. Schmajuk NA. Hippocampal dysfunction in schizophrenia. Hippocampus11: 599–613, 2001.489. Schmidt W <strong>and</strong> Reymann KG. Proliferating cells differentiate in<strong>to</strong>neurons in the hippocampal CA1 region of gerbils after globalcerebral ischemia. Neurosci Lett 334: 153–156, 2002.490. Schmidt-Hieber C, Jonas P, <strong>and</strong> Bischofberger J. Enhancedsynaptic plasticity in newly generated granule cells of the adulthippocampus. Nature 429: 184–187, 2004.491. Schweisguth F. Notch signaling activity. Curr Biol 14: R129–R138,2004.492. Scott BW, Wang S, Burnham WM, De Boni U, <strong>and</strong> Woj<strong>to</strong>wiczJM. Kindling-induced neurogenesis in the dentate gyrus of the rat.Neurosci Lett 248: 73–76, 1998.493. Seaberg RM <strong>and</strong> van der Kooy D. <strong>Adult</strong> rodent neurogenicregions: the ventricular subependyma contains neural stem cells,but the dentate gyrus contains restricted progeni<strong>to</strong>rs. J Neurosci22: 1784–1793, 2002.494. Seaberg RM <strong>and</strong> van der Kooy D. Stem <strong>and</strong> progeni<strong>to</strong>r cells: thepremature desertion of rigorous definitions. Trends Neurosci 26:125–131, 2003.495. Sekerkova G, Ilijic E, <strong>and</strong> Mugnaini E. Bromodeoxyuridineadministered during neurogenesis of the projection neurons causescerebellar defects in rat. J Comp Neurol 470: 221–239, 2004.496. Seki T. Expression patterns of immature neuronal markers PSA-NCAM, CRMP-4 <strong>and</strong> NeuroD in the hippocampus of young adult<strong>and</strong> aged rodents. J Neurosci Res 70: 327–334, 2002.497. Seki T. Hippocampal adult neurogenesis occurs in a microenvironmentprovided by PSA-NCAM-expressing immature neurons.J Neurosci Res 69: 772–783, 2002.498. Seki T <strong>and</strong> Arai Y. Highly polysialylated neural cell adhesionmolecule (NCAM-H) is expressed by newly generated granule cellsin the dentate gyrus of the adult rat. J Neurosci 13: 2351–2358, 1993.499. Seki T <strong>and</strong> Arai Y. Age-related production of new granule cells inthe adult dentate gyrus. Neuroreport 6: 2479–2482, 1995.500. Seri B <strong>and</strong> Alvarez-Buylla A. Neural stem cells <strong>and</strong> the regulationof neurogenesis in the adult hippocampus. Clin Neurosci Res2: 11–16, 2002.501. Seri B, Garcia-Verdugo JM, McEwen BS, <strong>and</strong> Alvarez-BuyllaA. Astrocytes give rise <strong>to</strong> new neurons in the adult mammalianhippocampus. J Neurosci 21: 7153–7160, 2001.502. Seroogy KB, Gall CM, Lee DC, <strong>and</strong> Kornblum HI. Proliferativezones of postnatal rat brain express epidermal growth fac<strong>to</strong>r recep<strong>to</strong>rmRNA. Brain Res 670: 157–164, 1995.503. Shastri L. Episodic memory <strong>and</strong> cortico-hippocampal interactions.Trends Cogn Sci 6: 162–168, 2002.504. Sheline YI, Sanghavi M, Mintun MA, <strong>and</strong> Gado MH. Depressionduration but not age predicts hippocampal volume loss in medicallyhealthy women with recurrent major depression. J Neurosci19: 5034–5043, 1999.505. Shen Q, Goderie S, Jin L, Karanth N, Sun Y, Abramova N,Vincent P, Pumiglia K, <strong>and</strong> Temple S. Endothelial cells stimulateself-renewal <strong>and</strong> exp<strong>and</strong> neurogenesis of neural stem cells. Science.In press.506. Shingo T, Gregg C, Enwere E, Fujikawa H, Hassam R, GearyC, Cross JC, <strong>and</strong> Weiss S. Pregnancy-stimulated neurogenesis inthe adult female forebrain mediated by prolactin. Science 299:117–120, 2003.507. Shingo T, Sorokan ST, <strong>and</strong> Shimazaki TWS. Erythropoietinregulates the in vitro <strong>and</strong> in vivo production of neuronal progeni<strong>to</strong>rsby mammalian forebrain neural stem cells. J Neurosci 21:9733–9743, 2001.508. Shors TJ. Memory traces of trace memories: neurogenesis, synap<strong>to</strong>genesis<strong>and</strong> awareness. Trends Neurosci 27: 250–256, 2004.509. Shors TJ, Miesegaes G, Beylin A, Zhao M, Rydel T, <strong>and</strong> GouldE. <strong>Neurogenesis</strong> in the adult is involved in the formation of tracememories. Nature 410: 372–376, 2001.510. Shors TJ, Townsend DA, Zhao M, Kozorovitskiy Y, <strong>and</strong> GouldE. <strong>Neurogenesis</strong> may relate <strong>to</strong> some but not all types of hippocampal-dependentlearning. Hippocampus 12: 578–584, 2002.511. Smart I. The subependymal layer of the mouse brain <strong>and</strong> its cellproduction as shown by radioau<strong>to</strong>graphy after thymidine-H 3 injection.J Comp Neurol 116: 325–347, 1961.512. Solomon PR, V<strong>and</strong>er Schaaf ER, Thompson RF, <strong>and</strong> Weisz DJ.Hippocampus <strong>and</strong> trace conditioning of the rabbit’s classicallyconditioned nictitating membrane response. Behav Neurosci 100:729–744, 1986.513. Song H, Stevens CF, <strong>and</strong> Gage FH. Neural stem cells from adulthippocampus develop essential properties of functional CNS neurons.Nat Neurosci 5: 438–445, 2002.514. Song H, Stevens CF, <strong>and</strong> Gage FH. Astroglia induce neurogenesisfrom adult neural stem cells. Nature 417: 39–44, 2002.515. Sousa N <strong>and</strong> Almeida OFX. Corticosteroids: sculp<strong>to</strong>rs of thehippocampal formation. Rev Neurosci 13: 59–84, 2002.516. Spranger M, Lindholm D, B<strong>and</strong>tlow C, Heumann R, Gnahn H,Njer-No M, <strong>and</strong> Thonen H. Regulation of nerve growth fac<strong>to</strong>r(NGF) synthesis in the rat central nervous system: comparisonbetween the effects of interleukin-1 <strong>and</strong> various growth fac<strong>to</strong>rs inastrocyte culture <strong>and</strong> in vivo. Eur J Neurosci 2: 69–76, 1990.517. Stanfield BB <strong>and</strong> Trice JE. Evidence that granule cells generatedin the dentate gyrus of adult rats extend axonal projections. ExpBrain Res 72: 399–406, 1988.518. Starkman MN, Giordani B, Gebarski SS, Berent S, SchorkMA, <strong>and</strong> Schteingart DE. Decrease in cortisol reverses humanhippocampal atrophy following treatment of Cushing’s disease.Biol Psychiatry 46: 1595–1602, 1999.519. Steffens DC, Byrum CE, McQuoid DR, Greenberg DL, PayneME, Blitching<strong>to</strong>n TF, MacFall JR, <strong>and</strong> Krishnan KR. Hippocampalvolume in geriatric depression. Biol Psychiatry 48: 301–309, 2000.520. Stein-Behrens BA, Lin WJ, <strong>and</strong> Sapolsky RM. Physiologicalelevations of glucocorticoids potentiate glutamate accumulation inthe hippocampus. J Neurochem 63: 596–602, 1994.521. Steiner B, Kronenberg G, Jessberger S, Br<strong>and</strong>t MD, Reuter K,<strong>and</strong> Kempermann G. Differential regulation of gliogenesis in thecontext of adult hippocampal neurogenesis in mice. Glia 46: 41–52,2004.522. Stenvers KL, Lund PK, <strong>and</strong> Gallagher M. Increased expressionof type 1 insulin-like growth fac<strong>to</strong>r recep<strong>to</strong>r messenger RNA in rathippocampal formation is associated with aging <strong>and</strong> behavioralimpairment. Neuroscience 72: 505–518, 1996.523. Stump G, Durrer A, Klein AL, Lu<strong>to</strong>lf S, Suter U, <strong>and</strong> Taylor V.Notch1 <strong>and</strong> its lig<strong>and</strong>s Delta-like <strong>and</strong> Jagged are expressed <strong>and</strong>active in distinct cell populations in the postnatal mouse brain.Mech Dev 114: 153–159, 2002.524. Sullivan EV, Marsh L, Mathalon DH, Lim KO, <strong>and</strong> PfefferbaumA. Anterior hippocampal volume deficits in nonamnesic,aging chronic alcoholics. Alcohol Clin Exp Res 19: 110–122, 1995.525. Sun W <strong>and</strong> Rebec GV. Lidocaine inactivation of ventral subiculumattenuates cocaine-seeking behavior in rats. J Neurosci 23: 10258–10264, 2003.526. Sun Y, Jin K, Xie L, Childs J, Mao XO, Logvinova A, <strong>and</strong>Greenberg DA. VEGF-induced neuroprotection, neurogenesis,<strong>and</strong> angiogenesis after focal cerebral ischemia. J Clin Invest 111:1843–1851, 2003.527. Sutherl<strong>and</strong> RJ, Weisend MP, Mumby D, Astur RS, Hanlon FM,Koerner A, Thomas MJ, Wu Y, Moses SN, Cole C, Hamil<strong>to</strong>nDA, <strong>and</strong> Hoesing JM. Retrograde amnesia after hippocampaldamage: recent vs. remote memories in two tasks. Hippocampus11: 27–42, 2001.528. Su<strong>to</strong> N, Austin JD, <strong>and</strong> Vezina P. Locomo<strong>to</strong>r response <strong>to</strong> noveltypredicts a rat’s propensity <strong>to</strong> self-administer nicotine. Psychopharmacologia158: 175–180, 2001.529. Sutula T, Cascino G, Cavazos J, Parada I, <strong>and</strong> Ramirez L.Mossy fiber synaptic reorganization in the epileptic human temporallobe. Ann Neurol 26: 321–330, 1989.530. Sutula T, Lauersdorf S, Lynch M, Jurgella C, <strong>and</strong> Woodard A.Deficits in radial arm maze performance in kindled rats: evidencefor long-lasting memory dysfunction induced by repeated briefseizures. J Neurosci 15: 8295–8301, 2003.Downloaded from physrev.physiology.org on November 25, 2005Physiol Rev • VOL 85 • APRIL 2005 • www.prv.org


568 ABROUS, KOEHL, AND LE MOAL531. Szele FG <strong>and</strong> Chesselet MF. Cortical lesions induce an increasein cell number <strong>and</strong> PSA-NCAM expression in the subventricularzone of adult rats. J Comp Neurol 368: 439–454, 1996.532. Tada E, Parent JM, Lowenstein DH, <strong>and</strong> Fike JR. X-irradiationcauses a prolonged reduction in cell proliferation in the dentategyrus of adult rats. Neuroscience 99: 33–41, 2000.533. Taepavarapruk P <strong>and</strong> Phillips AG. Neurochemical correlates ofrelapse <strong>to</strong> D-amphetamine self-administration by rats induced bystimulation of the ventral subiculum. Psychopharmacology 168:99–108, 2003.534. Takasawa K, Kitagawa K, Yagita Y, Sasaki T, Tanaka S, MatsushitaK, Ohstuki T, Miyata T, Okano H, Hori M, <strong>and</strong> Matsumo<strong>to</strong>M. Increased proliferation of neural progeni<strong>to</strong>r cells butreduced survival of newborn cells in the contralateral hippocampusafter focal cerebral ischemia in rats. J Cereb Blood Flow Metab22: 299–307, 2002.535. Tan SE. Impairing the amphetamine conditioning in rats throughthe inhibition of hippocampal calcium/calmodulin-dependent proteinkinase II activity. Neuropharmacology 42: 540–547, 2002.536. Tanapat P, Hastings NB, Reeves AJ, <strong>and</strong> Gould E. Estrogenstimulates a transient increase in the number of new neurons in thedentate gyrus of the adult female rat. J Neurosci 19: 5792–5801,1999.537. Tanapat P, Hastings NB, Rydel TA, Galea LA, <strong>and</strong> Gould E.Exposure <strong>to</strong> fox odor inhibits cell proliferation in the hippocampusof adult rats via an adrenal hormone-dependent mechanism.J Comp Neurol 437: 496–504, 2001.538. Teyler TJ <strong>and</strong> DiScenna P. The hippocampal memory indexingtheory. Behav Neurosci 100: 147–154, 1986.539. Tha KK, Okuma Y, Miyazaki H, Murayama T, Uehara T,Hatakeyama R, Hayashi Y, <strong>and</strong> Nomura Y. Changes in expressionsof proinflamma<strong>to</strong>ry cy<strong>to</strong>kines IL-1beta, TNF-alpha <strong>and</strong> IL-6 inthe brain of senescence accelerated mouse (SAM) P8. Brain Res885: 25–31, 2000.540. Tomasiewicz H, Ono K, Yee D, Thompson C, Goridis C, RutishauserU, <strong>and</strong> Magnuson T. Genetic deletion of a neural celladhesion molecule variant (N-CAM-180) produces distinct defectsin the central nervous system. Neuron 11: 1163–1174, 1993.541. Trachtenberg JT, Chen BE, Knott GW, Feng G, Sanes JR,Welker E, <strong>and</strong> Svoboda K. Long-term in vivo imaging of experience-dependentsynaptic plasticity in adult cortex. Nature 420:788–794, 2002.542. Trejo JL, Carro E, <strong>and</strong> Torres-Aleman I. Circulating insulin-likegrowth fac<strong>to</strong>r I mediates exercise-induced increases in the numberof new neurons in the adult hippocampus. J Neurosci 21: 1628–1634, 2001.543. Tropepe V, Craig CG, Morshead CM, <strong>and</strong> van der Kooy D.Transforming growth fac<strong>to</strong>r-alpha null <strong>and</strong> senescent mice showdecreased neural progeni<strong>to</strong>r cell proliferation in the forebrain subependyma.J Neurosci 17: 7850–7859, 1997.544. Tsai G <strong>and</strong> Coyle JT. N-aspartate in neuropsychiatric disorders.Prog Neurobiol 46: 531–540, 1995.545. Tzeng SF. Inhibi<strong>to</strong>rs of DNA binding in neural cell proliferation<strong>and</strong> differentiation. Neurochem Res 28: 45–52, 2003.546. Tzeng SF <strong>and</strong> Wu JP. Responses of microglia <strong>and</strong> neural progeni<strong>to</strong>rs<strong>to</strong> mechanical brain injury. Neuroreport 10: 2287–2292, 1999.547. Uchida K, Kumihashi K, Kurosawa S, Kobayashi T, I<strong>to</strong>i K, <strong>and</strong>Machida T. Stimula<strong>to</strong>ry effects of prostagl<strong>and</strong>in E 2 on neurogenesisin the dentate gyrus of the adult rat. Zool Sci 19: 1211–1216,2002.548. Ueki T, Tanaka M, Yamashita K, Mikawa S, Qiu Z, MaragakisNJ, Hevner RF, Miura N, Sugimura H, <strong>and</strong> Sa<strong>to</strong> K. A novelsecre<strong>to</strong>ry fac<strong>to</strong>r, Neurogenesin-1, provides neurogenic environmentalcues for neural stem cells in the adult hippocampus. J Neurosci23: 11732–11740, 2003.549. Vaidya VA, Siuciak JA, Du F, <strong>and</strong> Duman RS. Hippocampalmossy fiber sprouting induced by chronic electroconvulsive seizures.Neuroscience 89: 157–166, 1999.550. Vakili K, Pillay SS, Lafer B, Fava M, Renshaw PF, Bonello-Cintron CM, <strong>and</strong> Yurgelun-Todd DA. Hippocampal volume inprimary unipolar major depression: a magnetic resonance imagingstudy. Biol Psychiatry 47: 1087–1090, 2000.551. Vallée M, Mayo W, Darnaudéry M, Corpéchot C, Young J,Koehl M, Le Moal M, Baulieu EE, Robel P, <strong>and</strong> Simon H.Neurosteroids: deficient cognitive performance in aged rats dependson low pregnenolone sulfate levels in the hippocampus. ProcNatl Acad Sci USA 94: 14865–14870, 1997.552. Vallieres L, Campbell IL, Gage FH, <strong>and</strong> Sawchenko PE. Reducedhippocampal neurogenesis in adult transgenic mice withchronic astrocytic production of interleukin-6. J Neurosci 22: 486–492, 2002.553. Van Praag H, Christie BR, Sejnowski TJ, <strong>and</strong> Gage FH. Runningenhances neurogenesis, learning, <strong>and</strong> long-term potentiationin mice. Proc Natl Acad Sci USA 96: 13427–13431, 1999.554. Van Praag H, Kempermann G, <strong>and</strong> Gage FH. Running increasescell proliferation <strong>and</strong> neurogenesis in the adult mouse dentategyrus. Nat Neurosci 2: 266–270, 1999.555. Van Praag H, Kempermann G, <strong>and</strong> Gage FH. Neural consequencesof environmental enrichment. Nat Rev Neurosci 1: 191–198, 2000.556. Van Praag H, Schinder AF, Christie BR, Toni N, Palmer TD,<strong>and</strong> Gage FH. Functional neurogenesis in the adult hippocampus.Nature 415: 1030–1034, 2002.557. Vazquez DM. Stress <strong>and</strong> the developing limbic-hypothalamic-pituitary-adrenalaxis. Psychoneuroendocrinology 23: 663–700, 1988.558. Velakoulis D, Stuart GW, Wood SJ, Smith DJ, Brewer WJ,Desmond P, Singh B, Copolov D, <strong>and</strong> Pantelis C. Selectivebilateral hippocampal volume loss in chronic schizophrenia. BiolPsychiatry 50: 531–539, 2001.559. Viau V, Sharma S, <strong>and</strong> Meaney MJ. Changes in plasma adrenocorticotropin,corticosterone, corticosteroid-binding globulin, <strong>and</strong>hippocampal glucocorticoid recep<strong>to</strong>r occupancy/translocation inrat pups in response <strong>to</strong> stress. J Neuroendocrinol 8: 1–8, 1996.560. Vitry S, Avellana-Adalid V, Hardy R, Lachapelle F, <strong>and</strong> Baron-Van Evercooren A. Mouse oligospheres: from pre-progeni<strong>to</strong>rs <strong>to</strong>functional oligodendrocytes. J Neurosci Res 58: 735–751, 1999.561. Vorel SR, Liu X, Hayes RJ, Spec<strong>to</strong>r JA, <strong>and</strong> Gardner EL.Relapse <strong>to</strong> cocaine-seeking after hippocampal theta burst stimulation.Science 292: 1175–1178, 2001.562. Wakade CG, Mahadik SP, Waller JL, <strong>and</strong> Chiu FC. Atypicalneuroleptics stimulate neurogenesis in adult rat brain. J NeurosciRes 69: 72–79, 2002.563. Walker DW, Barnes DE, Zornetzer SF, Hunter BE, <strong>and</strong> KubanisP. Neuronal loss in hippocampus induced by prolonged ethanolconsumption in rats. Science 209: 711–713, 1980.564. Wang S, Scott BW, <strong>and</strong> Woj<strong>to</strong>wicz JM. Heterogeneous propertiesof dentate granule neurons in the adult rat. J Neurobiol 42:248–257, 2000.565. Weaver JD, Huang MH, Albert M, Harris T, Rowe JW, <strong>and</strong>Seeman TE. Interleukin-6 <strong>and</strong> risk of cognitive decline: MacArthurstudies of successful aging. Neurology 59: 371–378, 2002.566. Weinberg RA. The retinoblas<strong>to</strong>ma protein <strong>and</strong> cell cycle control.Cell 81: 323–330, 1995.567. Weinmaster G. Notch signal transduction: a real rip <strong>and</strong> more.Curr Opin Genet Dev 10: 363–369, 2000.568. Weinstein DE, Burrola P, <strong>and</strong> Kilpatrick TJ. Increased proliferationof precursor cells in the adult rat brain after targetedlesioning. Brain Res 743: 11–16, 1996.569. Weins<strong>to</strong>ck M. Alterations induced by gestational stress in brainmorphology <strong>and</strong> behaviour of the offspring. Prog Neurobiol 65:427–451, 2001.570. Weiss S, Reynolds BA, Vescovi AL, Morshead C, Craig CG,<strong>and</strong> van der Kooy D. Is there a neural stem cell in the mammalianforebrain? Trends Neurosci 19: 387–393, 1996.571. White NM. Addictive drugs as reinforcers: multiple partial actionson memory systems. Addiction 91: 921–949, 1996.572. Winner B, Cooper-Kuhn CM, Aigner R, Winkler J, <strong>and</strong> KuhnHG. Long-term survival <strong>and</strong> cell death of newly generated neuronsin the adult rat olfac<strong>to</strong>ry bulb. Eur J Neurosci 16: 1681–1689, 2002.573. Wittenberg GM <strong>and</strong> Tsien JZ. An emerging molecular <strong>and</strong> cellularframework for memory processing by the hippocampus.Trends Neurosci 25: 501–505, 2002.574. Wu W, Wong K, Chen J, Jiang Z, Dupuis S, Wu JY, <strong>and</strong> Rao Y.Directional guidance of neuronal migration in the olfac<strong>to</strong>ry systemby the protein Slit. Nature 400: 331–336, 1999.Downloaded from physrev.physiology.org on November 25, 2005Physiol Rev • VOL 85 • APRIL 2005 • www.prv.org


NEUROGENESIS IN THE ADULT BRAIN 569575. Yagita Y, Kitagawa K, Ohtsuki T, Takasawa K, Miyata T,Okano H, Hori M, <strong>and</strong> Matsumo<strong>to</strong> M. <strong>Neurogenesis</strong> by progeni<strong>to</strong>rcells in the ischemic adult rat hippocampus. Stroke 32: 1890–1896, 2001.576. Yagita Y, Kitagawa K, Sasaki T, Miyata T, Okano H, Hori M,<strong>and</strong> Matsumo<strong>to</strong> M. Differential expression of Musashi1 <strong>and</strong> nestinin the adult rat hippocampus after ischemia. J Neurosci Res 69:750–756, 2002.577. Yamaguchi M, Sai<strong>to</strong> H, Suzuki M, <strong>and</strong> Mori K. Visualization ofneurogenesis in the central nervous system using nestin promoter-GFP transgenic mice. Neuroreport 11: 1991–1996, 2000.578. Yanagisawa M, Takizawa T, Ochiai W, Uemura A, NakashimaK, <strong>and</strong> Taga T. Fate alteration of neuroepithelial cells from neurogenesis<strong>to</strong> astrocy<strong>to</strong>genesis by bone morphogenetic proteins.Neurosci Res 41: 391–396, 2001.579. Yoshida K, Kakihana M, Chen LS, Ong M, Baird A, <strong>and</strong> GageFH. Cy<strong>to</strong>kine regulation of nerve growth fac<strong>to</strong>r-mediated cholinergicneurotrophic activity synthesized by astrocytes <strong>and</strong> fibroblasts.J Neurochem 59: 919–931, 1992.580. Yoshikawa K. Cell cycle regula<strong>to</strong>rs in neural stem cells <strong>and</strong> postmi<strong>to</strong>ticneurons. Neurosci Res 37: 1–14, 2000.581. Yoshimizu T <strong>and</strong> Chaki S. Increased cell proliferation in the adultmouse hippocampus following chronic administration of group IImetabotropic glutamate recep<strong>to</strong>r antagonist, MGS0039. BiochemBiophys Res Commun 315: 493–496, 2004.582. Yoshimura S, Takagi Y, Harada J, Teramo<strong>to</strong> T, Thomas SS,Waeber C, Bakowska JC, Breakefield XO, <strong>and</strong> Moskowitz MA.FGF-2 regulation of neurogenesis in adult hippocampus after braininjury. Proc Natl Acad Sci USA 98: 5874–5879, 2001.583. Young D <strong>and</strong> Dragunow M. Neuronal injury following electricallyinduced status epilepticus with <strong>and</strong> without adenosine recep<strong>to</strong>rantagonism. Exp Neurol 133: 125–137, 1995.584. Young D, Lawlor PA, Leone P, Dragunow M, <strong>and</strong> During MJ.Environmental enrichment inhibits spontaneous apop<strong>to</strong>sis, preventsseizures <strong>and</strong> is neuroprotective. Nat Med 5: 448–453, 1999.585. Yu IT, Lee SH, Lee YS, <strong>and</strong> Son H. Differential effects of corticosterone<strong>and</strong> dexamethasone on hippocampal neurogenesis invitro. Biochem Biophys Res Commun 317: 484–490, 2004.586. Yuste R <strong>and</strong> Bonhoeffer T. Morphological changes in dendriticspines associated with long-term synaptic plasticity. Annu RevNeurosci 24: 1071–1089, 2001.587. Zhang R, Zhang L, Zhang Z, Wang Y, Lu M, Lapointe M, <strong>and</strong>Chopp M. A nitric oxide donor induces neurogenesis <strong>and</strong> reducesfunctional deficits after stroke in rats. Ann Neurol 50: 602–611,2001.588. Zhang R, Zhang Z, Wang L, Wang Y, Gousev A, Zhang L, Ho KL,Morshead C, <strong>and</strong> Chopp M. Activated neural stem cells contribute<strong>to</strong> stroke-induced neurogenesis <strong>and</strong> neuroblast migration <strong>to</strong>wardthe infarct boundary in adult rats. J Cereb Blood Flow Metab24: 441–448, 2004.589. Zhang RL, Zhang ZG, Zhang L, <strong>and</strong> Chopp M. Proliferation <strong>and</strong>differentiation of progeni<strong>to</strong>r cells in the cortex <strong>and</strong> the subventricularzone in the adult rat after focal cerebral ischemia. Neuroscience105: 33–41, 2001.590. Zhu DY, Liu SH, Sun HS, <strong>and</strong> Lu YM. Expression of induciblenitric oxide synthase after focal cerebral ischemia stimulates neurogenesisin the adult rodent dentate gyrus. J Neurosci 23: 223–229,2003.591. Zhu Y, Jin K, Mao XO, <strong>and</strong> Greenberg DA. Vascular endothelialgrowth fac<strong>to</strong>r promotes proliferation of cortical neuron precursorsby regulating E2F expression. FASEB J 17: 186–193, 2003.592. Zhu Z, Jiang W, <strong>and</strong> Thompson HJ. Mechanisms by which energyrestriction inhibits rat mammary carcinogenesis: in vivo effects ofcorticosterone on cell cycle machinery in mammary carcinomas.Carcinogenesis 24: 1225–1231, 2003.593. Zigova T, Pencea V, Wieg<strong>and</strong> SJ, <strong>and</strong> Luskin MB. Intraventricularadministration of BDNF increases the number of newly generatedneurons in the adult olfac<strong>to</strong>ry bulb. Mol Cell Neurosci 11:234–245, 1998.594. Zucconi GG <strong>and</strong> Giuditta A. Is it only neurogenesis? Rev Neurosci13: 375–382, 2002.595. Zwain IH <strong>and</strong> Yen SS. Neurosteroidogenesis in astrocytes, oligodendrocytes,<strong>and</strong> neurons of cerebral cortex of rat brain. Endocrinology140: 3843–3852, 1999.Downloaded from physrev.physiology.org on November 25, 2005Physiol Rev • VOL 85 • APRIL 2005 • www.prv.org

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!