28.10.2014 Views

Bone marrow as a priming site for naīve CD8+ T cells - Weizmann ...

Bone marrow as a priming site for naīve CD8+ T cells - Weizmann ...

Bone marrow as a priming site for naīve CD8+ T cells - Weizmann ...

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.

Thesis <strong>for</strong> the degree<br />

Doctor of Philosophy<br />

Submitted to the Scientific Council of the<br />

<strong>Weizmann</strong> Institute of Science<br />

Rehovot, Israel<br />

עבודת גמר ‏(תזה)‏ לתואר<br />

דוקטור לפילוסופיה<br />

מוגשת למועצה המדעית של<br />

מכון ויצמן למדע<br />

רחובות,‏ ישראל<br />

במתכונת ‏"רגילה"‏<br />

In a "Regular" Format<br />

By<br />

Anita Sapoznikov<br />

מאת<br />

אניטה ספוז'ניקוב<br />

הבנת תפקידיהם של תאים דנדריטיים באיברים הלימפואידים בעכבר<br />

Probing In-Vivo Dendritic Cell functions in Lymphoid Organs<br />

Advisor:<br />

Dr. Steffen Jung<br />

Prof. Idit Shachar<br />

מנחה:‏<br />

דר'‏ סטפן יונג<br />

פרופ'‏ עידית שחר<br />

July 2009<br />

תמוז,‏ תשס"ט<br />

0


Acknowledgements<br />

I am grateful to my supervisors, Dr. Steffen Jung and Prof. Idit Shachar <strong>for</strong> opening<br />

the door to the wonderful world of immunology science and <strong>for</strong> sharing with me their<br />

broad knowledge and experience.<br />

I am especially grateful to Dr. Yael Pewzner-Jung, Vyacheslav Kalchenko; Dr. Guy<br />

Shakhar and Idan Milo <strong>for</strong> the fruitful discussions, professional support and creative<br />

collaboration on this study.<br />

Finally, I would also like to thank my fellow members of Steffen Jung's laboratory <strong>for</strong><br />

sharing their expertise, technical <strong>as</strong>sistance and friendship.<br />

1


List of abbreviations<br />

APCs<br />

AFCs<br />

APRIL<br />

BAFF<br />

BCMA<br />

BM<br />

bmDCs<br />

BST 2<br />

cDCs<br />

CLPs<br />

CMTMR<br />

CTLs<br />

DN<br />

DTR<br />

DTx<br />

ECFP<br />

ELISPOT<br />

GFP<br />

HEVs<br />

IFN I<br />

IPCs<br />

i.v.<br />

LCs<br />

LNs<br />

MACS<br />

MDPs<br />

MHC<br />

MIF<br />

MLR<br />

OVA<br />

PDCs<br />

Antigen Presenting Cells<br />

Antibody-Forming Cells<br />

Proliferation-Inducing Ligand<br />

B cell-Activating Factor<br />

B Cell Maturation Antigen<br />

<strong>Bone</strong> Marrow<br />

<strong>Bone</strong> Marrow-resident Dendritic Cells<br />

<strong>Bone</strong> <strong>marrow</strong> Stromal cell antigen-2<br />

conventional Dendritic Cells<br />

Common Lymphoid Precursors<br />

ChloroMethyl benzoyl amino TetraMethylRhodamine<br />

Cytotoxic T Lymphocytes<br />

Double Negative<br />

Diphtheria Toxin Receptor<br />

Diphtheria Toxin<br />

Enhanced Cyan Fluorescent Protein<br />

Enzyme-Linked Immunospot <strong>as</strong>say<br />

Green Fluorescent Protein<br />

High Endothelial Venules<br />

Interferon type I<br />

Interferon-Producing Cells<br />

Intravenously<br />

Langerhans Cells<br />

Lymph Node<br />

Magnetic Cell Sorting<br />

Macrophage/Dendritic cell Precursors<br />

Major Histocompatiblity Complex<br />

Migration-Inhibitory Factor<br />

Mixed-Leukocyte Reaction<br />

Ovalbumin<br />

Pl<strong>as</strong>macytoid Dendritic Cells<br />

2


Rag 2 Recombination-Activating gene 2<br />

s.c. Subcutaneously<br />

SiglecH Sialic acid binding Ig-like lectin H<br />

S1P1 Sphingosine 1-Phosphate receptor-1<br />

Spl MΦ Splenic Macrophages<br />

TACI Transmembrane Activator<br />

TCR T Cell Receptor<br />

TLRs Toll-Like Receptors<br />

TNF Tumor Necrosis Factor<br />

TRITC Tetramethyl Rhodamine IsoThioCyanate<br />

3


Table of content<br />

Summary 6<br />

1. Introduction 8<br />

1.1 Dendritic <strong>cells</strong> (DCs) 8<br />

1.2 <strong>Bone</strong> <strong>marrow</strong> (BM) 12<br />

1.3 Intravital microscopy 15<br />

2. Periv<strong>as</strong>cular clusters of dendritic <strong>cells</strong> provide critical<br />

survival signals to B <strong>cells</strong> in bone <strong>marrow</strong> niches 17<br />

2.1 Introduction 17<br />

2.2 Results 18<br />

2.2.1 Phenotypic and functional characterization of bone <strong>marrow</strong> DCs 18<br />

2.2.2 Organization of bmDCs into periv<strong>as</strong>cular clusters 19<br />

2.2.3 DC ablation causes depletion of recirculating B <strong>cells</strong> from BM 21<br />

2.2.4 BmDCs provide a survival signal to recirculating mature B <strong>cells</strong> 27<br />

2.2.5 Recirculating mature B <strong>cells</strong> require MIF-producing bmDCs <strong>for</strong> survival 29<br />

2.3 Discussion 35<br />

3. The bone <strong>marrow</strong> immune niche: <strong>Bone</strong> <strong>marrow</strong> <strong>as</strong> a<br />

<strong>priming</strong> <strong>site</strong> <strong>for</strong> naïve CD8 + T <strong>cells</strong> 38<br />

3.1 Introduction 38<br />

3.2 Results 39<br />

3.2.1 Identification of bmDC clusters that contain B <strong>cells</strong> in the BM of the<br />

long bones 39<br />

3.2.2 BmDCs originate without monocytic intermediate from<br />

macrophage/DC precursors (MDPs) 41<br />

3.2.3 In the steady-state T lymphocytes are more motile than B <strong>cells</strong><br />

in the bmDC niche 43<br />

3.2.4 Naïve CD8 + T <strong>cells</strong> <strong>for</strong>m multi-cell clusters in the bmDC niches<br />

upon blood-borne antigen stimulation 45<br />

3.2.5 Naïve CD8 + T <strong>cells</strong> are efficiently activated in BM niches in<br />

absence of DCs 49<br />

3.2.6 Naïve CD8 + T <strong>cells</strong> fail to <strong>for</strong>m multi-cell clusters in mice<br />

4


harboring MHC cl<strong>as</strong>s-I-deficient stroma 53<br />

3.3 Discussion 54<br />

4. Organ-dependent in vivo <strong>priming</strong> of naïve CD4 + , but not<br />

CD8 + T <strong>cells</strong> by pl<strong>as</strong>macytoid dendritic <strong>cells</strong> 59<br />

4.1 Introduction 59<br />

4.2 Results 60<br />

4.2.1 PDCs are spared from ablation in DTx-treated CD11c-DTRtg mice 60<br />

4.2.2 PDCs fail to prime naïve T <strong>cells</strong> in the spleen 63<br />

4.2.3 PDCs can prime naïve CD4 + T <strong>cells</strong> in LNs, but fail to support<br />

CD8 + T cell <strong>priming</strong> 65<br />

4.2.4 LN PDCs efficiently acquire and process soluble antigen and<br />

prime productive T cell responses 72<br />

4.2.5 PDC-specific antigen targeting and PDC-mediated in vivo and<br />

in vitro CD4 + T cell <strong>priming</strong> 74<br />

4.3 Discussion 79<br />

5. Final discussion 82<br />

6. Material and Methods 86<br />

6.1 Periv<strong>as</strong>cular clusters of dendritic <strong>cells</strong> provide critical survival<br />

signals to B <strong>cells</strong> in bone <strong>marrow</strong> niches 86<br />

6.2 The bone <strong>marrow</strong> immune niche: <strong>Bone</strong> <strong>marrow</strong> <strong>as</strong> a <strong>priming</strong><br />

<strong>site</strong> <strong>for</strong> naïve CD8 + T <strong>cells</strong> 88<br />

6.3 Organ-dependent in vivo <strong>priming</strong> of naïve CD4 + , but not<br />

CD8 + T <strong>cells</strong> by pl<strong>as</strong>macytoid dendritic <strong>cells</strong> 90<br />

7. References 94<br />

8. List of publications 106<br />

9. Statement about independent collaboration 107<br />

5


Summary<br />

Dendritic <strong>cells</strong> (DCs) play a central role in T-cell activation and the control of the<br />

inherent autoreactivity of the T-cell compartment. Pleiotropic DC functions are likely<br />

<strong>as</strong>sociated with discrete DC subsets. However, the latter remain largely defined by<br />

phenotype and unique anatomic location, rather than function. The investigation of<br />

DC involvement in complex phenomena that rely on multicellular interactions, such<br />

<strong>as</strong> immuno-stimulation and tolerization calls <strong>for</strong> an <strong>as</strong>sessment of DC functions<br />

within physiological context. Given the highly dynamic DC compartment, in the<br />

current study, we investigated in vivo DC functions by their conditional ablation in the<br />

intact organism.<br />

In my PhD thesis we phenotypically and functionally characterized bone <strong>marrow</strong>resident<br />

DCs (bmDCs). We identified an architecturally-defined novel bone <strong>marrow</strong><br />

niche defined by DC clusters, which wrap distinct blood vessels in steady-state.<br />

Suggesting a potential role in adaptive immune responses, the niches harbored both<br />

mature B and T <strong>cells</strong>. Surprisingly, we found that bmDC ablation resulted in the<br />

specific loss of B <strong>cells</strong> from the niches. The combined use of transgenic and knockout<br />

mouse models, finally allowed us to define the molecular mechanism <strong>for</strong> the B<br />

cell loss. Thus, bmDC provide a critical survival factor, the cytokine MIF, to<br />

recirculating B <strong>cells</strong>, which ensures their maintenance in the bone <strong>marrow</strong> immune<br />

niches. The identification of lymphoid follicle-like niches in the BM represents a<br />

breakthrough in our understanding of this organ <strong>as</strong> a potential <strong>site</strong> of primary immune<br />

responses against blood-borne pathogens. Furthermore, we identified the origin of<br />

bmDCs by showing that they arose without monocytic intermediate from<br />

macrophage/DC precursor (MDP). In addition we studied the motility of T and B<br />

lymphocytes in the bmDC niches in steady-state. Application of state-of-the-art<br />

intravital microscopy, allowed us to show that naïve CD8 + T <strong>cells</strong> <strong>for</strong>med multi-cell<br />

clusters and differentiated into effector <strong>cells</strong> in the bmDC niches upon blood-borne<br />

antigen stimulation, which fulfill the function of BM <strong>as</strong> a secondary lymphoid organ.<br />

Unexpectedly, however, we observed that naïve CD8 + T <strong>cells</strong> were efficiently<br />

activated and primed in BM niches in absence of DCs, challenging the critical APC<br />

function of DCs observed <strong>for</strong> the spleen. Using the BM chimera approach, we<br />

revealed that radio-resistant stromal <strong>cells</strong> were critical <strong>for</strong> antigen cross-presentation<br />

6


and naïve CD8 + T cell activation in absence of DCs. Taken together, our results thus,<br />

suggest the novel unique role of nonhematopoietic <strong>cells</strong> in the BM.<br />

Finally, we unraveled the APC potential of pl<strong>as</strong>macytoid dendritic cell (PDCs), which<br />

due to the lack of suitable experimental system - h<strong>as</strong> remained controversial. In most<br />

p<strong>as</strong>t experimental systems any putative in vivo T cell <strong>priming</strong> potential of PDCs could<br />

have been m<strong>as</strong>ked by the presence of conventional DCs (cDCs). In contr<strong>as</strong>t here, we<br />

studied in vivo CD4 + and CD8 + T cell responses in mice that were conditionally<br />

depleted of cDCs, but retained PDCs. Furthermore, we use a novel antibody-mediated<br />

strategy to specifically target cDCs and PDCs. Using these approaches, we showed<br />

that PDCs were unable to support CD4 + and CD8 + T cell responses in cDC-depleted<br />

spleens. However, surprisingly, we found that in absence of cDCs, PDCs were able to<br />

stimulate productive naive CD4 + T cell responses in lymph nodes (LNs).<br />

Interestingly, and in contr<strong>as</strong>t to the cDC-driven immune responses these PDCtriggered<br />

responses were not <strong>as</strong>sociated with concomitant CTL responses. Our results,<br />

thus, characterized PDCs <strong>as</strong> bona fide DCs that can initiate unique CD4 + helper T<br />

cell-dominated primary immune responses.<br />

7


1. Introduction<br />

1.1 Dendritic <strong>cells</strong> (DCs)<br />

Dendritic <strong>cells</strong> (DCs) are hematopoietic <strong>cells</strong> that belong to the family of<br />

“professional” antigen-presenting <strong>cells</strong> (APCs), which also includes B <strong>cells</strong> and<br />

macrophages. In fact, DCs were originally discovered b<strong>as</strong>ed on their APC function,<br />

when Steinman and Cohn in 1973 first identified DCs in mouse spleen <strong>as</strong> potent<br />

stimulators of the primary immune response [1].<br />

DCs belong to myeloid-lineage derived mononuclear phagocytes and are a<br />

heterogeneous population of <strong>cells</strong> that can be divided into two major populations: (1)<br />

nonlymphoid tissue migratory (mainly called tissue DCs) and lymphoid tissue–<br />

resident DCs (also called “cl<strong>as</strong>sic” or “conventional” DCs (cDCs)) and (2)<br />

pl<strong>as</strong>macytoid DCs (PDCs) (also called natural interferon-producing <strong>cells</strong>).<br />

Migratory and resident DCs have two main functions: the maintenance of selftolerance<br />

and the induction of specific immune responses against invading pathogens<br />

[1,2], where<strong>as</strong> the main function of PDCs is to secrete large amounts of interferon-α<br />

in response to viral infections and to prime T <strong>cells</strong> against viral antigens [3].<br />

1.1.1 DCs in nonlymphoid and lymphoid tissues<br />

Nonlymphoid tissue DCs can be further subdivided into <strong>cells</strong> present in sterile tissues,<br />

such <strong>as</strong> the pancre<strong>as</strong> and the heart, DCs present in filtering <strong>site</strong>s, such <strong>as</strong> the liver and<br />

the kidney, and DCs present at environmental interfaces <strong>as</strong> lung, gut, and skin.<br />

Among interface DCs, are the epidermal Langerhans’ <strong>cells</strong> (LCs). LCs constitutively<br />

express major histocompatibility complex (MHC) cl<strong>as</strong>s II and high levels of the lectin<br />

langerin, that is required and sufficient to <strong>for</strong>m the LC-characteristic intracytopl<strong>as</strong>mic<br />

Birbeck granules [4].<br />

Lymphoid tissue-resident DCs are the most studied DC populations in mice. They<br />

populate the spleen. In mice, splenic DCs that originate from blood-derived<br />

circulating precursors (pre-cDCs) [5] constitutively express MHC cl<strong>as</strong>s II and the<br />

integrin CD11c. CD11c hi cDCs can be further subdivided into CD4 + CD8α - CD11b +<br />

DCs that localize mostly in the marginal zone and CD8α + CD4 - CD11b - DCs localized<br />

in the marginal and T-cell zone [6]. CD4 - CD8α - CD11b + DCs have also been<br />

identified and are called double-negative (DN) DCs. CD8α + DCs are specialized in<br />

MHC cl<strong>as</strong>s I presentation, where<strong>as</strong> CD4 + DC subset is specialized in MHC cl<strong>as</strong>s II<br />

8


presentation [7]. CD8α + DCs have also been shown to cross-present cell-<strong>as</strong>sociated<br />

antigens, where<strong>as</strong> CD4 + DCs are unable to do so [8,9].<br />

Lymph node (LN) DCs are more heterogeneous <strong>as</strong> they include blood-derived<br />

lymphoid tissue-resident CD8α + , CD4 + , and DN spleen-equivalent DCs, plus<br />

additional migratory DCs entering via the afferent lymphatics that vary according to<br />

the LN draining <strong>site</strong> [10]. Thus <strong>for</strong> instance, migratory epidermal LCs and dermal<br />

DCs are present in skin-draining LNs, but are absent from mesenteric LNs, which<br />

contain intestinal lamina propria derived <strong>cells</strong>.<br />

1.1.2 Pl<strong>as</strong>macytoid DCs in nonlymphoid and lymphoid tissues<br />

Similarly to DCs, PDCs constitutively express MHC cl<strong>as</strong>s II molecules, but opposed<br />

to cDC they are characterized by low CD11c expression [11]. Murine PDCs lack<br />

CD11b and express CD45RA (B220) and Ly6C (Gr-1). Two PDC-restricted surface<br />

markers have been reported. The monoclonal antibodies 128G [12] and mPDCA-1<br />

[13] recognize BST-2 (bone <strong>marrow</strong> stromal cell antigen-2) [14] on PDCs. Injection<br />

of these antibodies results in the depletion of PDCs [15,16]. BST-2 h<strong>as</strong> been recently<br />

described <strong>as</strong> a ‘‘tetherin’’ that holds newly <strong>for</strong>med virions on the surface of infected<br />

<strong>cells</strong> to limit viral spread [17]. It also mediates endocytosis of virions, so it may<br />

function <strong>as</strong> a receptor <strong>for</strong> presentation of antigens contained in viral particles.<br />

Notably, the specificity of BST-2 <strong>as</strong> a PDC marker w<strong>as</strong> recently challenged, because<br />

BST-2 is inducible by IFN-γ treatment on other <strong>cells</strong>, including T <strong>cells</strong>, B <strong>cells</strong> and<br />

pl<strong>as</strong>ma <strong>cells</strong> [14]. The second PDC-restricted surface marker is Siglec-H, a member<br />

of the sialic acid binding Ig-like lectin (Siglec) family, which is recognized by the<br />

440C antibody that blocks type I IFN production by PDCs, but does not induce<br />

depletion [18].<br />

PDCs circulate in blood and are found in steady-state BM, spleen, thymus, LN, and<br />

the liver. They enter the LNs through the high endothelial venules (HEVs) and<br />

accumulate in the paracortical T cell-rich are<strong>as</strong> [19]. PDCs do not efficiently migrate<br />

to peripheral tissue in the steady state with the exception of the liver. The PDC<br />

proliferation rate in lymphoid organs is very low, and less than 0.3% PDCs are in cell<br />

cycle. On continuous BrdU labeling over 14 days in vivo, 90% of PDCs were found<br />

positive [11]. Thus, the lifespan of differentiated PDCs in the spleen and LNs is short<br />

and continuous replacement via blood is essential.<br />

9


The most characteristic functional feature of PDCs is their unique propensity to<br />

secrete large amounts of type I interferons (IFN I) in response to viral infections,<br />

owing to their constitutive expression of the transcription factor IRF-7 [20]. Helixloop-helix<br />

transcription factor (E protein) E2-2/Tcf4 is preferentially expressed in<br />

murine and human PDCs and is required <strong>for</strong> their development of IFN I response<br />

[21]. To respond to pathogens, PDCs do not need to be infected. They can detect the<br />

unique structural features of viral nucleic acids, such <strong>as</strong> unmethylated CpG-rich DNA<br />

motifs or double-stranded RNA, by employing Toll-like receptors (TLRs). When<br />

TLRs engage these motifs, they initiate a signaling c<strong>as</strong>cade that results in PDC<br />

activation. In addition to secreting IFN I, activated PDCs undergo other important<br />

phenotypic changes, notably the acquisition of a dendritic morphology and the<br />

upregulation of MHC and T cell costimulatory molecules, which enable PDCs to<br />

engage and activate naive T <strong>cells</strong> [11,22,23,24,25,26]. These are also the major<br />

changes that activated cDCs go through when they detect pathogens, and these<br />

changes epitomize the so-called maturation process [27]. By secreting IFN I, PDCs<br />

can activate both the innate (e.g., natural killer <strong>cells</strong>) and the acquired (e.g., cDCs and<br />

B <strong>cells</strong>) arms of the immune system.<br />

There h<strong>as</strong> been some controversy regarding whether PDCs are capable of <strong>priming</strong>.<br />

The expression of MHC and T cell costimulatory molecules on activated PDCs is not<br />

<strong>as</strong> high <strong>as</strong> on their cDC counterparts, and this is probably why PDCs tend to be less<br />

efficient at T cell <strong>priming</strong> than cDCs. Nevertheless, PDCs can stimulate immunity<br />

upon adoptive transfer [28,29], or sustain protective responses at <strong>site</strong>s of infection<br />

[30], demonstrating their immunogenic potential. Several reports have indicated that<br />

PDCs can also induce T cell tolerance, primarily through the induction of regulatory T<br />

<strong>cells</strong>, a capacity they also share with cDCs [31,32,33,34,35]. This may enable PDCs<br />

to promote regulatory T cell <strong>for</strong>mation within the infection <strong>site</strong> to dampen the<br />

immune response and prevent immunopathology [36,37,38].<br />

1.1.3 Antigen presentation by DCs<br />

The functional feature that defines DCs is their unrivaled capacity to capture, process<br />

and present antigens, which is a prerequist <strong>for</strong> T cell <strong>priming</strong>. However, not all DCs<br />

have equivalent antigen-presenting roles in vivo. Differences may derive from<br />

intrinsic abilities of individual DC types to capture and process antigens, and to load<br />

the resulting antigenic peptides onto their MHC molecules. All DC subtypes express<br />

high levels of MHC cl<strong>as</strong>s I and II molecules, and thus can potentially present peptide<br />

10


antigens to CD8 + and CD4 + T <strong>cells</strong>, respectively. Antigens can be categorized<br />

according to their source <strong>as</strong> endogenous (that is synthesized by the APC itself) or<br />

exogenous (that is synthesized by other <strong>cells</strong> and taken up by the APC) [39]. Most<br />

endogenous antigens are derived from proteosomal degradation of proteins in the<br />

cytopl<strong>as</strong>m. Peptides are subsequently transported into the ER and loaded onto MHC<br />

cl<strong>as</strong>s I molecules [39] More recently, it h<strong>as</strong> been shown that endogenous proteins can<br />

also reach the MHC cl<strong>as</strong>s II pathway, via autophagy [40]. Exogenous antigens are<br />

internalized by pinocytosis, phagocytosis or receptor-mediated endocytosis [39].<br />

Then, these antigens become accessible to the endosomal prote<strong>as</strong>es, are processed<br />

into peptides that can be loaded in specialized endosomal compartments on MHC<br />

cl<strong>as</strong>s II molecules. Presentation of exogenous antigens on MHC cl<strong>as</strong>s I molecules, a<br />

process known <strong>as</strong> cross-presentation, requires the presence of a unique phagosome-tocytosol<br />

pathway. As most <strong>cells</strong> express MHC cl<strong>as</strong>s I molecules, but only a few can<br />

cross-present, this pathway must rely on specific machinery to handle endocytosed<br />

antigens, but how this machinery works remains under debate [41].<br />

Like direct presentation, cross presentation can have two out-comes: it can lead to the<br />

triggering of protective CD8 + T cell responses (cross-<strong>priming</strong>) or to the neutralization<br />

of self-reactive CD8 + T <strong>cells</strong> (cross-tolerance). Cross-<strong>priming</strong> is essential <strong>for</strong> the<br />

defense against intracellular pathogens, incl. bacteria and viruses. Importantly, DCs<br />

seem to be uniquely able to cross-prime naïve T <strong>cells</strong> in vivo and are absolutely<br />

required <strong>for</strong> certain responses [42]. Cross <strong>priming</strong> is also believed to play a role in<br />

transplant rejection and cancer immuno-surveillance, although the latter remains<br />

under debate [43,44]<br />

1.1.4 Differentiation of DCs from hematopoietic progenitor <strong>cells</strong><br />

The “early DC progenitors” present in the BM, have high proliferative capacity and<br />

express CD117 (c-kit, the receptor <strong>for</strong> stem cell factor), but no mature lineage marker.<br />

In clonal in vitro <strong>as</strong>says and on in vivo transfer, they generate offspring <strong>cells</strong><br />

severalfold the progenitor input numbers. These so-called “macrophage and DC<br />

progenitors” (MDPs) [45] give rise to monocytes, preDCs, macrophages, and DCs, <strong>as</strong><br />

well <strong>as</strong> PDCs.<br />

Monocytes develop in the BM from dividing myeloid precursors, and are rele<strong>as</strong>ed to<br />

the peripheral circulation <strong>as</strong> non-dividing <strong>cells</strong> [46], be<strong>for</strong>e they enter tissues. Murine<br />

BM and blood monocytes are best defined by their expression of the macrophagecolony-stimulating-factor<br />

receptor (M-CSF receptor, CD115), CD11b (membrane-<br />

11


activating complex 1) and the F4/80 antigen [47]. Distinct surface expression levels of<br />

the chemokine receptor allow the identification of two discrete Ly6C high and Ly6C low<br />

monocyte subsets [48,49]. The monocyte subsets are also functionally distinct: the<br />

Ly6C low population w<strong>as</strong> characterized by homing to non-inflamed tissues, where<strong>as</strong><br />

Ly6C high<br />

<strong>cells</strong> were shown to be actively recruited to <strong>site</strong>s of inflammation.<br />

Adoptively transferred Ly6C high monocytes efficiently seeded the recipient spleen<br />

with CD11c int progeny, however, failed under non-inflammatory conditions to give<br />

rise to splenic conventional CD11c high DCs [50].<br />

1.2 <strong>Bone</strong> <strong>marrow</strong> (BM)<br />

The bone <strong>marrow</strong> occupies the cavities of the bones throughout the skeleton and<br />

contains a dense network of medullary v<strong>as</strong>cular sinuses. The densely packed extrav<strong>as</strong>cular<br />

spaces between the sinuses are the <strong>site</strong>s where lymphoid and myeloid<br />

lineages of hematopoietic precursors arise from common multi-potential progenitors.<br />

BM v<strong>as</strong>culature is distinctive because it h<strong>as</strong> similar characteristics to v<strong>as</strong>culature in<br />

inflammed tissues with a greater expression of leukocyte adhesion molecules <strong>as</strong><br />

compared with normal vessels [51]. Endothelial <strong>cells</strong> direct trafficking of <strong>cells</strong> into<br />

the BM via series of molecular interactions between specific v<strong>as</strong>cular cell adhesion<br />

molecules and chemokines <strong>as</strong> well <strong>as</strong> their cognate ligands and receptors on<br />

circulating <strong>cells</strong> [51].<br />

BM contains at le<strong>as</strong>t seven types of stromal <strong>cells</strong> (osteobl<strong>as</strong>ts, osteocl<strong>as</strong>ts, fat <strong>cells</strong>,<br />

m<strong>as</strong>t <strong>cells</strong>, fibrobl<strong>as</strong>ts, endothelial <strong>cells</strong> and smooth muscle <strong>cells</strong>) that undergo<br />

complex interactions with hematopoietic <strong>cells</strong> [52]. Thus, BM is organized in distinct<br />

"niches", which provide support <strong>for</strong> hematopoiesis, <strong>as</strong> well <strong>as</strong> the proper environment<br />

<strong>for</strong> the maintenance of <strong>cells</strong> that homed to the BM from the periphery [53]. The<br />

spatial organization of these <strong>cells</strong> and their interaction with the microenvironment<br />

within the BM remain poorly understood.<br />

Recent studies show that in addition to its role in hematopoesis, the BM is also an<br />

important <strong>site</strong> <strong>for</strong> cytotoxic T cell activation and memory T cell generation [54]. In<br />

addition, it w<strong>as</strong> suggested that the BM might support <strong>for</strong> B cell responses to antigen<br />

challenge [55]. Together these findings indicate that the BM might fulfill a dual<br />

function <strong>as</strong> primary and secondary lymphoid organ.<br />

12


1.2.1 B cell development<br />

B <strong>cells</strong> are generated from T, B and NK cell-committed common lymphoid precursors<br />

(CLPs) that derive from haematopoietic stem <strong>cells</strong> (HSCs) in the fetal liver and adult<br />

BM. In B cell differentiation, these CLPs give initially rise to B cell precursors that<br />

are negative <strong>for</strong> surface Ig but positive <strong>for</strong> the B cell lineage marker B220. According<br />

to their differential expression of additional cell surface markers these precursors have<br />

been divided into four subsets termed pre-pro-B <strong>cells</strong>, pro-B <strong>cells</strong>, pre-B <strong>cells</strong> and<br />

immature B <strong>cells</strong>. Pre-pro B <strong>cells</strong> retain all Ig heavy and light chain genes in germline<br />

configuration, where<strong>as</strong> pro-B <strong>cells</strong> have rearranged heavy chain D-J (diversity and<br />

junction) segments. Successful heavy-chain gene rearrangement leads to generation of<br />

pre-B <strong>cells</strong>, which are characterized by surface expression of pre-B-cell receptor (pre-<br />

BCR). At this stage light-chain rearrangement takes place, and if successful results in<br />

surface expression of a BCR on the immature B <strong>cells</strong> [53,56]. Immature IgM + B <strong>cells</strong><br />

exit the BM and reach the spleen, where they mature into peripheral mature IgM +<br />

IgD + B <strong>cells</strong>. Newly <strong>for</strong>med B <strong>cells</strong> give rise to either follicular or marginal zone B<br />

<strong>cells</strong>. Follicular B <strong>cells</strong> comprise the bulk of the peripheral B cell pool, are the main<br />

cellular constituent of the splenic and lymph node follicles, recirculate throughout the<br />

blood and lymphatic systems and contribute to the primary and memory humoral<br />

response by generating both antibody secreting pl<strong>as</strong>ma <strong>cells</strong> and germinal center B<br />

<strong>cells</strong>. In contr<strong>as</strong>t, marginal zone B <strong>cells</strong> constitute only 5% of all peripheral B <strong>cells</strong>,<br />

are localized to the marginal sinus of the spleen, do not recirculate and contribute to<br />

the early primary humoral response [57]. Pl<strong>as</strong>ma <strong>cells</strong> are terminally differentiated B<br />

<strong>cells</strong> specialized in Ig secretion. They develop in response to activation of mature B<br />

<strong>cells</strong> by antigen in peripheral lymphoid organs, and then return to colonize the BM<br />

[58].<br />

The identity of the BM stromal <strong>cells</strong> establishing the niches <strong>for</strong> B (and T)<br />

lymphocytes remains uncertain. By using CXCL12 GFP knockin mice, it h<strong>as</strong> been<br />

shown that BM stromal <strong>cells</strong> expressing high levels of CXCL12 are different from<br />

those producing IL-7. The two types of BM stromal <strong>cells</strong> were found to be <strong>as</strong>sociated<br />

with B lymphocytes at different developmental stages, so that pre-pro-B <strong>cells</strong> and<br />

pl<strong>as</strong>ma <strong>cells</strong> adjoined CXCL12-expressing stromal <strong>cells</strong>, where<strong>as</strong> pro-B <strong>cells</strong> were in<br />

contact with IL-7-producing stromal <strong>cells</strong> [59].<br />

13


1.2.2 B cell maintenance<br />

The steady- state B cell pool size is determined by the B cell production rate and<br />

lifespan of mature B <strong>cells</strong>. The majority of the peripheral B <strong>cells</strong> are small resting<br />

<strong>cells</strong> that are programmed to respond rapidly to antigenic challenge. They are actively<br />

supported in this quiescent state by extrinsic survival factors that prevent death by<br />

apoptosis. The ligands and receptors necessary <strong>for</strong> B cell growth and survival have<br />

been identified <strong>as</strong> members of the tumor necrosis factor (TNF) superfamily [90]. The<br />

primary ligand is B cell-activating factor (BAFF), which is synthesized by<br />

radioresistant stromal <strong>cells</strong> [91], macrophages, DCs and neutrophils [92]. BAFF binds<br />

three distinct receptors: transmembrane activator (TACI), B cell maturation antigen<br />

(BCMA) and BAFF-R. A related ligand, a proliferation-inducing ligand (APRIL),<br />

binds only TACI and BCMA and is critical <strong>for</strong> pl<strong>as</strong>ma cell survival [196]. Studies of<br />

BAFF - / - mice showed that BAFF is not required <strong>for</strong> B cell development in the BM<br />

[197], however, in these mice, B cell development is blocked at the immature stage<br />

and no mature B <strong>cells</strong> are present in BM or periphery [197]. In addition, treatment<br />

with soluble BAFF receptor (TACI-Ig) results in rapid loss of mature peripheral B<br />

<strong>cells</strong> [198].<br />

CD74 (invariant chain, Ii) is a nonpolymorphic type II integral membrane protein that<br />

w<strong>as</strong> originally thought to function mainly <strong>as</strong> an MHC cl<strong>as</strong>s II chaperone [199].<br />

However, CD74 w<strong>as</strong> recently found to play an additional role <strong>as</strong> accessory signaling<br />

molecule. A small proportion of CD74 is expressed on the surface of antigenpresenting<br />

<strong>cells</strong> together with CD44 [200]. In macrophages, CD74 w<strong>as</strong> recently<br />

reported to demonstrate high-affinity binding to the proinflammatory cytokine,<br />

macrophage migration-inhibitory factor (MIF). MIF binds to the extracellular domain<br />

of CD74, and this interaction is required <strong>for</strong> MIF-mediated cell proliferation [95]. In<br />

B <strong>cells</strong>, MIF initiates a signaling c<strong>as</strong>cade that involves Syk and Akt, leading to NF- B<br />

activation, proliferation, and survival in a CD74- and CD44-dependent manner [201].<br />

CD74 stimulation results in activation of the p53 family member, TAp63, which in<br />

turn transactivates the Bcl2 gene and augment B-cell survival [98].<br />

Thus, MIF regulates the adaptive immune response by maintaining the mature B cell<br />

population in the periphery.<br />

14


1.2.3 T <strong>cells</strong> in the BM<br />

Mature T <strong>cells</strong> circulating in the periphery enter primary lymphoid organs through the<br />

blood route. In the thymus, single positive CD4 and CD8 T <strong>cells</strong> represent recently<br />

generated naive T <strong>cells</strong>, which have completed their development and are ready to<br />

egress [60]. In the BM, mature T <strong>cells</strong> represent about 3–8% of total nucleated <strong>cells</strong>.<br />

The CD4:CD8 ratio in the BM is approximately 1:2 [61]. BM T <strong>cells</strong> arrive from the<br />

blood route and, after homing to the BM, can move back to the blood and then<br />

migrate to other lymphoid organs [61]. In addition, memory CD8 + T <strong>cells</strong> reside and<br />

proliferate in the BM [62]. Intravital microscopy studies of mouse BM showed that<br />

central memory CD8 T cell home preferentially to the BM. Their rolling in BM<br />

microvessels occurs through L-, P- and E-selectins, where<strong>as</strong> sticking is mostly<br />

mediated by the interaction between the lymphocyte integrin α4β1 and the endothelial<br />

adhesion molecule VCAM-1 [63]. Chemokines, such <strong>as</strong> CXCL12 mediate α4β1<br />

integrin activation [63]. This is in agreement with the finding that high levels of<br />

CXCL12, a m<strong>as</strong>ter regulator of HSC recruitment into the BM, are present in normal<br />

BM sinusoidal endothelium.<br />

The anatomical location of the niches where T <strong>cells</strong> localize in the BM and the<br />

cellular components of such niches are current topics of investigation. In vivo imaging<br />

studies showed that adoptively transferred T <strong>cells</strong> migrated to specific BM<br />

periv<strong>as</strong>cular regions, which were localized par<strong>as</strong>agittally within the frontal and<br />

parietal bones of the mouse skull and were defined by a specialized endothelium by<br />

leukemic <strong>cells</strong> <strong>for</strong> BM seeding. T <strong>cells</strong> were found in these regions <strong>for</strong> at le<strong>as</strong>t 2<br />

weeks after transfer [64].<br />

1.3 Intravital microscopy<br />

Two-photon microscopy is a relatively new imaging method that allows studying<br />

dynamic cell behavior in vivo and in situ. Two-photon microscopy overcomes<br />

previous limitations of deep tissue imaging by using femtosecond-pulsed infra-red<br />

l<strong>as</strong>er sources, which feature the following advantages: 1) deeper tissue penetration of<br />

up to several hundred microns; 2) reduced phototoxicity and photobleaching,<br />

permitting long-term imaging of living tissue; 3) simultaneous visualization of several<br />

fluorophores using multichannel photodetectors; and 4) visualization of unlabeled<br />

15


tissue components (i.e. collagen, laminin, and el<strong>as</strong>tin) using second harmonic<br />

generation [65,66].<br />

Using intravital microscopy investigators are dissecting the cellular and molecular<br />

underpinnings controlling immune cell motility and interactions in the tussues. Twophoton<br />

microscopy enables to observe, quantify and probe the complex behaviors of<br />

units such <strong>as</strong> T <strong>cells</strong>, B <strong>cells</strong>, granulocytes and DCs in their native anatomic context.<br />

The l<strong>as</strong>er allows rapid acquisition of stacks of 3D stacks, which are used to produce<br />

3D time-lapse movies. From the movies, tracking of migration paths of individual<br />

<strong>cells</strong> are me<strong>as</strong>ured. Then, the track coordinates are exported <strong>as</strong> numerical datab<strong>as</strong>es<br />

and used to compute various parameters defining cell migration [65].<br />

1.3.1 BM <strong>as</strong> a model <strong>for</strong> intravital imaging<br />

The thickness of bone surrounding BM cavities in most anatomic locations renders<br />

these tissues inaccessible <strong>for</strong> in vivo imaging. However, recently, a new method that<br />

allows microscopic observation in BM of anesthetized mice h<strong>as</strong> been developed [52].<br />

It exploits the fact that the thin flat bones that <strong>for</strong>m the skull's calvaria are sufficiently<br />

transparent to allow imaging. This technique that maintains tissue integrity without<br />

requiring surgical manipulation, except a small skin incision [52]. To enhance the<br />

visibility of the BM microcirculation and allow me<strong>as</strong>urements of its luminal<br />

dimentions, an inert, high molecular weight pl<strong>as</strong>ma marker that remains strictly<br />

confined to the intrav<strong>as</strong>cular compartment is injected [52]. The organization of the<br />

v<strong>as</strong>culature in murine skull BM h<strong>as</strong> parallels to that of long bones. Moreover, the<br />

space between and around venules and sinusoids that is occupied by hematopoietic<br />

tissue contains hematopoietic precursor <strong>cells</strong> at a similar frequency <strong>as</strong> the femoral<br />

BM [67].<br />

16


2. Periv<strong>as</strong>cular clusters of dendritic <strong>cells</strong> provide critical<br />

survival signals to B <strong>cells</strong> in bone <strong>marrow</strong> niches<br />

2.1 Introduction<br />

The bone <strong>marrow</strong> (BM) is generally considered a primary lymphoid organ providing a<br />

unique microenvironment with defined niches <strong>for</strong> stem <strong>cells</strong>, <strong>as</strong> well <strong>as</strong> <strong>for</strong> lymphoand<br />

myelogenesis [53,68,69,70]. Early B cell development is confined to the BM<br />

where it comprises a number of defined states (pre-pro-B, pro-B, pre-B) [71].<br />

Immature B <strong>cells</strong> then leave the BM and mature in the spleen [72]. Notably, up to a<br />

quarter of the mature B cell pool positive <strong>for</strong> immunoglobulin D (IgD) and IgM can<br />

subsequently be found recirculating in steady state through the BM [55]. Moreover,<br />

these BM-resident mature B <strong>cells</strong> can participate in situ in T-independent humoral<br />

immune responses to blood-borne microbes [55]. Finally the BM constitutes a storage<br />

<strong>site</strong> <strong>for</strong> terminally differentiated B <strong>cells</strong>, i.e. long-lived pl<strong>as</strong>ma <strong>cells</strong> [59,73]. T cell<br />

development occurs in the thymus, but mature CD4 and CD8 T <strong>cells</strong> continuously<br />

home to the BM <strong>as</strong> well [61]. In addition, it is well established that this organ<br />

represents a principal reservoir <strong>for</strong> both CD4 + and CD8 + memory T <strong>cells</strong>, which<br />

represent a chief obstacle to BM transplantation [74,75]. BM-resident naïve and<br />

memory T <strong>cells</strong> can be both activated in situ, suggesting the presence of professional<br />

antigen presenting <strong>cells</strong> (APC) in the BM [54,75,76]. Collectively these findings show<br />

that in addition to its established function <strong>as</strong> the primary lymphoid organ in<br />

hematopoiesis, the BM can also act <strong>as</strong> secondary lymphoid organ, providing a suitable<br />

microenvironment <strong>for</strong> initiation and restimulation of adaptive immune responses.<br />

Recent histological and imaging data emph<strong>as</strong>ize the importance of functional<br />

compartmentalization of lymphoid organs. The existence and localization of a<br />

postulated BM immune niche [77], however, remains elusive.<br />

Dendritic <strong>cells</strong> (DCs) are mononuclear phagocytes specialized in the stimulation and<br />

tolerization of T lymphocytes [1,78]. Emph<strong>as</strong>izing their function in<br />

immunosurveillance, most tissues are populated with DCs that translocate to draining<br />

lymph nodes (LNs) after encountering antigen. In addition to those migratory DCs,<br />

there are also lymphoid tissue-resident DCs, which are believed to collect and present<br />

antigen in the tissue itself [78]. Not surprisingly, <strong>as</strong> an established hallmark of<br />

adaptive immunity, DCs are found in greatest numbers and complexity at <strong>site</strong>s where<br />

17


immune responses are initiated, such <strong>as</strong> the secondary lymphoid organs [79].<br />

Here we report the phenotypic characterization of BM-resident Dendritic <strong>cells</strong><br />

(bmDCs). In situ imaging revealed that these rare <strong>cells</strong> were concentrated in the BM<br />

in unique periv<strong>as</strong>cular clusters enveloping blood vessels and seeded with mature B<br />

and T lymphocytes. Highlighting a critical role of the bmDCs in the maintaining the<br />

integrity of these putative BM immune niches, conditional bmDC ablation resulted in<br />

a specific loss of recirculating mature B <strong>cells</strong> from the BM. This phenotype could be<br />

reversed by transgenic overexpression of the antiapoptotic factor Bcl-2 in B <strong>cells</strong>.<br />

Finally, the survival of recirculating B <strong>cells</strong> in the niches specifically required the<br />

presence of bmDCs that produced macrophage migration-inhibitory factor (MIF).<br />

2.2 Results<br />

2.2.1 Phenotypic and functional characterization of bone <strong>marrow</strong> DCs<br />

The BM of naïve mice contained a sizable population of DCs phenotypically defined<br />

by the expression of major histocompatibility complex (MHC) cl<strong>as</strong>s II and the β<br />

integrin subunit CD11c (Fig. 1a); we obtained 1.1x10 6 ± 0.2x10 6 bmDCs from one<br />

femur and tibia of each wild-type C57BL/6 mouse (± s.d.; n=10). Comparative flow<br />

cytometry showed that bmDCs differed from the bulk of splenic conventional DCs<br />

(cDCs) by prominent expression of CD103 (α E integrin) and F4/80, a surface marker<br />

expressed on monocytes, macrophages and DC subsets (Fig. 1b). In addition, bmDCs<br />

had lower expression of CD62L and higher expression of Ly6C/G (Gr-1), CD11b and<br />

CD115 than did splenic cDCs. Furthermore, bmDCs had an activated phenotype, <strong>as</strong><br />

indicated by high expression of MHC cl<strong>as</strong>s II (I-A b ), CD40, CD44 and the costimulatory<br />

molecule CD86. DCs were originally identified by their ability to<br />

stimulate naive in vitro T cell responses and this functional feature remains the best<br />

criteria to distinguish them from the closely related macrophages [80]. To functionally<br />

define bmDCs, we there<strong>for</strong>e tested their ability to prime naïve alloreactive T <strong>cells</strong> in a<br />

mixed-leukocyte reaction (MLR). In contr<strong>as</strong>t to BM-resident monocytes and<br />

macrophages, defined <strong>as</strong> F4/80 + CD11c - MHC II - and F4/80 + CD11c- MHC II+ <strong>cells</strong>,<br />

respectively, the F4/80 + CD11chigh MHC cl<strong>as</strong>s II high bmDCs efficiently stimulated<br />

allogeneic naive CD4 + T <strong>cells</strong> (Fig. 1C). Hence, these <strong>cells</strong> can be cl<strong>as</strong>sified <strong>as</strong> true<br />

DCs.<br />

18


Figure 1. Characterization of the mouse bmDC compartment<br />

(a) Identification of CD11c high DCs (outlined) in BM and spleen by flow cytometry.<br />

(b) Flow cytometry of the expression of cell surface markers by bmDCs and splenic cDCs from<br />

C57BL/6 wt mice (gated <strong>as</strong>CD11c high MHC cl<strong>as</strong>s II + <strong>cells</strong>, <strong>as</strong> indicated in A). MFI, mean fluorescence<br />

intensity.<br />

(c) Mixed-leukocyte reaction with mononuclear phagocyte populations isolated from BM and spleen.<br />

Stimulator <strong>cells</strong> were sorted according to the following markers: splenic cDCs (CD11c-GFP + , F4/80 - ,<br />

MHC II + ); bmDCs (CD11c-GFP + , F4/80 + , MHC II + ); splenic macrophages (Spl MΦ) (CD11c-GFP - ,<br />

F4/80 + , MHC II + ); bmMΦ (CD11c-GFP - , F4/80 + , MHC II + ); and BM monocytes (CD11c-GFP - , F4/80 + ,<br />

MHC II - ). CD4 + T cell responses were me<strong>as</strong>ured by thymidine incorporation. Error bars represent SED.<br />

2.2.2 Organization of bmDCs into periv<strong>as</strong>cular clusters<br />

The DC compartment of secondary lymphoid organs is highly organized. Although<br />

adoptively transferred splenic cDCs have been visualized in the BM of recipient<br />

mice [54], the localization and organization of endogenous bmDCs remains unknown.<br />

To study the in vivo distribution of DCs in the BM, we used an in situ imaging<br />

19


strategy with mice containing DCs expressing green fluorescent protein (GFP; called<br />

Cx3cr1 gfp/+ mice) [81,82,83,84]. CD11chigh bmDCs of these Cx3cr1 gfp/+ mice were<br />

brightly labeled with green fluorescence (Fig. 2a). Furthermore, histologically,<br />

bmDCs could be distinguished from other <strong>cells</strong> expressing GFP-tagged CX3CR1 in<br />

Cx3cr1 gfp/+ BM, such <strong>as</strong> rare lymphocytes and monocytes [50,81], by their<br />

characteristic spindle-shaped morphology (Fig. 2). Surprisingly, two-photon<br />

microscopy of skull BM [67] of Cx3cr1 gfp/+ mice showed that the DCs in the cranial<br />

BM parenchyma were organized in unique periv<strong>as</strong>cular clusters wrapped around<br />

distinct blood vessels and occupying architecturally definable niches (Fig. 2b-d).<br />

Notably, bmDCs were not in direct contact with the vessel walls and thus were<br />

distinct from rare periv<strong>as</strong>cular BM macrophages present tight <strong>as</strong>sociation with the<br />

endothelium (Fig. 2e). The bmDC clusters were reminiscent of the perisinusoidal<br />

niches identified by adoptive B cell transfer [55]. To determine whether grafted B<br />

lymphocytes indeed localized together with the bmDC clusters, we adoptively<br />

transferred Hoechst-labeled splenic B <strong>cells</strong> into Cx3cr1 gfp/+ recipient mice. The<br />

grafted mature B <strong>cells</strong> homed to the BM, and their distribution w<strong>as</strong> restricted to the<br />

extrav<strong>as</strong>cular are<strong>as</strong> covered by bmDCs (Fig. 2f). Finally, we determined whether the<br />

niches defined by bmDCs also harbored T <strong>cells</strong>. Indeed, adoptively transferred T <strong>cells</strong><br />

homed to the periv<strong>as</strong>cular bmDC clusters and localize there together with bmDCs and<br />

B <strong>cells</strong> (Fig. 2g,h). We conclude that bmDCs define novel previously unidentified<br />

specific anatomic locations or niches in the BM that harbor DCs and recirculating B<br />

and T lymphocytes.<br />

20


Figure 2. bmDCs are organized into unique periv<strong>as</strong>cular clusters that define BM immune niches<br />

harboring B and T <strong>cells</strong><br />

(a) Flow cytometry of wt and Cx3cr1 gfp/+ BM, indicating GFP expression by CD11c high MHC II +<br />

bmDCs.<br />

(b, c, d) Two-photon microscopy of cranial BM cavity of Cx3cr1 gfp/+ mice (bmDCs – green; blood<br />

vessels- red).<br />

(e) Two-photon microscopy of cranial BM cavity of Cx3cr1 gfp/+ mouse. Note periv<strong>as</strong>cular MΦ (green),<br />

that are tightly <strong>as</strong>sociated with the endothelium (upper blood vessel) and distinct from bmDC clusters<br />

(green); blood vessels (red).<br />

(f) Two-photon microscopy of the colocalization of adoptively transferred B <strong>cells</strong> (blue), host bmDCs<br />

(green) and blood vessels (red) in BM cavities of Cx3cr1 gfp/+ mice at 5 h after transfer.<br />

(g) Two-photon microscopy of the localization of adoptively transferred T <strong>cells</strong> (blue), host bmDCs<br />

(green) and blood vessels (red) in BM cavities of Cx3cr1 gfp/+ mice 5 h after transfer of splenic CD4 +<br />

and CD8 + T <strong>cells</strong>. (h) Two-photon microscopy of the localization of adoptively transferred T <strong>cells</strong><br />

(blue), B cell (red) and host bmDCs (green) in BM cavities of Cx3cr1 gfp/+ mice at 5 h after transfer.<br />

Scale bars: 20 μm. Data are from one representative experiment of three.<br />

2.2.3 DC ablation causes depletion of recirculating B <strong>cells</strong> from BM<br />

To probe <strong>for</strong> bmDC functions, we next took advantage of the CD11c-diphtheria toxin<br />

receptor (DTR)-transgenic (CD11c-DTRtg) mouse model, which allows the<br />

conditional ablation of CD11c high DCs through the use of diphtheria toxin (DTx) [42].<br />

BM chimer<strong>as</strong> generated by the reconstitution of lethally irradiated recipient mice with<br />

21


CD11c-DTRtg BM tolerate repeated DTx injection without adverse side effects,<br />

which allows prolonged DC ablation [85]. Intraperitoneal DTx injection into chimer<strong>as</strong><br />

of wild-type mice reconstituted with CD11c-DTRtg BM [CD11c-DTRtg > wt]<br />

resulted in systemic depletion of DCs, including 90% of the bmDCs (Fig. 3a). The<br />

ablation of BM-resident DCs w<strong>as</strong> confirmed by two-photon imaging of cranial BM of<br />

CD11c-DTRtg Cx3cr1 gfp/+ (double-transgenic) mice. Notably, the periv<strong>as</strong>cular BM<br />

immune niches of these mice harbor both GFP + bmDCs and monocytes [50], which<br />

can be distinguished by their morphology. Only the characteristically spindle-shaped<br />

bmDCs expressed CD11c and were thus depleted by the DTx treatment of CD11c-<br />

DTRtg Cx3cr1 gfp/+ mice, where<strong>as</strong> monocytes were resistant to DTx (Fig. 3b).<br />

Figure 3. Conditional ablation of bmDCs<br />

(a) Flow cytometry of BM from CD11c-DTRtg Cx3cr1 gfp/+ mice not treated or 24 hafter intraperitoneal<br />

injection of DTx to deplete CD11c high DCs. Numbers adjacent to outlined are<strong>as</strong> indicate percent<br />

CD11c + MHC cl<strong>as</strong>s II + <strong>cells</strong>.<br />

(b) Two-photon microscopy of cranial BM cavity of CD11c-DTRtg Cx3cr1 gfp/+ mice, untreated or<br />

treated with DTx <strong>as</strong> described in A. Note distinct morphology of bmDCs and monocytes, which define<br />

the BM niche in the absence of bmDCs. Green – monocytes and bmDCs; red – blood vessels. Scale<br />

bars: 20 μm. Data are from one representative experiment of four.<br />

We next studied the outcome of the bmDC depletion on the cellular composition of<br />

the BM immune niches. Flow cytometry of the BM of untreated mice confirmed<br />

the presence of a distinct population of recirculating B <strong>cells</strong>, defined <strong>as</strong><br />

B220 + IgM + IgD + <strong>cells</strong> [72] or CD93 - (AA4.1) CD23 + <strong>cells</strong> [86]. Unexpectedly,<br />

bmDC-depleted mice exhibited a profound reduction of these mature B <strong>cells</strong>, which<br />

22


decre<strong>as</strong>ed in abundance with time (Fig. 4a,b,d). B cell development w<strong>as</strong> not<br />

substantially affected by bmDC ablation, <strong>as</strong> indicated by the presence of pro-B <strong>cells</strong>,<br />

pre-B <strong>cells</strong> and immature B <strong>cells</strong> (B220 + IgM- and IgM + IgD - CD93 + <strong>cells</strong>) (Fig. 4a,b).<br />

Moreover, the effect w<strong>as</strong> specific to the BM, since mature B cell numbers in spleen<br />

and LNs were not affected (Fig. 4c). BM-resident mature B <strong>cells</strong> have been shown to<br />

respond to antigenic challenge by in situ differentiation into antibody-secreting<br />

pl<strong>as</strong>mabl<strong>as</strong>ts [55]. The bmDC depletion would be expected to impair such humoral<br />

BM immune responses and thus have functional consequences. It h<strong>as</strong> been reported<br />

that short-lived pl<strong>as</strong>ma <strong>cells</strong> express CD11c and are hence DTx-sensitive in CD11c-<br />

DTRtg mice [87]. To test the outcome of bmDC ablation on short-lived pl<strong>as</strong>ma cell<br />

generation in the BM, we generated mixed BM chimer<strong>as</strong> by reconstitution of lethally<br />

irradiated wt recipients with a 95 : 5 mixture of CD11c-DTRtg recombinationactivating<br />

gene 2-defecient (RAG - / - CD45.2 + ) BM and wild-type (CD45.1 + ) BM. All<br />

B <strong>cells</strong> in the recipient mice were derived from the wild-type BM (CD45.1), where<strong>as</strong><br />

the bulk of the DCs expressed the DTR-GFP transgene [87] (Fig. 5a). Chimer<strong>as</strong> were<br />

inoculated daily with DTx to deplete DCs. To exclude an influx by pl<strong>as</strong>ma <strong>cells</strong> from<br />

the spleen, the animals were splenectomized [55]. Mice were immunized with the T-<br />

independent antigen NP-Ficoll, then 4 d later, BM <strong>cells</strong> were isolated and analyzed<br />

with an IgM enzyme-linked immunospot <strong>as</strong>say (ELISPOT) [87]. Ablation of bmDCs<br />

impaired the <strong>for</strong>mation of NP-specific IgM-secreting short-lived pl<strong>as</strong>ma cell in the<br />

BM of the <strong>as</strong>plenic mice (Fig. 5b). Cumulatively, these results indicate the critical<br />

requirement of bmDCs in the recruitment or maintenance of mature recirculating B<br />

<strong>cells</strong> inthe BM.<br />

23


Figure 4. BmDCs are required <strong>for</strong> the maintenance of recirculating mature B <strong>cells</strong> in<br />

the BM<br />

(a) Flow cytometry of BM B cell compartment of untreated and bmDC-depleted [CD11c-<br />

DTRtg > wt] BM chimer<strong>as</strong>. Pro, pre-B <strong>cells</strong> are plotted <strong>as</strong> B220 + IgM - IgD - , immature B <strong>cells</strong> are<br />

B220 + IgM + IgD - and mature B <strong>cells</strong> are B220 + IgM + IgD + . Note reduction of mature B <strong>cells</strong>.<br />

Bar diagram summarizes cell numbers obtained from flow cytometry <strong>for</strong> <strong>cells</strong> isolated from two fibiae<br />

and femurs (3 mice/ group). Data are from one representative experiment of five.<br />

(b) Flow cytometry of BM B cell compartment of untreated and bmDC-depleted [CD11c-DTRtg > wt]<br />

BM chimer<strong>as</strong>. Pro, pre-B <strong>cells</strong> and immature B <strong>cells</strong> are plotted <strong>as</strong> B220 + CD93 (AA4.1) + CD23 - <strong>cells</strong><br />

and mature B <strong>cells</strong> are B220 + CD93 (AA4.1) - CD23 + <strong>cells</strong>. Note reduction of mature B <strong>cells</strong>.<br />

(c) Summary of numbers of mature B <strong>cells</strong> in the BM, spleen and LNs of [CD11c-DTRtg > wt] BM<br />

chimer<strong>as</strong> left untreated or administered DTx <strong>for</strong> 5 d. Error bars represent s. d.<br />

of data obtained from analysis of three experiments with 9 mice / group.<br />

(d) Flow cytometry of BM B cell compartment of untreated and DTx-treated [CD11c-DTRtg > wt] BM<br />

chimer<strong>as</strong> showing disappearance kinetics of endogenous mature BM B <strong>cells</strong>. Of note, not all mature<br />

BM B <strong>cells</strong> are dependent on bmDCs.<br />

25


Figure 5. Ablation of bmDCs impaired the <strong>for</strong>mation of IgM-secreting short-lived pl<strong>as</strong>ma <strong>cells</strong><br />

(a) Flow cytometry of spleens of untreated and DTx-injected [95%CD11c-DTRtg Rag - / - / 5%wt > wt]<br />

BM chimer<strong>as</strong> <strong>for</strong> DCs and B <strong>cells</strong>. Note that most DCs are DTR-GFP + and ablated by the DTx<br />

treatment, where<strong>as</strong> all mature IgD + B <strong>cells</strong> are of wt origin (CD45.1) and unaffected by the treatment.<br />

(b) Ablation of bmDCs impairs NP-specific IgM-secreting pl<strong>as</strong>ma cell <strong>for</strong>mation in the BM of <strong>as</strong>plenic<br />

mixed BM chimer<strong>as</strong>. Scheme illustrating the experimental protocol. Graph showing the result of<br />

ELISPOT <strong>as</strong>say <strong>for</strong> NP-specific antibody-<strong>for</strong>ming <strong>cells</strong> (AFCs) in the BM of the various mice.<br />

26


2.2.4 BmDCs provide a survival signal to recirculating mature B <strong>cells</strong><br />

BmDCs could be a source of chemokines needed to attract mature B <strong>cells</strong> from the<br />

blood circulation. Alternatively, bmDCs could provide critical B survival factors in<br />

the periv<strong>as</strong>cular BM niches. To address those options, we isolated B <strong>cells</strong> from wildtype<br />

mice and mice overexpressing the antiapoptotic factor Bcl2 (Eμ-bcl-2-22 mice<br />

[88]; called Bcl-2-transgenic), labeled the <strong>cells</strong> with cell-tracking dye CMTMR and<br />

adoptively transferred them into recipient mice. When grafted into untreated [CD11c-<br />

DTRtg > wt] BM chimer<strong>as</strong>, wild-type B <strong>cells</strong> were readily detected in <strong>as</strong>sociation<br />

with the bmDC clusters; however, we failed to locate significant numbers of these<br />

<strong>cells</strong> in bmDC-depleted BM immune niches (Fig. 6a,b). In contr<strong>as</strong>t, we detected<br />

transferred Bcl-2tg B <strong>cells</strong> regardless of bmDC ablation (Fig. 6c,d). To confirm this<br />

result and to allow quantification, we repeated the experiment using allotype-marked<br />

B cell grafts and flow cytometry. As shown above (Fig. 4a,b), depletion of bmDCs<br />

resulted in a significant loss of mature host B <strong>cells</strong> from the BM of [CD11c-DTRtg ><br />

wt] BM chimer<strong>as</strong> (Fig. 6e). Furthermore, mice treated with DTx had half <strong>as</strong> many<br />

transferred wild-type B <strong>cells</strong> <strong>as</strong> did untreated controls mice, but there w<strong>as</strong> no<br />

significant loss of cell survival in treated mice after transfer of Bcl-2-tg B <strong>cells</strong> (Fig.<br />

6f). Importantly, the DTx-induced loss of the B cell graft w<strong>as</strong> specific to the BM, and<br />

did not occur in the spleens or LNs of the recipient mice (Fig. 6g). The finding that<br />

the B cell reduction could be observed with adoptively transferred wild-type B <strong>cells</strong><br />

ruled out the possibility that it w<strong>as</strong> due to a direct effect of the toxin on CD11c-DTRtg<br />

B <strong>cells</strong>. These results allow two conclusions to be drawn. First, bmDCs are not<br />

required <strong>for</strong> B cell homing to the BM, <strong>as</strong> Bcl-2tg B <strong>cells</strong> were presented in DCdepleted<br />

BM. Second and more importantly, the finding that en<strong>for</strong>ced overexpression<br />

of Bcl-2 restored the BM-resident mature B <strong>cells</strong> indicates that bmDCs provide these<br />

<strong>cells</strong> directly or indirectly with a critical survival signal.<br />

27


Figure 6. BmDCs provide survival signals to mature B <strong>cells</strong> in the BM<br />

(a-d) Multiphoton microscopy of cranial BM cavities of untreated and DTx-treated CD11c-DTRtg<br />

Cx3cr1 gfp/+ mice that received a wt (A,B) or Bcl-2 transgenic (C,D) B cell graft 24 h be<strong>for</strong>e B cell<br />

analysis; B <strong>cells</strong> (CMTMR, red) host bmDCs (green). Note absence of host bmDCs (green) from DTx-<br />

28


treated mice and unique survival of Bcl-2 transgenic B <strong>cells</strong> (CMTMR, red). Scale bars: 20 μm. Data<br />

are from one representative experiment of two.<br />

(e,f,g) Flow cytometry of BM B cell compartment of untreated and DTx-treated [CD11c-DTRtg > wt]<br />

BM chimer<strong>as</strong> 24 h after engraftment with splenic wt or Bcl-2tg B <strong>cells</strong>.<br />

(e) Analysis of host <strong>cells</strong> (CD45.1 - ) and donor B <strong>cells</strong> (CD45.1 + ) in BM.<br />

(f) Analysis of grafted <strong>cells</strong> in BM.<br />

(g) Analysis of grafted <strong>cells</strong> in spleen and LNs. Bar diagram summarizes results obtained from one<br />

representative experiment of two (3 mice/group).<br />

2.2.5 Recirculating mature B <strong>cells</strong> require MIF-producing bmDCs <strong>for</strong> survival<br />

Adoptively transferred T <strong>cells</strong> also home to the bmDC clusters and localized together<br />

with B <strong>cells</strong> (Fig. 2g,h). However, the abundance of BM T <strong>cells</strong> remained unaffected<br />

by the bmDC depletion (Fig. 7). It is thus unlikely that bmDCs act via T <strong>cells</strong> that<br />

provide an indirect B cell survival signal, such <strong>as</strong> CD40L [89].<br />

One of the best-characterized B cell survival factors is the B-cell activating factor<br />

(BAFF) [90], a member of the TNF family, which is required by mature splenic<br />

follicular and marginal zone B cell subpopulations. Although the splenic BAFF signal<br />

is provided by nonhematopoietic <strong>cells</strong>, including stromal and follicular DCs [91],<br />

BAFF production h<strong>as</strong> also been reported in immune <strong>cells</strong>, including in vitro cultured<br />

DCs [92,93]. To test whether bmDCs promote the mature BM B cell survival by<br />

secreting BAFF, we generated mixed BM chimer<strong>as</strong> by injecting BM from BAFFdeficient<br />

mice [94] and CD11c-DTRtg mice into wild-type mice. Injection of DTx<br />

into the resultant these [BAFF - / - / CD11c-DTRtg > wt] chimer<strong>as</strong> resulted in the<br />

specific ablation of BAFF + / + bmDCs expressing the GFP-DTR fusion protein [42],<br />

but it spared GFP - BAFF - / - bmDCs (Fig. 8a,b). In this setting, a loss of recirculating<br />

mature B <strong>cells</strong> would indicate that the critical survival factor provided by bmDC is<br />

BAFF. However, mature BM B <strong>cells</strong> were retained in the DTx-treated mixed<br />

chimer<strong>as</strong> that had lost CD11c-DTRtg BAFF + / + bmDCs and exclusively retained<br />

BAFF-deficient bmDCs (Fig. 8c). Importantly, this experimental set up did not<br />

address any potential contribution of BAFF-producing non-DCs to B cell survival, <strong>as</strong><br />

their wild type counterparts were not be ablated in the CD11c-DTRtg system. Rather<br />

our results establish that mature BM B cell survival w<strong>as</strong> independent of BAFFproducing<br />

bmDCs.<br />

29


Figure 7. T cell compartment in the BM remains unaffected by bmDC ablation<br />

Flow cytometry of BM T cell compartment of untreated and DTx-treated [CD11c-DTRtg > wt] BM<br />

chimer<strong>as</strong>. Note that the frequency of the CD4 + and CD8 + T <strong>cells</strong> remains unaffected by ablation of the<br />

bmDCs.<br />

30


Figure 8. Mature B cell survival in BM is independent of BAFF-producing bmDCs<br />

(a) Approach used to determine molecular contribution of DCs in BM chimer<strong>as</strong>. Note that after DTx<br />

treatment [CD11c-DTRtg / mutant > wt] mice are left with only mutant DCs.<br />

(b) Flow cytometry of DTx-induced DC depletion in [BAFF - / - /CD11c-DTRtg > wt] chimer<strong>as</strong>, on day<br />

6 after 5 daily intraperitoneal DTx injections. CD11c high DCs generated from CD11c-DTR BM and<br />

BAFF - / - BM are GFP + and GFP - , respectively. Data are from one representative experiment of three.<br />

(c) Flow cytometry of BM of [wt > wt], [BAFF - / - > wt] and [BAFF - / - /CD11c-DTRtg > wt] BM with<br />

or witout 5 daily intraperitoneal injections of DTx be<strong>for</strong>e analysis on day 6.chimer<strong>as</strong>. Bar diagram<br />

summarizes results obtained from one representative<br />

experiment of two (3 mice/group).<br />

The chemokine MIF h<strong>as</strong> been found to activate an antiapoptotic PI3K-Akt signaling<br />

pathway via triggering of a CD74-CD44 receptor complex [95,96]. Moreover, data<br />

indicate involvement of MIF in controlling the survival of mature B <strong>cells</strong> [97,98].<br />

Both CD74 and CD44 were expressed by BM-resident mature B <strong>cells</strong> (Fig. 9a). MIF<br />

is widely expressed by myeloid <strong>cells</strong>, including DCs [99]. RT-PCR analysis showed<br />

that where<strong>as</strong> all the mononuclear phagocyte populations in the BM transcribed MIF,<br />

its expression w<strong>as</strong> greater in bmDCs (Fig. 9b). To investigate the possible<br />

involvement of MIF in the maintenance of recirculating B <strong>cells</strong>, we analyzed the<br />

31


spleens and BM of MIF - / - mice and age-matched wild-type controls [99]. The<br />

frequency of splenic CD93 - CD23 + mature B <strong>cells</strong> w<strong>as</strong> not affected by the MIF<br />

deficiency (Fig. 9c). However, the BM of MIF - / - mice had significantly fewer mature<br />

B <strong>cells</strong> than did the BM of wild-type control mice (Fig. 9c). Although these data<br />

suggest that MIF might indeed play a role in the maintenance of the mature B <strong>cells</strong> in<br />

the BM, <strong>cells</strong> other than bmDCs could provide the critical MIF signal. To directly test<br />

whether MIF-expressing bmDCs are required <strong>for</strong> the maintenance of the mature B<br />

<strong>cells</strong>, we generated mixed BM chimer<strong>as</strong> by injecting wild-type mice with BM from<br />

MIF-deficient mice and CD11c-DTRtg mice. DTx treatment of these [MIF - / - / CD11c-<br />

DTRtg > wt] chimer<strong>as</strong> resulted in the specific ablation of GFP + MIF + / + bmDCs,<br />

sparing GFP - MIF - / - bmDCs (Fig. 9d). Importantly, and in contr<strong>as</strong>t to the [BAFF - / - /<br />

CD11c-DTRtg > wt] chimer<strong>as</strong> included <strong>as</strong> additional control, mice that retained after<br />

DTx treatment only MIF-deficient bmDCs in their BM, showed a significant loss of<br />

mature B <strong>cells</strong> from the BM, but not from the spleen (Fig. 9e). Notably, the mixedchimera<br />

approach specifically targeted DCs, but did not impair other MIF-producing<br />

<strong>cells</strong> in the BM, such <strong>as</strong> monocytes or macrophages (Fig. 9b). Depletion of bmDCs<br />

equally affected wild-type (CD45.1 + ) and MIF - / - (CD45.1 - ) B <strong>cells</strong> equally in the<br />

mixed chimer<strong>as</strong> (Fig. 9f), which indicated that the phenotype w<strong>as</strong> not due to an<br />

intrinsic defect of MIF - / - B <strong>cells</strong>. Collectively, these data establish that bmDCs<br />

produce MIF, which is a critical steady-state survival factor <strong>for</strong> mature recirculating B<br />

<strong>cells</strong> in the BM.<br />

32


Figure 9. Mature B cell survival in BM requires MIF-producing bmDCs<br />

(a) Flow cytometry of mature B <strong>cells</strong> in the BM <strong>for</strong> surface expression of components of the MIF<br />

receptor complex CD44 and CD74. Histograms are gated according to dot plot. Open histograms<br />

represent isotype control (CD74) or BM B <strong>cells</strong> of CD44 - / - (CD44).<br />

(b) RT-PCR analysis of MIF expression by monocytes (Mono), macrophages, DCs and unfractionated<br />

<strong>cells</strong> (UF) from BM. Cells are defined <strong>as</strong> described in Fig. 1c and sorted accordingly.<br />

(c) Flow cytometry of B220 + CD93 - CD23 + mature B <strong>cells</strong> from the spleen and BM of MIF - / - mice and<br />

age-matched wt controls. Bar diagram summarizes results obtained from representative experiment of<br />

two (3 mice/group).<br />

34


(d) Flow cytometry of splenic DCs in control [MIF - / - / CD11c-DTRtg > wt] BM chimer<strong>as</strong>, left<br />

untreated or treated with 5 daily intraperitoneal DTx injections, be<strong>for</strong>e analysis on day 6. Note<br />

conditional ablation of DTR/GFP + MIF + / + DCs.<br />

(e) Flow cytometry of BM and spleen B cell compartment of [wt > wt], [CD11c-DTRtg > wt] and<br />

[MIF - / - / CD11c-DTRtg > wt] BM chimer<strong>as</strong>. Bar diagram summarizes results obtained from two<br />

representative experiments of two (3 mice/group). Note inclusion of data of [BAFF - / - /CD11c-DTRtg<br />

> wt] chimer<strong>as</strong> in this experiment that did not show mature B cell loss in response to bmDC depletion.<br />

(f) Flow cytometry of the distribution of MIF - / - B cell (CD45.1 - ) and MIF + / + B <strong>cells</strong> (CD45.1 + ) in the<br />

BM B cell compartment. Bar diagram summarizes results obtained from two representative<br />

experiments of two (4 mice/group). Note that CD11c-DTRtg and MIF - / - B <strong>cells</strong> are equally affected by<br />

the DC depletion.<br />

2.3 Discussion<br />

Here we have reported the phenotypic and functional characterization of the dendritic<br />

cell compartment in unmanipulated murine BM. We found bmDCs were organized<br />

into unique periv<strong>as</strong>cular clusters that wrapped distinct blood vessels. Moreover, the<br />

bmDCs seemed to mark unique BM immune niches, <strong>as</strong> the clusters were seeded by<br />

recirculating B and T <strong>cells</strong>. Conditional ablation of bmDCs resulted in the specific<br />

loss of the mature B <strong>cells</strong> from the niches, <strong>as</strong> B cell maintenance required the<br />

presence of bmDCs that expressed the survival factor MIF.<br />

The mammalian BM is the major <strong>site</strong> of adult hematopoiesis. The advent of advanced<br />

imaging studies h<strong>as</strong> led to the identification of unique niches that provide a highly<br />

specialized microenvironment <strong>for</strong> distinct developmental processes. These include<br />

anatomically defined niches <strong>for</strong> hematopoietic stem <strong>cells</strong> [68,70], B cell development<br />

[59] and thrombopoiesis [70]. A far less appreciated role of the BM is its involvement<br />

in the generation of adaptive immune responses to blood-borne antigens. Naïve B and<br />

T <strong>cells</strong> can be found in the BM, and have been shown to undergo in situ antigenspecific<br />

activation [55,76]. Moreover, the BM is a major reservoir <strong>for</strong> memory T<br />

<strong>cells</strong>, which undergo b<strong>as</strong>al proliferation [61,75] and can be activated in this <strong>site</strong> [54].<br />

The BM is also known to harbor DCs, however, these <strong>cells</strong> have not been<br />

characterized in depth so far. Here we have defined CD11c high bmDCs functionally <strong>as</strong><br />

myeloid BM <strong>cells</strong> that most efficiently primed naïve alloreactive T <strong>cells</strong> in vitro.<br />

Phenotypic characterization showed that bmDCs had an activated phenotype and<br />

expressed unique markers that distinguished them from their splenic counterpart.<br />

Most notably, multiphoton analysis of the cranial BM of mice containing GFP-labeled<br />

bmDCs (Cx3cr1 gfp/+ mice) revealed that these <strong>cells</strong> were concentrated in unique<br />

periv<strong>as</strong>cular clusters that wrapped a distinct set of sinusoids and venules. Adoptive<br />

transfer of labeled B and T lymphocytes led to the seeding of the DC clusters with<br />

35


grafted <strong>cells</strong>, which suggested that the structures represent previously sought BM<br />

immune niches [76]. Here we focused on unmanipulated naïve mice and it remains to<br />

be shown, if T cell responses that have been <strong>as</strong>signed to the BM, are impaired in<br />

bmDC-depleted mice. The notion that bmDCs are critical APC that initiate BM T cell<br />

responses would be supported by the studies [76] and our in vitro MLR results. The<br />

effect of the bmDC depletion on the generation of BM-resident pl<strong>as</strong>ma <strong>cells</strong> in<br />

response to NP-Ficoll challenge suggests that B <strong>cells</strong> might be primed in the niches.<br />

However, in vivo triggering of B <strong>cells</strong> by antigen-bearing DCs in the BM immune<br />

niches, <strong>as</strong> reported <strong>for</strong> the LNs [100], remains to be shown. An intriguing question<br />

raised by our finding is what restricts the size of the DC clusters and BM immune<br />

niches. Published studies have reported the existence of specialized endothelial<br />

microdomains in the BM v<strong>as</strong>culature that express the adhesion molecule E-selectin<br />

and present the chemokine SDF-1 (CXCL12) [101]. These domains, like the DC<br />

clusters, are found in par<strong>as</strong>agittal locations in the skull and were shown to abruptly<br />

terminate. If these domains represent the entry ports to the BM immune niches, they<br />

might provide longitudinal restrictions. The widths of the niches could be determined<br />

by distinct stromal cell cuffs on selected blood vessels. The importance of stromal cell<br />

subsets in the organization of lymphoid organs h<strong>as</strong> been demonstrated in studies of<br />

LNs [102].<br />

Highlighting the functional importance of bmDCs, their conditional ablation resulted<br />

in the specific loss of both endogenous and adoptively transferred mature B <strong>cells</strong> from<br />

the BM immune niches. This failure to engraft bmDC-depleted BM could be<br />

overcome by the overexpression of the antiapoptotic factor Bcl-2 in mature B <strong>cells</strong>,<br />

which suggested that bmDCs provide a unique survival factor. Studies of mixed BM<br />

chimer<strong>as</strong> subsequently allowed us to show that this factor w<strong>as</strong> MIF. Although we<br />

would like to mention that we did not covered in this study other factors than survival<br />

that could also affect accumulation or migration of circulating B <strong>cells</strong> in the BM.<br />

The cytokine MIF h<strong>as</strong> pleiotropic functions and is important in acute and chronic<br />

inflammatory dise<strong>as</strong>es such <strong>as</strong> septic shock, rheumatoid arthritis, colitis and<br />

atherosclerosis [103]. A critical breakthrough in the understanding of MIF's action<br />

w<strong>as</strong> provided by expression cloning of its cellular receptor complex CD74-CD44<br />

[95]. Importantly, MIF engagement of the receptor complex h<strong>as</strong> been shown to trigger<br />

the antiapoptotic PI3K/-Akt signaling pathway in cultured fibrobl<strong>as</strong>ts, tumor <strong>cells</strong> and<br />

primary B <strong>cells</strong> and thus promote their survival [96,97,98]. Here we provided a first in<br />

36


vivo evidence <strong>for</strong> th<strong>as</strong> MIF activity, by demonstrating that mature B cell maintenance<br />

required MIF-producing bmDCs.<br />

It h<strong>as</strong> been shown that splenic marginal zone B <strong>cells</strong> depend on DCs <strong>for</strong> survival and<br />

differentiation into IgM-secreting pl<strong>as</strong>mabl<strong>as</strong>ts [104]. However, in this c<strong>as</strong>e, the TNF<br />

superfamily members BAFF and APRIL were identified <strong>as</strong> critical factors provided<br />

by the DCs. Using a mixed-BM chimera approach, we have ruled out the possibility<br />

that mature B <strong>cells</strong> in the BM require BAFF-producing DCs; however, it remains to<br />

be determined whether APRIL is involved in the BM immune niche. Such a scenario<br />

could indicate similarities between the splenic marginal zone and the BM immune<br />

niches, both of which receive their antigenic input via the blood and not the<br />

lymphatics and thus likely share the t<strong>as</strong>k to providing systemic immunity to bloodborne<br />

pathogens. Notably, not all mature B <strong>cells</strong> were lost from the bmDC-depleted<br />

BM. This could suggest that other <strong>cells</strong> can provide B <strong>cells</strong> with survival signals in<br />

the BM immune niches. Alternatively, the niches could be inhabited by mature B <strong>cells</strong><br />

of two origins: recirculating B <strong>cells</strong> that matured in the spleen, and B <strong>cells</strong> that<br />

matured in the BM, <strong>as</strong> proposed be<strong>for</strong>e [105]. Given their distinct history, the later<br />

IgD + IgM + B <strong>cells</strong> might be independent of MIF and they might thus persist in the DCdepleted<br />

BM immune niches. Such <strong>cells</strong> would be expected to accumulate in BM<br />

niches of MIF - / - mice, which might explain the less severe B cell loss in these mice<br />

relative to the loss in those with conditional ablation.<br />

Finally, it is noteworthy that in many mutant mice with enhanced B cell signaling,<br />

mature B <strong>cells</strong> fail to accumulate in the BM [106,107,108,109,110]. Although this h<strong>as</strong><br />

been hypothesized to be due to impaired B cell homing [111], our finding that mature<br />

B <strong>cells</strong> in the BM immune niches uniquely depended on a specific survival factor<br />

raises the possibility that local B cell apoptosis might account <strong>for</strong> some of the<br />

phenotypes.<br />

Cumulatively, we have reported here the existence of a novel BM immune niches that<br />

harbored DCs, mature B and T lymphocytes. Beyond their established role <strong>as</strong> APC,<br />

DCs residing in this microenvironment needed to provide a critical cytokine, MIF to<br />

maintain the survival of recirculating B <strong>cells</strong> in the niche. Our identification of the<br />

lymphoid follicle- like BM structure should have a critical effect on future studies<br />

investigating the role of this organ in the immune defense against blood borne<br />

pathogens.<br />

37


3. The bone <strong>marrow</strong> immune niche: <strong>Bone</strong> <strong>marrow</strong> <strong>as</strong> a<br />

<strong>priming</strong> <strong>site</strong> <strong>for</strong> naïve CD8 + T <strong>cells</strong><br />

3.1 Introduction<br />

The unique cellular composition and the architecture of the T cell area<br />

microenvironment of secondary lymphoid organs are thought to be prerequi<strong>site</strong>s <strong>for</strong><br />

primary T cell responses [112]. It is in these places where naive T <strong>cells</strong> encounter<br />

antigen-ladden DCs [113]. The BM is a part of the lymphocyte recirculation network<br />

and similar to spleen, is highly v<strong>as</strong>cularized, but lacks a lymphatic drainage [114]. It<br />

h<strong>as</strong> been shown that the parenchyma of the mouse BM contains 3-8% CD3 + T<br />

lymphocytes [76]. T lymphocyte homing to the BM requires VCAM-1 and ICAM-1<br />

that are constitutively expressed in on the stroma [115]. Furthermore, BM is enriched<br />

with antigen-specific memory T <strong>cells</strong> [61]. CD8 + memory T <strong>cells</strong> in the BM were<br />

shown to undergo more vigorous homeostatic proliferation and respond f<strong>as</strong>ter to<br />

antigen stimulation than those in other tissues [116,117]. The BM accumulates CD8 +<br />

central memory T <strong>cells</strong>, which are more frequent among T <strong>cells</strong> in the BM than<br />

elsewhere [63]. Antigen-specific CD8 <strong>cells</strong> persist in the BM <strong>for</strong> several months after<br />

resolution of acute infection [118]. BM-resident T <strong>cells</strong> are a major obstacle to BM<br />

transplantation, because contaminating T <strong>cells</strong> in the BM allografts can cause graft<br />

versus host dise<strong>as</strong>e (GvHD). On the other hand, T cell depletion from allogenic BM<br />

grafts compromises engraftment [119], because CD8 + T <strong>cells</strong> and DCs facilitate<br />

engraftment [120]. On the b<strong>as</strong>is of these observations, the hypothesis is that BM is not<br />

only a <strong>site</strong> that harbors antigen-experienced memory T <strong>cells</strong>, but also a <strong>site</strong> <strong>for</strong><br />

initiation of naïve T cell responses - a function that is generally restricted to secondary<br />

lymphoid organs. The existence of bmDC clusters that contain mature B and T<br />

lymphocytes could indicate that the BM displays structural feature of a secondary<br />

lymphoid organs that contain follicle-like structures. The BM represents a major part<br />

of the lymphocyte recirculation network [115], with billions of lymphocytes<br />

recirculating through it per day. Thus, understanding whether the identified niches<br />

harbor similar function of secondary lymphoid organs, i.e. initiation of primary T cell<br />

responses, can be of major clinical importance.<br />

38


3.2 Results<br />

3.2.1 Identification of bmDC clusters that contain B <strong>cells</strong> in the BM of the long<br />

bones<br />

We have shown that in the cranial BM parenchyma bmDCs are organized into unique<br />

periv<strong>as</strong>cular clusters that occupy structurally definable niches and contain B <strong>cells</strong> and<br />

T <strong>cells</strong>. However, it remained unclear whether similar DC niches exist in the BM of<br />

the long bones. Since these bones are not accessible to intravital two-photon<br />

microscopy, we per<strong>for</strong>med cl<strong>as</strong>sical immunohistochemistry of femura fragments of<br />

Cx3cr1 gfp/+ mice. As seen in Fig. 10A we identified DC clusters reminiscent of the<br />

ones detected in the skull according to CD11c expression, CX3CR1-GFP<br />

fluorescence and characteristic spindle-shape DC morphology (Fig. 10A). BmDC<br />

clusters could mainly be visualized in the subendosteal region of BM cavities (Fig.<br />

10B) that is known to be populated by most primitive stem <strong>cells</strong> [68]. Moreover,<br />

staining <strong>for</strong> B <strong>cells</strong> revealed that these <strong>cells</strong> were located in close contacts with<br />

bmDCs (Fig. 10C). Furthermore, all bmDC clusters were found densely populated by<br />

B lymphocytes (Fig. 10D). In addition, preliminary results indicate that bmDC<br />

depletion in CD11c-DTRtg Cx3cr1 gfp/+ double-transgenic mice leads to a reduction of<br />

the B220 + B <strong>cells</strong> (Fig. 10E). These results suggest that the organization of bmDCs is<br />

similar in the BM of skull and the long bones. These bmDCs reside in close proximity<br />

to B <strong>cells</strong> and are needed <strong>for</strong> their survival in the niche.<br />

39


A<br />

B<br />

C<br />

D<br />

E<br />

DTx<br />

Figure 10. bmDCs in the long bones are organized in clusters containing B <strong>cells</strong><br />

(A) Immunohistochemistry of femura fragment from Cx3cr1 gfp/+ mouse (bmDCs-green), stained with<br />

DAPI (blue), anti-CD11c antibody (red) at x40 magnification.<br />

(B) Fluorescence microscopy analysis of Cx3cr1 gfp/+ femura (bmDCs-green) at x10 magnification.<br />

(C) Fluorescence and immunostaining microscopic analysis of Cx3cr1 gfp/+ femura section, stained with<br />

anti-B220 (red), (bmDCs-green) at x40 magnification.<br />

(D) Fluorescence and immunostaining microscopy analysis of Cx3cr1 gfp/+ femura section, stained with<br />

anti-B220 (blue), (bmDCs-green) at x20 magnification.<br />

(E) Fluorescence and immunostaining microscopy analysis of femura section from untreated CD11c-<br />

DTR Cx3cr1 gfp/+ double-transgenic mouse and 1 d after i.p. injection of DTx. Sections are stained with<br />

anti-B220 antibody (blue), (bmDCs-green) at x20 magnification.<br />

40


3.2.2 BmDCs originate without monocytic intermediate from macrophage/DC<br />

precursors (MDPs)<br />

Macrophage/DC precursors (MDPs), defined <strong>as</strong> cKit + CD11b - Cx3cr1 gfp/+ Gr-1 - BM<br />

<strong>cells</strong> [45], act <strong>as</strong> in vivo precursors of both BM and blood Gr-1 high and Gr-1 low<br />

monocytes [50]. We previously showed that Gr-1 high monocytes differentiate under<br />

resting conditions into mucosal DCs in the intestinal lamina propria [50] and in the<br />

lungs [84]. However, monocytes do not give rise to cl<strong>as</strong>sical splenic CD11c hi DCs<br />

[50,121], which arise from MDPs via circulating non-monocytic precursors, the<br />

preDCs [5,50]. To investigate the origin of bmDCs, we took advantage of CD11c-<br />

DTR Cx3cr1 gfp/+ double-transgenic mice. In these mice, systemic injection of DTx<br />

depleted endogenous bmDCs from the niches (Fig. 11A, middle panel), but spared<br />

round-shaped monocytes. The ablation w<strong>as</strong> however transient, since already four days<br />

after DTx treatment, we found the niches reconstituted with bmDCs (Fig. 11A, right<br />

panel). To identify the bmDC precursors, we adoptively transferred sorted MDPs<br />

(cKit + CD11b - Gr-1 - Cx3cr1 gfp/+ ) or monocytes (cKit - CD11b + Gr-1 + Cx3cr1 gfp/+ or<br />

cKit - CD11b + Gr-1 - Cx3cr1 gfp/+ high) from Cx3cr1 gfp/+ mice into CD11c-DTRtg mice,<br />

that were depleted of their endogenous bmDCs. To prevent bmDC regeneration from<br />

endogenous progenitors the mice were repeatedly injected with DTx, until they were<br />

sacrificed <strong>for</strong> analysis (Fig. 11B). Notably, neither transferred Gr-1 high , nor Gr-1 low<br />

monocytes gave rise to bmDCs (Fig. 11C, two left panels), where<strong>as</strong> in the same<br />

experiment, the Gr-1 high monocytes efficiently reconstituted intestinal lamina propria<br />

DCs (Fig. 11C, third panel from the left). In stark contr<strong>as</strong>t, the MDP transfer resulted<br />

in the efficient reconstitution of bmDCs (Fig. 11C, right panel). Collectively, these<br />

findings show that bmDCs, similarly to their splenic counterparts arise from MDPs<br />

and not from monocytes <strong>as</strong> does a large fraction of DCs in non-lymphoid organs.<br />

41


A<br />

be<strong>for</strong>e DTx<br />

24h after DTx<br />

4d after DTx<br />

B<br />

Day 1 2 3 …………………...….. 14<br />

DTx<br />

DTx DTx analysis<br />

Monocyte/<br />

MDP<br />

transfer into<br />

DTR chimer<strong>as</strong><br />

C<br />

Gr-1 low monocytes<br />

Gr-1 high monocytes<br />

Lamina Propria –<br />

monocyte transfer<br />

MDP transfer<br />

Figure 11. bmDCs arise without monocytic intermediate from MDPs<br />

(A) Two-photon microscopy of cranial BM cavities of CD11c-DTRtg Cx3cr1 gfp/+ mice left untreated or<br />

24 h and 4 d after i.p. DTx injection. Note that 24 h after DTx treatment, monocytes (green, roundshape)<br />

define the BM niche in absence of DCs (green, spindle-shape).<br />

(B) Schedule used to determine the origin of bmDCs. CD11c-DTRtg chimer<strong>as</strong> were treated with DTx 1<br />

d be<strong>for</strong>e transfer of sorted Cx3cr1 gfp/+ monocytes or MDP. Then, every second day, the mice were<br />

injected with DTx and cranial BM cavities and lamina propria were analyzed on day 14.<br />

(C) Cranial BM cavities and lamina propria of DTx treated CD11c-DTRtg chimer<strong>as</strong> 14 d after transfer<br />

of Cx3cr1 gfp/+ monocytes or MDP.<br />

42


3.2.3 In the steady-state T lymphocytes are more motile than B <strong>cells</strong> in the bmDC<br />

niche<br />

Although secondary T-cell responses in the BM are well established, the motility of<br />

naïve T <strong>cells</strong>, their positioning in specific niches and primary immune responses to<br />

blood-borne antigens in this organ have yet to be explored. After exiting the thymus<br />

or the BM, naïve T <strong>cells</strong> and B <strong>cells</strong> search <strong>for</strong> an antigen by recirculation between<br />

blood and secondary lymphoid organs. Using intravital imaging, it h<strong>as</strong> been reported<br />

that T <strong>cells</strong> in the LNs move at high velocities [122]. Their trajectories are chaotic and<br />

random in absence of appropriate antigen [122]. To investigate naïve T and B cell<br />

motility in the BM under the steady-state conditions, we transferred 5x10 7 CMTMRlabeled<br />

T <strong>cells</strong> (with equal ratio of CD4 + and CD8 + T <strong>cells</strong>) and CFP + B <strong>cells</strong> (isolated<br />

from β-actin-Enhanced Cyan Fluorescent Protein transgenic mice) into Cx3cr1 gfp/+<br />

mice, and examined the motility of the <strong>cells</strong> 4 h later by intravital microscopy. We<br />

observed that grafted T and B <strong>cells</strong> dispersed equally in the BM (Fig. 12A, Movie 1).<br />

Time-lapse imaging and tracking of individual <strong>cells</strong> in three dimensions over 20-45<br />

min showed that T cell trajectories are longer and more random than those of B <strong>cells</strong><br />

(Fig. 12A, Movie 1). Quantification revealed that T <strong>cells</strong> crawled with mean velocity<br />

of 3.6 μm per minute and their displacement rate w<strong>as</strong> 2 μm per minute, where<strong>as</strong> B cell<br />

mean velocity w<strong>as</strong> 2 μm per minute and displacement rate w<strong>as</strong> 1 μm per minute (Fig.<br />

12B). Next, we analyzed the differences in motility of naïve CD8 + T <strong>cells</strong> only versus<br />

mature B lymphocytes. The me<strong>as</strong>urement of velocity and displacement rate showed<br />

again that T <strong>cells</strong> are more motile than their B cell counterparts (with mean velocity<br />

of 6.3 versus 4.05 μm per minute; and displacement rate of 3.6 versus 2.02 μm per<br />

minute, respectively) (Fig. 12C, Movie 2). The observation of Movies 1, 2 revealed<br />

that most of the B <strong>cells</strong> are in close contacts with DCs that are display frequent shape<br />

changes and dendrite extensions. Taken together, our analysis showed that although in<br />

these setting, no antigen is added to the system, B <strong>cells</strong> movement is confined;<br />

where<strong>as</strong> T <strong>cells</strong> display significant and random cell movement within the BM, similar<br />

to their motility in the LNs.<br />

43


A<br />

T cell (yellow)<br />

B cell (blue)<br />

B<br />

Velocity (µ/min)<br />

27.5<br />

25.0<br />

22.5<br />

20.0<br />

17.5<br />

15.0<br />

12.5<br />

10.0<br />

7.5<br />

5.0<br />

2.5<br />

0.0<br />

T <strong>cells</strong><br />

P=0.0009<br />

B <strong>cells</strong><br />

Displacement rate (µ/min)<br />

20.0<br />

17.5<br />

15.0<br />

12.5<br />

10.0<br />

7.5<br />

5.0<br />

2.5<br />

0.0<br />

T <strong>cells</strong><br />

P


Figure 12. T lymphocytes are more motile than B <strong>cells</strong> in the bmDC niche under steady-state<br />

conditions<br />

(A) Time lapse images of CMTMR-labeled wild type T <strong>cells</strong> (yellow) and CFP + B <strong>cells</strong> (blue)<br />

transferred into Cx3cr1 gfp/+ mice (bmDCs – green), BM microv<strong>as</strong>culature w<strong>as</strong> delineated by i.v.<br />

injection of Quantum dots (red). Tracks of one T cell and one B cell (white circles) at different time<br />

points. The l<strong>as</strong>t images on the right show the entire track (white line).<br />

(B) Velocity and displacement rate of transferred T (total CD4 + and CD8 + ) and B <strong>cells</strong>. Data points<br />

represent individual <strong>cells</strong> (n of T <strong>cells</strong> = 228, n of B <strong>cells</strong> = 178).<br />

(C) Velocity and displacement rate of transferred CD8 + T <strong>cells</strong> and B <strong>cells</strong>. Data points represent<br />

individual <strong>cells</strong> (n of T <strong>cells</strong> = 236, n of B <strong>cells</strong> = 168).<br />

3.2.4 Naïve CD8 + T <strong>cells</strong> <strong>for</strong>m multi-cell clusters in the bmDC niches upon bloodborne<br />

antigen stimulation<br />

In the LNs, T <strong>cells</strong> home to a specific area, termed T cell zone, to which DCs that<br />

captured antigens in peripheral tissue migrate. A naïve T cell spends an average of 24<br />

hours in a given LN, but its duration is extended to 3-4 days when the T cell is<br />

exposed to its cognate antigen [122]. If a T cell encounters a DC presenting its<br />

cognate peptide, the T cell arrests on the DC and a stable contact is established,<br />

l<strong>as</strong>ting <strong>for</strong> 36-48h to fully activate T cell [123]. Although we have a considerable<br />

understanding of T cell motility, their interaction with DCs and activation in the LNs,<br />

these processes in the BM remain poorly defined. There<strong>for</strong>e, we decided to study<br />

motility of naive CD8 + T <strong>cells</strong> within the BM in detail, focusing first on the initial<br />

steps leading to T cell activation, by examining T <strong>cells</strong> motility upon antigen<br />

administration. To test whether CD8 + T <strong>cells</strong> can sense antigen presented by <strong>cells</strong> in<br />

the BM, we purified naïve OVA-specific TCR transgenic CD8 + T <strong>cells</strong> from the<br />

spleens of OT-I mice (Fig. 13A) and labeled them with CMTMR dye. Cx3cr1 gfp/+<br />

recipient mice were engrafted with 5x10 7 labeled CD8 + T <strong>cells</strong>. Four hours later, the<br />

mouse w<strong>as</strong> prepared <strong>for</strong> intravital imaging of the BM and quantum dots were injected<br />

just be<strong>for</strong>e beginning of imaging to visualize blood vessels. After the start of the<br />

recording, 0.5 mg SIINFEKL peptide and FITC-dextran, were i.v. coinjected to the<br />

recipient. Coinjection of FITC-dextran into the blood stream facilitates the<br />

observation of the peptide effect in real-time <strong>as</strong> it diffuses. Peptide administration<br />

resulted in delineation of BM microv<strong>as</strong>culature in green color and immediate<br />

immobilization of the majority of CD8 + OT-I <strong>cells</strong> (Movie 3). Analysis of one<br />

representative movie using computerized 3D tracking software demonstrated that T<br />

<strong>cells</strong> covered a longer distance be<strong>for</strong>e the administration of the peptide than following<br />

its injection (Fig. 13B). Velocity analysis showed that most of the <strong>cells</strong> that were<br />

motile at the beginning of the movie, moved at a significantly slower rate after OVA<br />

45


peptide administration (Fig. 13C). We could observe that T cell immobilization w<strong>as</strong><br />

general and not restricted to the specific are<strong>as</strong> where DCs reside. This is likely, since<br />

the peptide does not have to be processes by APC, but is extracellulary loaded on all<br />

MHC cl<strong>as</strong>s I expressing <strong>cells</strong> [124]. There<strong>for</strong>e, it is expected that OT-I T <strong>cells</strong><br />

immediately stop in vicinity of <strong>cells</strong> that bound the peptide.<br />

A<br />

OT-I <strong>cells</strong><br />

99%<br />

CD8<br />

CD62L<br />

B<br />

FSC<br />

be<strong>for</strong>e immunization<br />

CD44<br />

after immunization<br />

C<br />

50<br />

Instantaneous velocity (μm/min)<br />

45<br />

40<br />

35<br />

30<br />

25<br />

20<br />

15<br />

10<br />

OVA peptide<br />

+FITC-dextran<br />

injection<br />

5<br />

0<br />

0 8.5 17 25.5 34 42.5 51<br />

Time (min)<br />

46


Figure 13. Motility of naïve CD8 + T <strong>cells</strong> upon peptide immunization<br />

(A) Flow cytometry of enriched naïve CD8 + T <strong>cells</strong> by immunomagnetic cell separation with anti-CD8<br />

beads. Staining with anti-CD62L and anti-CD44 shows that 99% of the purified <strong>cells</strong> are naïve.<br />

(B) Time lapse images of CMTMR-labeled wild type T <strong>cells</strong> (yellow) transferred into Cx3cr1 gfp/+ mice<br />

(bmDCs – green), be<strong>for</strong>e (left panel) and after i.v. injection of 0.5 mg OVA peptide and FITC-dextran<br />

(right panel). Note that BM microv<strong>as</strong>culature, be<strong>for</strong>e the immunization, w<strong>as</strong> delineated by Quantum<br />

dots (red) and upon immunization by FITC-dextran (green). Tracks of T <strong>cells</strong> are in purple. The l<strong>as</strong>t<br />

images on the right show the entire track (white line).<br />

(C) Instantaneous velocity of individual T <strong>cells</strong> be<strong>for</strong>e and after peptide immunization. Note the<br />

incre<strong>as</strong>e in the velocity at the time point of peptide injection and then robust decre<strong>as</strong>e.<br />

Next, we decided to study naïve CD8 + T cell <strong>priming</strong> within the BM, focusing on the<br />

first steps to T cell activation after intact antigen administration.<br />

We purified naïve OVA-specific TCR transgenic CD8 + T <strong>cells</strong> from the spleens of<br />

OT-I mice (CD45.1) and transferred them into CD45.2 WT recipients that 16h later<br />

were immunized with 0.5 mg OVA protein. Two hours after immunization, spleen,<br />

LNs and BM of the mice were <strong>as</strong>sessed <strong>for</strong> the activation status of OT-I <strong>cells</strong> by<br />

me<strong>as</strong>uring CD69 expression. CD69 is an early membrane receptor, which is rapidly<br />

and transiently expressed on lymphocytes during activation and is not detected on<br />

resting lymphocytes [125]. Interestingly, analysis of naive OT-I donor mice had<br />

revealed that the b<strong>as</strong>al levels of CD69 expression on CD8 T <strong>cells</strong> in the BM, were<br />

higher than in spleen or LNs (Fig. 14A). In addition, after antigen challenge, CD69<br />

expression on the transferred OT-I <strong>cells</strong> in the BM w<strong>as</strong> notably higher than in other<br />

organs (Fig. 14A). These results are in agreement with the findings of Feuerer et al.<br />

[76]. To visualize the change in T cell behavior after antigen challenge, we next<br />

immunized Cx3cr1 gfp/+ mice i.v. with 0.5 mg of OVA protein and 24h later<br />

transferred into these mice a mix of two populations of naïve CD8 + T <strong>cells</strong> and<br />

polyclonal CD8 + T <strong>cells</strong> purified from CFP + mice and CMTMR-labeled OT-I T <strong>cells</strong><br />

(5x10 7 each). Four hours later, the skull BM of the recipient mice w<strong>as</strong> analyzed by<br />

intravital imaging. Three-dimensional reconstitutions generated by time-lapse images<br />

revealed that solely OT-I <strong>cells</strong> clustered in are<strong>as</strong> enriched with DCs (Fig. 14B, Movie<br />

4). This suggested that bmDCs efficiently captured the exogenous antigen and rapidly<br />

processed and presented it to the specific T <strong>cells</strong>. Polyclonal T <strong>cells</strong> that served <strong>as</strong><br />

internal control, moved with higher velocities (11 μm per minute compared to 4 μm<br />

per minute) and displacement rate (5 μm per minute compared to 1 μm per minute)<br />

than OT-I T <strong>cells</strong> that responded to their cognate antigen and arrested (Fig. 14C). In<br />

addition, the trajectories of the polyclonal T cell movement were random and spread,<br />

where<strong>as</strong> OT-I T cell behavior w<strong>as</strong> more restricted and showed a notable antigen-<br />

47


dependent suppression in motility (Fig. 14D). These results indicate that the BM is<br />

accessible to blood-borne antigens, which can be processed and presented by BMresident<br />

<strong>cells</strong>, resulting in early activation of T <strong>cells</strong>. Moreover, our findings establish<br />

that T <strong>cells</strong> arrest in the BM niches during antigenic challenge and this arrest leads to<br />

their activation.<br />

A<br />

CD45.1<br />

B<br />

CD8<br />

CD8<br />

CD69<br />

MFI of CD69 expression<br />

90<br />

80<br />

no treatment<br />

70<br />

OVA<br />

60<br />

50<br />

40<br />

30<br />

20<br />

10<br />

0<br />

BM SP LNs<br />

48


C<br />

Velocity (µ/min)<br />

22.5<br />

20.0<br />

17.5<br />

15.0<br />

12.5<br />

10.0<br />

7.5<br />

5.0<br />

2.5<br />

0.0<br />

Polyclonal<br />

T <strong>cells</strong><br />

P


ate of specific OT-I T <strong>cells</strong> w<strong>as</strong> significantly lower than of polyclonal T <strong>cells</strong> upon<br />

immunization with the antigen and regardless of DC depletion (Fig. 15B). Moreover,<br />

when OT-I T <strong>cells</strong> were tested <strong>for</strong> CD69 expression by flow cytometry, BM-resident<br />

OT-I <strong>cells</strong> in DC depleted hosts expressed comparable levels of CD69 to DC-bearing<br />

recipients (Fig. 15C). Interestingly, analysis of splenic OT-I <strong>cells</strong> served <strong>as</strong> internal<br />

control <strong>for</strong> efficient DC depletion, <strong>as</strong> in this organ, depletion of DCs significantly<br />

impaired CD69 expression on T <strong>cells</strong> (Fig. 15C). To verify that naïve CD8 + T <strong>cells</strong><br />

were primed in the BM in absence of DCs, we transferred CFSE-labeled OT-I<br />

CD45.1 T <strong>cells</strong> into control mice and mice that were treated with DTx throughout the<br />

experiment. The mice were immunized with OVA and 3 d after immunization spleen<br />

and BM were isolated and analyzed <strong>for</strong> the proliferation status of the grafted T <strong>cells</strong>.<br />

The transferred T <strong>cells</strong> readily proliferated in all organs of DC-proficient mice in<br />

response to immunization with OVA (Fig. 5D). As reported previously, the DTxinduced<br />

DC depletion abrogated the expansion of CD8 + T <strong>cells</strong> in the spleen.<br />

Surprisingly, however, we observed a proliferate response of the grafted T <strong>cells</strong> in the<br />

DC-depleted BM (Fig. 5D). Furthermore, since proliferative T cell expansion can be<br />

<strong>as</strong>sociated with the generation of cytokine-producing effector T <strong>cells</strong>, we investigated<br />

polarization of CD8 + T <strong>cells</strong> in DC-depleted BM. To analyze T cell polarization of the<br />

grafted OT-I <strong>cells</strong>, we per<strong>for</strong>med an in vivo restimulation by rechallenging the OVAimmunized<br />

mice with an i.v. injection of OVA peptide [126]. In presence of DCs,<br />

CD8 + T <strong>cells</strong> in the spleen and in the BM differentiated into IFN-γ-producing <strong>cells</strong><br />

(Fig. 5E). Depletion of DCs, impaired IFN-γ secretion in the spleen, but absence of<br />

DCs did not affect the differentiation of CD8 + T <strong>cells</strong> into effector <strong>cells</strong> in the BM<br />

(Fig. 5E). Collectively, our data show that BM contains APCs other than bmDCs that<br />

are capable of stimulating naïve CD8 + T <strong>cells</strong>. Although DCs are known to be the<br />

most potent cross-presenting <strong>cells</strong> [42,127], it could be that in the unique BM<br />

microenvironment, other <strong>cells</strong>, such <strong>as</strong> B <strong>cells</strong>, macrophages, neutrophils, stromal<br />

<strong>cells</strong> or other MHC cl<strong>as</strong>s I expressing <strong>cells</strong> would be able to per<strong>for</strong>m the t<strong>as</strong>k.<br />

50


A<br />

DTx<br />

B<br />

Velocity (µ/min)<br />

13<br />

12<br />

11<br />

10<br />

9<br />

8<br />

7<br />

6<br />

5<br />

4<br />

3<br />

2<br />

1<br />

0<br />

P


D<br />

no immunization<br />

CD45.1<br />

CD8<br />

OVA DTx OVA<br />

SP<br />

counts<br />

CFSE<br />

BM<br />

E<br />

SP<br />

non treated OVA DTx OVA<br />

CD45.1<br />

CD8<br />

1% 21%<br />

5%<br />

CD8<br />

BM<br />

IFN gamma<br />

CD45.1<br />

CD8<br />

CD8<br />

1% 5%<br />

6%<br />

IFN gamma<br />

Figure 15. Naïve CD8 + T <strong>cells</strong> are efficiently activated in BM niches in absence of DCs.<br />

(A) Two-photon imaging of CMTMR-labeled OT-I T <strong>cells</strong> (yellow) and CFP + polyclonal T <strong>cells</strong> (blue)<br />

transferred into CD11c-DTR Cx3cr1 gfp/+ mice that were untreated or injected with DTx twice, 24 h and<br />

3 h prior to the transfer. Then, 6 h after the first DTx injection, recipient mice were immunized with<br />

0.5mg OVA. Note that after DTx treatment, monocytes (green, round-shape) define the BM niche in<br />

absence of DCs (green, spindle-shape).<br />

(B) Velocity and displacement rate of CMTMR-labeled OT-I T <strong>cells</strong> (yellow) and CFP + polyclonal T<br />

<strong>cells</strong> (blue) transferred into CD11c-DTR Cx3cr1 gfp/+ mice that were treated <strong>as</strong> described in (A). Data<br />

points represent individual <strong>cells</strong> (n of polyclonal T <strong>cells</strong> = 37, n of OT-I <strong>cells</strong> = 45).<br />

(C) Flow cytometry of CD69 expression of transferred OT-I CD45.1 naïve T <strong>cells</strong> into CD11c-DTRtg<br />

chimer<strong>as</strong> that were left untreated or were injected with DTx and then, and 2 h be<strong>for</strong>e the analysis were<br />

immunized with 0.5mg OVA. The bar graph represents mean fluorescent intensity (MFI) of CD69<br />

expression in the spleen and BM.<br />

(D) Flow cytometry of transferred CFSE-labeled CD45.1 OT-I <strong>cells</strong> into untreated or DTx-injected<br />

CD11c-DTRtg chimer<strong>as</strong>, 3 d after i.v. immunization with 0.5 mg OVA. Histograms represent <strong>cells</strong><br />

gated according to scatter, CD45.1 and CD8 expression, <strong>as</strong> indicated in the dot plots.<br />

52


(E) CD45.1 OT-I <strong>cells</strong> were transferred into CD11c-DTRtg chimer<strong>as</strong>. One day later, mice were treated<br />

with DTx and 8 hours later i.v. immunized with OVA and CpG. Three days after immunization, INF-γ<br />

production in the spleen and BM w<strong>as</strong> <strong>as</strong>sessed after in vivo restimulation with OVA peptide, 2 h be<strong>for</strong>e<br />

sacrificing the mice. Data represent INF-γ profiles after gating on CD45.1 and CD8 lymphocytes.<br />

3.2.6 Naïve CD8 + T <strong>cells</strong> fail to <strong>for</strong>m multi-cell clusters in mice harboring MHC<br />

cl<strong>as</strong>s-I-deficient stroma<br />

Few studies demonstrated the capacity of nonhematopoietic <strong>cells</strong>, i.e. stromal <strong>cells</strong> to<br />

present antigens and stimulate CD8 + T <strong>cells</strong>. For instance, after ingestion, oral<br />

antigens distribute systemically and provoke T cell stimulation outside the<br />

g<strong>as</strong>trointestinal tract. Within the liver, scavenger liver sinusoidal endothelial <strong>cells</strong><br />

cross-present oral antigens on H-2K b to CD8 + T <strong>cells</strong> [128]. In other study, osteocl<strong>as</strong>ts<br />

that resorb the bone throughout the life of an animal, in presence of OVA induce<br />

proliferation of CD8 + T <strong>cells</strong> [129]. Given the notion that BM is populated by high<br />

variety of stromal <strong>cells</strong> and that bmDCs are not crucial <strong>for</strong> generation of T cell<br />

clusters after immunization, we decided to test the possibility that stromal <strong>cells</strong> are<br />

capable to do so. To this end, we generated BM chimer<strong>as</strong> by the reconstitution of<br />

lethally irradiated H-2K b -deficient recipient mice [130] with Cx3cr1 gfp/+ BM<br />

[Cx3cr1 gfp/+ > H-2K b- / - ]. These mice contain <strong>cells</strong> of hematopoietic origin (e.g.,<br />

macrophages, DCs, and B <strong>cells</strong>) that are capable of presenting antigenic peptides on<br />

MHC cl<strong>as</strong>s I molecules, where<strong>as</strong> the radiation-resistant <strong>cells</strong>, such <strong>as</strong> stromal <strong>cells</strong>,<br />

lack the capacity to express MHC cl<strong>as</strong>s I molecules. The chimer<strong>as</strong> were transferred<br />

with equal amounts of purified polyclonal CFP + and CMTMR-labeled OT-I CD8 + T<br />

<strong>cells</strong>, and immunized with OVA protein. Analysis of the behavior of transferred T<br />

<strong>cells</strong>, unexpectedly, revealed that MHC cl<strong>as</strong>s I deprivation on stromal <strong>cells</strong> w<strong>as</strong><br />

sufficient to impair <strong>for</strong>mation of OT-I T cell clusters [Fig. 16, Movie 7]. Although<br />

these are only preliminary results, and analysis of both DC-depleted [CD11c-DTRtg<br />

Cx3cr1 gfp/+ > H-2K b - / - ] chimer<strong>as</strong> and reciprocal [H-2K b - / - > CD11c-DTRtg<br />

Cx3cr1 gfp/+ ] chimer<strong>as</strong> will be required, our results suggest that stromal <strong>cells</strong> in the<br />

BM are essential <strong>for</strong> CD8 + T cell <strong>priming</strong> in the BM.<br />

53


Figure 16. MHC cl<strong>as</strong>s I deprivation on stromal <strong>cells</strong> impairs <strong>for</strong>mation of OT-I <strong>cells</strong> after<br />

immunization with antigen<br />

Two-photon imaging of CMTMR-labeled OT-I T <strong>cells</strong> (yellow) and CFP + polyclonal T <strong>cells</strong> (blue)<br />

transferred into [Cx3cr1 gfp/+ > H-2K b - / - ] chimer<strong>as</strong> that were 24 h prior to transfer immunized with<br />

0.5mg OVA. T <strong>cells</strong> were analyzed 4 h after transfer. BM microv<strong>as</strong>culature w<strong>as</strong> delineated by<br />

Quantum dots (red).<br />

3.3 Discussion<br />

After birth, the BM is the principal <strong>site</strong> of hematopoiesis in mammals and thus, it is<br />

mainly considered to serve <strong>as</strong> a primary lymphoid organ. However, its potential to<br />

serve <strong>as</strong> a secondary immune organ h<strong>as</strong> hardly been explored.<br />

The BM microenvironment consists of many types of <strong>cells</strong>, which provide unique<br />

cues <strong>for</strong> the development of blood <strong>cells</strong> and <strong>for</strong> the survival and maintenance of HSCs<br />

and mature <strong>cells</strong>, such <strong>as</strong> mature B <strong>cells</strong>, pl<strong>as</strong>ma <strong>cells</strong> and memory T <strong>cells</strong>. It is<br />

believed that to fulfill these different t<strong>as</strong>ks the BM includes functionally distinct<br />

are<strong>as</strong>, termed niches, <strong>for</strong> different processes that take place in this organ. In the<br />

previous study, we phenotypically and functionally characterized a unique population<br />

of BM-resident DC CD11c high MHCII high DCs (bmDCs). Intravital multiphoton<br />

imaging revealed that these bmDCs were concentrated in unique periv<strong>as</strong>cular clusters<br />

enveloping selected blood vessels and seeded with mature B and T lymphocytes, <strong>as</strong><br />

well <strong>as</strong> monocytes. Moreover, we showed that bmDCs are critical <strong>for</strong> the maintenance<br />

of recirculating B <strong>cells</strong> in the BM through the production of MIF. These findings<br />

suggested that the clusters seeded with DCs and lymphocytes function <strong>as</strong> niches.<br />

Here, we investigated the dynamic behavior of mature lymphocytes in the BM and<br />

specifically in the are<strong>as</strong> enriched with DCs and tested the hypothesis that BM is<br />

capable of antigen presentation and initiation of primary T-cell responses – a function<br />

thought to be restricted to secondary lymphoid organs. First, we elucidated the origin<br />

of bmDCs. Interestingly, our results established that like splenic cDCs, bmDCs seem<br />

54


to originate in the steady state exclusively from MDPs, rather than from recirculating<br />

monocytes. However, these results do not exclude the possibility that monocytes may<br />

carry antigen into the BM to support T-cell responses, <strong>as</strong> shown earlier <strong>for</strong> adoptively<br />

transferred DCs [54]. We next focused on naïve T and B lymphocyte motility in<br />

bmDC niches. We discovered that B cell migration is restricted, where<strong>as</strong> T <strong>cells</strong><br />

move more freely and f<strong>as</strong>ter than B <strong>cells</strong> in the BM. Our findings regarding B<br />

lymphocytes are consistent with published observation that B <strong>cells</strong> are organized in<br />

defined niches near blood vessels and do not migrate from one B cell niche to another<br />

[55]. However, this is the first report examining the motility of naïve T <strong>cells</strong> in the<br />

BM. We have found that T <strong>cells</strong> move approximately twice <strong>as</strong> f<strong>as</strong>t <strong>as</strong> B <strong>cells</strong> in the<br />

BM, similarly to their motolity in the LNs [122]. These results suggest that<br />

differences in cell motility are intrinsic to the type of lymphocyte and do not depend<br />

on the specific microenvironment in the organ. The similarities in lymphocyte<br />

behavior in BM and LNs also argue <strong>for</strong> possible shared functional features in both<br />

organs.<br />

A microenvironment <strong>for</strong> efficient induction of a primary immune response requires an<br />

architecture facilitating homing of naïve T <strong>cells</strong> to are<strong>as</strong> of antigen presentation,<br />

thereby allowing efficient scanning of peptide-MHC complexes on DCs by T <strong>cells</strong>.<br />

These conditions seem to be met in the parenchyma of mouse BM, which contains<br />

both T lymphocytes and DCs. Indeed, Feuerer et al. showed that CD8 + T <strong>cells</strong> bearing<br />

a specific TCR can be activated and proliferate in the BM by administration of its<br />

cognate antigen into the circulation [76]. In addition, central memory T <strong>cells</strong> can be<br />

activated in the BM by blood-borne DCs [54]. In another study, mature recirculating<br />

B <strong>cells</strong> in the BM were demonstrated to be activated by Salmonella typhimurium to<br />

generate short-lived pl<strong>as</strong>ma <strong>cells</strong> [55]. These results show that B <strong>cells</strong> can be activated<br />

in the BM in response to blood-borne microbes. We, there<strong>for</strong>e, decided to further<br />

investigate these findings and focused on primary T-cell responses to blood-borne<br />

antigens. We aimed at examining whether naïve T <strong>cells</strong> can respond to blood-borne<br />

antigens presented by DCs in the BM. To address this question, we transferred OT-I<br />

<strong>cells</strong> to mice expressing green fluorescent DCs in the BM. After T cell homing to the<br />

BM, we imaged the mice in are<strong>as</strong> rich in DCs and T <strong>cells</strong> (BM immune niches).<br />

During the imaging session, TCR-specific peptide w<strong>as</strong> administered and an almost<br />

immediate arrest of T <strong>cells</strong> w<strong>as</strong> observed. This T cell arrest w<strong>as</strong> not restricted only to<br />

the immediate vicinity of DCs, since most BM <strong>cells</strong> expresses cognate MHC-<br />

55


molecules, there<strong>for</strong>e the injected peptide can bind to any cell in the BM and<br />

consequently T <strong>cells</strong> stop immediately on a cell that bound the peptide in their<br />

vicinity. To circumvent this obstacle, we took advantage of the ability of DCs to<br />

cross-present extracellular antigens to CD8 + T <strong>cells</strong>. Intravital imaging of the BM<br />

after injecting a specific protein revealed clusters of specific T <strong>cells</strong> in are<strong>as</strong> enriched<br />

with DCs, where<strong>as</strong> polyclonal T <strong>cells</strong> continued to migrate in chaotic and random<br />

f<strong>as</strong>hion. We have confirmed that the change in T cell dynamics (arrest) w<strong>as</strong> indeed<br />

accompanied by cell activation <strong>as</strong> CD69 w<strong>as</strong> upregulated in the BM, even to greater<br />

extent than in the spleen, during antigenic challenge. During steady-state, naïve<br />

lymphocytes continually enter and exit lymphoid organs in a recirculation process that<br />

is essential <strong>for</strong> immune surveillance. In contr<strong>as</strong>t, during immune responses, the egress<br />

process can be shut down transiently. When this occurs locally, it incre<strong>as</strong>es<br />

lymphocyte numbers in the responding lymphoid organ [131]. Lymphocyte egress<br />

requires sphingosine 1-phosphate receptor-1 (S1P1), where<strong>as</strong>, CD69 negatively<br />

regulates S1P1 and promotes lymphocyte retention in lymphoid organs [132].<br />

Interestingly, the b<strong>as</strong>al levels of CD69 in the BM were significantly higher than in<br />

other organs, suggesting that BM CD8 + T <strong>cells</strong> are in an activated state, and they are<br />

retained in the BM in order sample antigens and to respond in situ in c<strong>as</strong>e of antigen<br />

stimulation.<br />

To further establish if bmDCs are critical antigen-presenting <strong>cells</strong> that initiate BM T-<br />

cell responses, we analyzed the responses of naïve CD8 + T <strong>cells</strong> to antigen challenge<br />

in mice that were depleted of DCs. Surprisingly, ablation of bmDCs did not impair the<br />

capacity of antigen-specific T <strong>cells</strong> to cluster. In support of previous reports [42,133],<br />

we found that the capacity to prime CD8 + T <strong>cells</strong> in the spleen is uniquely restricted to<br />

cDCs. Thus, in DC-depleted spleens, T <strong>cells</strong> neither upregulated CD69 expression and<br />

proliferated, nor secreted IFN-γ. However, unexpectedly, in the BM of the same mice,<br />

T <strong>cells</strong> showed prompt CD69 upregulation, in vivo proliferation after antigen<br />

challenge and IFN-γ production.<br />

Lymphoid organs have a highly organized and complex architecture composed of<br />

distinct cellular compartments, at heart of which is a nonhematopoietic cell backbone<br />

[134]. In the LNs, nonhematopoietic microanatomy actively organizes the cellular<br />

encounters by providing pre<strong>for</strong>med migration tracks that create dynamic, but highly<br />

ordered movement patterns [135]. Although it is established that the main function of<br />

56


the stroma is to orchestrate cell migration within lymphoid organs, recently it h<strong>as</strong><br />

been shown that stromal <strong>cells</strong> can cross-present tumor-<strong>as</strong>sociated antigens to CD8 +<br />

cytotoxic T lymphocytes that mediate tumor rejection [136]. Studies of the BM<br />

chimera approach, allowed us to address the question which <strong>cells</strong> activate and prime<br />

naïve CD8 + T <strong>cells</strong> in DC-ablated BM niches. Chimer<strong>as</strong>, in which H-2K b -deficient<br />

recipients were reconstituted with Cx3cr1 gfp/+ BM, the MHC cl<strong>as</strong>s I-deprived stroma<br />

w<strong>as</strong> incapable of sustaining primary CD8 + T-cell response. These data suggest a<br />

novel unique role of nonhematopoietic <strong>cells</strong> in the BM, although a more detailed<br />

study is required to elucidate the nature of these <strong>cells</strong>.<br />

Collectively, our data suggest that BM is unique among lymphoid organs in<br />

per<strong>for</strong>ming primary and secondary immune functions. It contains a microenvironment<br />

that interacts with naïve, circulating antigen-specific T <strong>cells</strong> in absence of antigenpresenting<br />

DCs and leads to induction of primary CD8 + T-cell responses.<br />

57


Movies are added on disk<br />

Movie 1<br />

Two-photon imaging of CMTMR-labeled wild type CD4 + and CD8 + T <strong>cells</strong> (yellow) and CFP + B <strong>cells</strong><br />

(blue) transferred into Cx3cr1 gfp/+ mice (bmDCs – green), blood vessels are stained with Quantum<br />

dots (red).<br />

Movie 2<br />

Two-photon imaging of CMTMR-labeled wild type CD8 + T <strong>cells</strong> (yellow) and CFP + B <strong>cells</strong> (blue)<br />

transferred into Cx3cr1 gfp/+ mice (bmDCs – green), blood vessels are stained with Quantum dots (red).<br />

Movie 3<br />

Two-photon imaging of CMTMR-labeled wild type T <strong>cells</strong> (yellow) transferred into Cx3cr1 gfp/+ mice<br />

(bmDCs – green), be<strong>for</strong>e and after i.v. injection of 0.5 mg OVA peptide and FITC-dextran. Note that<br />

BM microv<strong>as</strong>culature, be<strong>for</strong>e the immunization, w<strong>as</strong> delineated by Quantum dots (red) and upon<br />

immunization by FITC-dextran it became green.<br />

Movie 4<br />

Two-photon imaging of CMTMR-labeled OT-I T <strong>cells</strong> (yellow) and CFP + polyclonal T <strong>cells</strong> (blue)<br />

transferred into Cx3cr1 gfp/+ mice (bmDCs - green) that w<strong>as</strong> 24 h prior to transfer immunized with<br />

0.5mg OVA. T <strong>cells</strong> were analyzed 4 h after transfer. BM microv<strong>as</strong>culature w<strong>as</strong> delineated by<br />

Quantum dots (red).<br />

Movie 5<br />

Two-photon imaging of CMTMR-labeled OT-I T <strong>cells</strong> (yellow) and CFP + polyclonal T <strong>cells</strong> (blue)<br />

transferred into CD11c-DTRtg Cx3cr1 gfp/+ double transgenic mice that were untreated or injected with<br />

DTx twice, 24 h and 3 h prior to the transfer. Then, 6 h after the first DTx injection, recipient mice<br />

were immunized with 0.5mg OVA. Note that after DTx treatment, monocytes (green, round-shape)<br />

define the BM niche in absence of DCs (green, spindle-shape).<br />

Movie 6<br />

Two-photon imaging of CMTMR-labeled OT-I T <strong>cells</strong> (yellow) and CFP + polyclonal T <strong>cells</strong> (blue)<br />

transferred into CD11c-DTRtg Cx3cr1 gfp/+ double transgenic mice that were untreated or injected with<br />

DTx twice, 24 h and 3 h prior to the transfer. Then, 6 h after the first DTx injection, recipient mice<br />

were immunized with 0.5mg OVA. Note that after DTx treatment, monocytes (green, round-shape)<br />

define the BM niche in absence of DCs (green, spindle-shape).<br />

Movie 7<br />

Two-photon imaging of CMTMR-labeled OT-I T <strong>cells</strong> (yellow) and CFP + polyclonal T <strong>cells</strong> (blue)<br />

transferred into [CD11c-DTRtg Cx3cr1 gfp/+ > H-2K b - / - ] chimer<strong>as</strong> that were 24 h prior to transfer<br />

immunized with 0.5mg OVA. T <strong>cells</strong> were analyzed 4 h after transfer. BM microv<strong>as</strong>culature w<strong>as</strong><br />

delineated by Quantum dots (red).<br />

58


4. Organ-dependent in vivo <strong>priming</strong> of naïve CD4 + , but not<br />

CD8 + T <strong>cells</strong> by pl<strong>as</strong>macytoid dendritic <strong>cells</strong><br />

4.1 Introduction<br />

T lymphocytes respond to antigenic peptides presented on major MHC molecules.<br />

Cytotoxic CD8 + T <strong>cells</strong> recognize peptides in the context of MHC cl<strong>as</strong>s I, where<strong>as</strong><br />

CD4 + helper T <strong>cells</strong> recognize peptides in complex with MHC cl<strong>as</strong>s II. Naïve T cell<br />

activation is restricted to secondary lymphoid organs, such <strong>as</strong> the spleen and LNs, and<br />

thought to rely on specialized APCs comprising B <strong>cells</strong>, macrophages and DCs. The<br />

APC composition of lymphoid organs likely encodes a potential to initiate distinct T<br />

cell responses. However, the differential contribution of APC to in vivo immune<br />

responses remains poorly understood.<br />

Natural IFN-producing <strong>cells</strong> (IPCs) are a unique subset of hematopoetic <strong>cells</strong> that play<br />

a critical role in the innate and adaptive immune defense against infections [3].<br />

Viruses (or unmethylated CpG DNA) engage Toll-like-receptors (TLR) 7 and TLR 9<br />

on IPC [25] and trigger secretion of m<strong>as</strong>sive amounts of type I interferons (IFN-α, -β,<br />

-ω and -λ) [137]. This specialization of IPCs in cytokine production is reflected in<br />

their distinct "pl<strong>as</strong>macytoid" morphology resembling Ig-secreting pl<strong>as</strong>ma <strong>cells</strong> [138].<br />

IPC-derived cytokines are critical <strong>for</strong> the initiation of early anti-viral NK cell<br />

responses [139,140]. In addition, IPC-derived type I IFNs boost the ability of cDCs to<br />

mature and stimulate T <strong>cells</strong>, thus promoting antiviral CTL responses [141,142].<br />

Furthermore, IPCs provide a critical costimulatory signal <strong>for</strong> the CpG-induced<br />

activation of cDCs [143,144]. Independent of their cytokine- and CD40L-mediated<br />

accessory function to control NK and cDC activities, IPCs have also been proposed to<br />

play a direct role <strong>as</strong> APC in the onset of T cell stimulation. Constitutive presence of<br />

IPCs in lymphoid organs, their expression of MHC molecules and acquisition of DC<br />

morphology upon culture h<strong>as</strong> led to the concept that IPCs represent a distinct DC<br />

subset, <strong>as</strong> so-called pl<strong>as</strong>macytoid DCs (PDCs). However, the original discovery of<br />

DCs defined them by the capacity to prime naïve T <strong>cells</strong> [80] and this activity h<strong>as</strong><br />

remained their hallmark since. Moreover, recent in vivo DC depletion experiments<br />

indicate that CD11c high cDCs are required to initiate cytotoxic CD8 + T cell responses<br />

against intracellular pathogens [42,145]. The potential of IPC/PDCs to prime naïve T<br />

<strong>cells</strong> remains however controversial. Freshly isolated PDCs are generally poor T cell<br />

stimulators. Thus, human blood-derived PDCs do not stimulate naïve CD4 + T <strong>cells</strong> in<br />

59


a MLR, unless cultured in the presence of IL-3 or virus (HSV) [146]. Moreover,<br />

murine splenic PDCs fail to induce naïve T cell proliferation to endogenous antigens<br />

even after virus exposure [147]. In contr<strong>as</strong>t, murine peptide-pulsed PDCs derived<br />

from Flt-3-driven BM cultures or spleens can promote the in vitro expansion of CD4 +<br />

T <strong>cells</strong> and T H polarization [148]. Adoptive transfer experiments with splenic and BM<br />

culture-derived, CpG-matured PDCs showed that PDCs can elicit responses of naïve<br />

CD8 + T <strong>cells</strong> to endogenous, but not exogenous antigens [149]. These conflicting<br />

results are likely due to the different source of the PDCs used in the reports.<br />

Moreover, none of the studies addressed the in vivo potential of un-manipulated PDCs<br />

to prime naïve T <strong>cells</strong>.<br />

Recently, IPC/PDCs have been shown to play a critical role in the control of airway<br />

inflammation [36,150] and allo-transplant rejection 151,152]. Although these findings<br />

mark PDCs <strong>as</strong> potential candidates <strong>for</strong> future adoptive cell therapies, such approaches<br />

will require better understanding of the in vivo characteristics of this unique cell type.<br />

Here we analyze CD4 + and CD8 + T cell responses to antigen challenge in mice that<br />

were conditionally depleted of CD11c high cDCs, but retain IPC/PDCs. We show that<br />

depletion of cDCs impairs splenic T cell responses, indicating that PDCs are<br />

incapable of T cell <strong>priming</strong> in this organ. In contr<strong>as</strong>t, the combined use of cDC<br />

ablation and a novel PDC-specific antigen targeting strategy revealed that in LNs,<br />

PDCs can efficiently trigger productive naïve CD4 + T cell responses. Interestingly,<br />

and in contr<strong>as</strong>t cDC-driven responses, the PDC-triggered CD4 + T cell stimulations<br />

lack a concomitant CD8 + T cell expansion. Our in vivo results, thus, characterize<br />

PDCs <strong>as</strong> bona fide DCs that can initiate unique CD4 + Th cell-dominated primary<br />

immune responses.<br />

4.2 Results<br />

4.2.1 PDCs are spared from ablation in DTx-treated CD11c-DTR transgenic<br />

mice<br />

To investigate differential in vivo functions of APC in lymphoid organs we recently<br />

developed a DTR-b<strong>as</strong>ed system that allows the conditional ablation of CD11c high<br />

cDCs (12). DTR expression in CD11c-DTR transgenic mice is driven by a DNA<br />

fragment flanking the Itgax gene [153], which encodes the α X subunit of the CD11c<br />

integrin. Murine CD11c expression is found in all cDCs, but h<strong>as</strong> also been reported<br />

60


<strong>for</strong> certain macrophages [154,155,156], activated T <strong>cells</strong> [157], NK <strong>cells</strong> [158] and<br />

pl<strong>as</strong>mabl<strong>as</strong>ts [87].<br />

Murine PDCs are defined <strong>as</strong> being CD11c low B220 + Ly6C + and can furthermore be<br />

identified by the PDC-specific antibodies 120G8 [12], 440c [159] and mPDCA-1<br />

[160]. To test the DTx-sensitivity of CD11c-DTRtg CD11c low PDCs, we injected<br />

CD11c-DTRtg C57BL/6 mice with DTx. Flow cytometry of the spleens one day after<br />

DTx treatment revealed that PDCs were spared from DTx-induced ablation, where<strong>as</strong><br />

cDCs were depleted (Fig. 17A). The DTx resistance of PDCs is explained by fact that<br />

the Itgax promoter fragment does not carry all transcription control elements required<br />

<strong>for</strong> CD11c expression in all the <strong>cells</strong> where it is active. Thus, this DNA fragment is<br />

inactive in PDCs that transcribe their endogenous Itgax allele. DTx resistance w<strong>as</strong><br />

also confirmed <strong>for</strong> LN PDCs (Fig. 17C,D; Fig. 19A).<br />

To investigate whether the toxin treatment results in a functional impairment of PDCs<br />

we tested type I interferon responses. As seen in Fig. 17E, DTx treatment neither<br />

affected the IFN-α response to in vivo CpG treatment nor compromised the ability of<br />

PDCs to produce IFN-α in response to an in vitro influenza virus challenge. Of note,<br />

we consistently observed in spleens, but not LNs of CD11c-DTRtg C57BL/6 mice a<br />

small fraction of mPDCA-1 + CD11c low <strong>cells</strong> that were DTR-GFP + and DTx<br />

sensitive(Fig. 17B,D) Collectivelly, these results indicate that PDCs of CD11c-DTR<br />

transgenic mice are resistant to DTx treatment. CD11c-DTRtg mice, thus, provide a<br />

unique model to study the role of PDCs in the <strong>priming</strong> of immune responses in the<br />

absence of conventional CD11c high DCs.<br />

61


Figure 17. PDCs are spared from ablation in DTx-treated CD11c-DTRtg mice<br />

(A) Flow cytometry of the spleens of untreated controls and CD11c-DTRtg mice 1 d after i.v. injection<br />

of DTx (4 ng/body weight). cDCs are gated <strong>as</strong> CD11c high mPDCA-1 neg <strong>cells</strong>; PDCs are defined <strong>as</strong> being<br />

CD11c int mPDCA-1 + . Bar diagram represents analysis of splenic cDCs and PDCs of untreated mice<br />

and mice 24 h after treatment (i.p. DTx ; 10 μg OVA i.v.). (each group n=5). Dot blots show <strong>cells</strong><br />

gated according to scatter.<br />

(B) DTR-GFP expression profiles of splenic cDCs and PDCs isolated from CD11c-DTRtg mice<br />

untreated or treated with DTx. Cells are defined and gated <strong>as</strong> in A. Graphs are representative of five<br />

repeats. Percentage refers to DTx-sensitive GFP + mPDCA-1 + CD11c low <strong>cells</strong> (filled area).<br />

(C) Flow cytometry of popliteal LNs of control mice CD11c-DTRtg mice 1 d after s.c. injection of 20<br />

ng DTx into the hind footpads. Bar diagram represents analysis of splenic cDCs and PDCs of untreated<br />

mice and mice 24 h upon indicated treatment (i.p. DTx ; 10 μg OVA i.v.). (each group n=5). Dot blots<br />

show <strong>cells</strong> gated according to scatter.<br />

(D) DTR-GFP expression profiles of LN PDCs isolated from untreated or treated CD11c-DTRtg mice.<br />

Note absence of GFP + mPDCA-1 + CD11c low subpopulation observed in spleen (B).<br />

(E) IFN-α production by splenic PDC and non-PDC cell fractions isolated from noninjected or DTxinjected<br />

CD11c-DTRtg mice and incubated in vitro <strong>for</strong> 20 h in the presence or absence of 400 HAU/<br />

ml influenza virus and blood sera analysis of DTx-treated and untreated CD11c-DTRtg mice, 6 h after<br />

PBS or 100 μg CpG i.v. injection. Error bars depicts the s.d.<br />

(F) Immunohistochemistry of popliteal LN from wt mouse, untreated CD11c-DTRtg mouse and<br />

CD11c-DTRtg mouse, 1 d after footpad injection of 20 ng DTx. Sections are stained with anti-GFP<br />

antibody (brown). Scale bar refers to 100 μm.<br />

(G) Flow cytometry of CD8 - and CD8 + cDC subsets in popliteal LNs, 1 d after footpad injection of 20<br />

ng DTx. Dot plots were gated according to the scatter and exclude B220 + <strong>cells</strong>.<br />

4.2.2 PDCs fail to prime naïve T <strong>cells</strong> in the spleen<br />

We first investigated the potential of splenic PDCs to stimulate naïve T <strong>cells</strong> in the<br />

absence of CD11c high cDCs. We isolated TCR transgenic Ovalbumin (OVA)-specific<br />

CD8 + and CD4 + T <strong>cells</strong> from OT-I and OT-II mice, respectively [161,162]. Donor<br />

mice carried an allotypic marker (CD45.1) that allowed <strong>for</strong> detection of the<br />

transferred <strong>cells</strong> in the recipient mice. Prior to transfer, the T <strong>cells</strong> were labeled with<br />

63


the intracellular dye CFSE, which allows the monitoring of in vivo proliferation [163].<br />

T <strong>cells</strong> were cotransferred into wt and CD11c-DTRtg C57BL/6 mice. The grafted<br />

mice were treated with DTx and eight hours later immunized by intravenous (i.v)<br />

injection of soluble OVA (10μg) or OVA-loaded splenocytes [8]. Four days after<br />

immunization, the mice were sacrificed, spleens were isolated and analyzed <strong>for</strong> the<br />

proliferation status of the T cell grafts (Fig. 18). All challenges resulted in vigorous<br />

expansion of both CD4 + and CD8 + T cell grafts in the wt recipient mice. As<br />

previously reported [42], depletion of CD11c high cDCs impaired the response of CD8 +<br />

T <strong>cells</strong> to cell-<strong>as</strong>sociated antigen. Presentation of antigen in the context of MHC cl<strong>as</strong>s<br />

I to CD8 + T <strong>cells</strong> requires a unique phagosome-to-cytosol pathway resulting in crosspresentation,<br />

which is an activity proposed to be uniquely <strong>as</strong>sociated with cDCs or<br />

cDC subsets [8,42,127]. In support of this notion, we found that cDCs were also<br />

needed <strong>for</strong> CD8 + T cell <strong>priming</strong> after challenge with exogenous soluble antigen (Fig.<br />

18). Moreover, depletion of splenic cDCs also abrogated OVA-specific CD4 + T cell<br />

responses to cell-<strong>as</strong>sociated and soluble antigen, thus extending the study by Tian et<br />

al. [133]. Importantly, <strong>as</strong> opposed to a recent report [164], we evaluated the T cell<br />

<strong>priming</strong> potential of PDCs in the absence of cDCs. Taken together, splenic PDCs are<br />

unable to prime naïve CD4 + and CD8 + T <strong>cells</strong> in absence of CD11c high cDCs.<br />

64


Figure 18. cDC ablation impairs splenic CD4 + and CD8 + T cell <strong>priming</strong><br />

Flow cytometry of cotransferred CFSE-labeled CD45.1 + OT-I and OT-II T cell grafts (10 6 <strong>cells</strong> each)<br />

in DTx-treated nontransgenic and CD11c-DTRtg mice (CD45.2), 4 d after i.v. immunization with<br />

OVA-loaded splenocytes or soluble OVA. Histograms represent <strong>cells</strong> gated according to scatter,<br />

CD45.1 and CD8 or CD4 surface expression, <strong>as</strong> indicated in the dot plots. Data show results of one<br />

representative experiment out of three.<br />

4.2.3 PDCs can prime naïve CD4 + T <strong>cells</strong> in LNs, but fail to support CD8 + T cell<br />

<strong>priming</strong><br />

T cell responses are initiated in specialized secondary lymphoid organs, such <strong>as</strong> the<br />

spleen and LNs, which differ considerably in their micro-architecture reflecting their<br />

specialization <strong>for</strong> the handling of blood- and tissue-borne antigens, respectively. We<br />

next investigated whether CD11c high DCs would also be required in the LNs. Local,<br />

subcutaneous (s.c.) DTx injection results in the rapid depletion of LN-resident <strong>cells</strong><br />

expressing the DTR-GFP fusion protein, including both CD8α + and CD8α - CD11c high<br />

cDCs and sinusoidal macrophages [154], but not PDCs (Fig. 17C,F,G; Fig 19A).<br />

Repetitive s.c. DTx injection results in persistent cDC ablation (Fig. 19A; Fig. 20A).<br />

To investigate the impact of the LN cDC depletion on T cell responses in this organ,<br />

we engrafted wt and CD11c-DTRtg C57BL/6 recipient mice with a mixture of OVAspecific<br />

CD4 + (OT-II) and CD8 + T <strong>cells</strong> (OT-I). The mice were subjected to the DTx<br />

65


treatment (which w<strong>as</strong> continued throughout the experiment to maintain cDC depletion<br />

(Fig. 19A)) and eight hours later immunized by s.c. injection of 10μg OVA. Three<br />

days after immunization, we isolated the antigen-draining popliteal LNs and analyzed<br />

them <strong>for</strong> the proliferation status of grafted T <strong>cells</strong>. The adoptively cotransferred<br />

CD45.1 + CD4 + and CD8 + T <strong>cells</strong> readily proliferated in the LNs of DTx-treated wt<br />

mice in response to immunization with soluble OVA (Fig. 19B). Importantly, the<br />

DTx-induced cDC depletion abrogated the expansion of CD8 + T <strong>cells</strong> indicating that,<br />

<strong>as</strong> in the spleen, <strong>priming</strong> of naïve CD8 + T <strong>cells</strong> required the presence of cDCs.<br />

Surprisingly, however, we observed a proliferative response of the grafted CD4 + T<br />

<strong>cells</strong> in the cDC-depleted LNs (Fig. 19B). Importantly, this w<strong>as</strong> unlikely to be caused<br />

due to residual cDCs, since the DTx treatment impaired in the same LNs the CD8 + T<br />

cell response, which is generally more sensitive to antigen challenge [165]. The<br />

persistence of the CD4 + T cell <strong>priming</strong> in absence of cDCs w<strong>as</strong> also observed in<br />

CD11c-DTRtg BALB/c mice, in combination with an OVA-specific TCR transgenic<br />

naïve CD4 + T cell graft isolated from DO11.10 mice [166] (Fig. 21). Moreover, to<br />

confirm our observation in a non-TCR transgenic system and with an antigen other<br />

than OVA, we challenged untreated and cDC-depleted DTRtg mice with KLH/ CpG<br />

and analyzed the resulting response of endogenous CD4 + T <strong>cells</strong>. On day 7 after s.c.<br />

antigen challenge, we purified CD4 + T <strong>cells</strong> from popliteal LNs and subjected them to<br />

an in vitro restimulation protocol with KLH-pulsed APC. Again, CD4 + T <strong>cells</strong> were<br />

efficiently primed irrespective of the presence of cDCs (Fig. 20).<br />

66


Figure 19. Ablation of cDCs impairs CD8 + T cell <strong>priming</strong>, but not CD4 + T cell <strong>priming</strong> in the LNs<br />

(A) Flow cytometry of the popliteal LNs of CD11c-DTRtg mice during the course of <strong>priming</strong><br />

experiment. Mice were injected twice with DTx into the hind footpads, on day 1 and 3. The line graph<br />

represents the analysis of LN cDCs and PDCs <strong>as</strong> indicated in the FACS blots (each group n=3). The<br />

bottom schedule refers to T cell <strong>priming</strong> experiments per<strong>for</strong>med in B.<br />

(B) Flow cytometry of cotransferred CFSE-labeled CD45.1 + OT-I and OT-II <strong>cells</strong> (bottom row: OT-II<br />

transfer only) into DTx-treated nontransgenic and CD11c-DTRtg mice (CD45.2 + ), 4 d after s.c.<br />

immunization with soluble OVA. Histograms represent <strong>cells</strong> gated according to scatter, CD45.1 and<br />

67


CD8 or CD4 expression, <strong>as</strong> indicated in the dot plots. Data show results of two representative<br />

experiments out of ten repeats. Graph summarizes percentages of dividing CFSE-labeled CD45.1 + OT-<br />

II <strong>cells</strong> in mice (n=14) that were treated with DTx and immunized, compared with proliferation in<br />

immunized wt control mice (n=8) (set <strong>as</strong> 100%). Each dot represents an independent mouse.<br />

Figure 20. Priming of endogenous, nontransgenic CD4 + T <strong>cells</strong> in the LNs of cDC-depleted mice<br />

(FVB/N background)<br />

(A) Dynamics of CD11c high cDC and CD4 + T cell (B) numbers throughout course of experiment<br />

involving KLH/CpG immunization (d1) and daily s.c. injections of DTx (10ng/ footpad). Both FVB/N<br />

and CD11c-DTRtg FVB/N mice were treated with DTx.<br />

(C) In vitro restimulation <strong>as</strong>say of MACS-enriched CD4 + T <strong>cells</strong> with KLH-pulsed, irradiated FVB/N<br />

splenocytes. T cell proliferation w<strong>as</strong> <strong>as</strong>sessed by addition of 1 μCi of [H 3 ] thymidine, 72 h after coculture<br />

of T <strong>cells</strong> and splenocytes and me<strong>as</strong>ured 8 h later. Note that incre<strong>as</strong>e in proliferation observed<br />

in absence of cDCs is likely due to absence of recruitment of bystander T <strong>cells</strong> to the LNs [189].<br />

68


Figure 21. CD4 + T lymphocyte <strong>priming</strong> in the LNs of cDC-depleted BALB/c mice<br />

(A) Flow cytometry of adoptively transferred CFSE-labeled OVA specific CD4 + T <strong>cells</strong> (DO11.10).<br />

Cells were transferred into nontransgenic and CD11c-DTRtg BALB/c mice, that were subsequently<br />

treated with DTx and immunized by s.c. injection with soluble OVA (10mg/ footpad). Histograms<br />

represent CFSE profiles of <strong>cells</strong> gated according to expression of the TCR clonotype (KJ126) and CD4.<br />

Data show one representative experiment out of three.<br />

(B) Priming of CD4 + T <strong>cells</strong> (DO11.10) by OVA-pulsed PDC in vitro. T <strong>cells</strong> (1 x 10 5 ) were cocultured<br />

with OVA-pulsed cDCs or PDCs (2 x 10 4 ) isolated from LNs of naïve BALB/c mice. The<br />

positive PDC fraction, which w<strong>as</strong> obtained by high-speed sorting according to CD11c and mPDCA-1<br />

expression contained no contaminating CD11c high cDCs. Proliferation w<strong>as</strong> <strong>as</strong>sessed by addition of 1<br />

μCi of [H 3 ] thymidine, after 72 h and me<strong>as</strong>ured 8 h later.<br />

Our data show that LNs contain APC other than CD11c high DCs or macrophages. that<br />

are capable of stimulating naïve CD4 + T <strong>cells</strong>. Although PDCs are potential<br />

candidates, other MHC cl<strong>as</strong>s II + <strong>cells</strong> could be responsible <strong>for</strong> the CD4 + T cell<br />

<strong>priming</strong>. By far, the largest MHC cl<strong>as</strong>s II + population in the LNs are B lymphocytes,<br />

which, even when naïve, have been shown to efficiently ingest and process<br />

noncognate protein antigens [167]. However, the ability of B <strong>cells</strong> to activate naïve<br />

CD4 + T <strong>cells</strong> remains under debate [168,169,170]. Moreover, pMHC encounter on B<br />

<strong>cells</strong> is believed to result in non-productive T cell <strong>priming</strong> or tolerization [171]. To<br />

investigate the role of B <strong>cells</strong> in our system, we crossed the CD11c-DTR transgene<br />

onto a B cell-deficient genetic background (C57BL/10-Igh-6tm1Cgn (μMT) [172].<br />

CD11c-DTRtg μMT and μMT C57BL/6 mice were engrafted with OVA-specific<br />

CD4 + T <strong>cells</strong> (OT-II) and subjected to the DTx treatment and OVA immunization. B<br />

cell deficiency resulted in a reduced recovery of grafted CD4 + T <strong>cells</strong> in DTx-treated<br />

69


CD11c-DTRtg μMT and μMT C57BL/6 mice. However, the frequency of OT-II <strong>cells</strong><br />

that lost the CFSE-label and thus proliferated in response to the OVA challenge w<strong>as</strong><br />

unaffected by the absence of B <strong>cells</strong> (Fig. 22A,B). CD4 + T cell <strong>priming</strong> in the LNs,<br />

thus, persisted in the combined in absence of B <strong>cells</strong> and CD11c high cDCs. Another<br />

potential APC candidate are epidermal Langerhans <strong>cells</strong> (LCs), which are CD11c neg ,<br />

but up-regulate CD11c expression upon migration to the skin draining LNs [4]. LCs<br />

would, hence, be expected to become DTx-sensitive by the time they reach the LNs.<br />

To nevertheless exclude the possibility that the CD4 + T cell response we observed<br />

w<strong>as</strong> due to residual LCs, we crossed the CD11c-DTRtg mice to mice that harbor the<br />

DTR gene under the LC-specific Langerin promoter [173]. DTx-treatment of CD11c-<br />

DTRtg LanDTRtg mice results in ablation of cDCs and LCs (data not shown).<br />

However, the response of the adoptively transferred OT-II CD4 + T <strong>cells</strong> to the OVA<br />

challenge persisted (Fig. 22C). Collectively, these data suggest that <strong>cells</strong> other than<br />

cDCs, macrophages, B <strong>cells</strong> or LCs are responsible <strong>for</strong> the <strong>priming</strong> of CD4 + T <strong>cells</strong><br />

and point at a role of PDCs in antigen presentation.<br />

70


Figure 22. LN CD4 + T cell <strong>priming</strong> in combined absence of cDCs and B <strong>cells</strong>, <strong>as</strong> well <strong>as</strong> cDCs and<br />

Langerhans <strong>cells</strong><br />

(A) Flow cytometry of cDC depletion in popliteal LNs of CD11c-DTRtg μMT mice be<strong>for</strong>e and 1 d<br />

after footpad injection of 20 ng DTx.<br />

(B) CD4 + T cell <strong>priming</strong> in popliteal LNs persists in combined absence of B <strong>cells</strong> and CD11c high cDCs.<br />

Flow cytometry of transferred CFSE-labeled CD45.1 OT-II <strong>cells</strong> in DTx treated wt, μMT, CD11c-<br />

DTRtg and CD11c-DTRtg μMT mice (CD45.2), 4 d after s.c. footpad immunization with 10 μg soluble<br />

OVA. Histograms represent CFSE profiles of <strong>cells</strong> gated according to CD45.1 and CD4 expression.<br />

Data show one representative experiment out of three.<br />

(C) CD4 + T cell <strong>priming</strong> persists in combined absence of Langerhans <strong>cells</strong> and CD11c high cDCs. Flow<br />

cytometry of transferred CFSE-labeled CD45.1 OT-II <strong>cells</strong> after transfer in DTx treated CD11c-DTRtg<br />

and CD11c-DTRtg Lan-DTRtg mice (CD45.2), 4 d after s.c. footpad immunization with 10 μg soluble<br />

OVA. Histograms represent CFSE profiles of <strong>cells</strong> gated according to CD45.1 and CD4 expression.<br />

71


4.2.4 LN PDCs efficiently acquire and process soluble antigen and prime<br />

productive T cell responses<br />

Antigen presentation and CD4 + T cell <strong>priming</strong> by LN-resident PDCs would require<br />

their antigen uptake. Importantly, soluble antigens of low molecular weight (


Proliferative T cell expansion can be <strong>as</strong>sociated with the generation of cytokineproducing<br />

effector T <strong>cells</strong>, but also abortive T cell responses [176,177,178]. We<br />

there<strong>for</strong>e investigated the quality of the primed LN CD4 + T <strong>cells</strong> by studying (Th) cell<br />

polarization and long-term T cell survival. To analyze Th polarization of the grafted<br />

OVA-specific CD4 + T <strong>cells</strong>, we per<strong>for</strong>med an in vivo restimulation <strong>as</strong>say by<br />

rechallenging the OVA-immunized mice with an i.v. injection of OVA peptide [126].<br />

Both in presence and absence of CD11c high cDCs, CD4 + T <strong>cells</strong> differentiated<br />

efficiently into IFN-γ-producing effector T <strong>cells</strong> (Fig. 24A). Abortive T cell<br />

stimulation is characterized by a collapse of the antigen-specific T cell population<br />

after its initial expansion [176,177]. To test the persistence of CD4 + T <strong>cells</strong> that were<br />

primed in absence of cDCs we used mixed CD11c-DTRtg BM chimer<strong>as</strong>, which allow<br />

<strong>for</strong> repetitive DTx treatment and hence extended cDC depletion [85]. In both<br />

untreated and cDC-depleted mice, the OT-II CD4 + T cell graft clonally expanded and<br />

persisted well beyond the initial antigen challenge (Fig. 24B). Furthermore, in<br />

presence and absence of cDCs the grafted naïve CD4 + T <strong>cells</strong> differentiated into T<br />

<strong>cells</strong> of CD45RB low memory phenotype [179].Taken together, PDCs seem capable of<br />

stimulating naïve CD4 + T <strong>cells</strong>, resulting in a persistent response including IFN-γproducing<br />

effector T <strong>cells</strong> of memory phenotype. Importantly, and in contr<strong>as</strong>t to the<br />

immune reaction triggered by cDCs, this response is not accompanied by a<br />

concomitant CD8 + T cell response.<br />

73


Figure 24. CD4 + T <strong>cells</strong> that are primed in absence of cDCs differentiate into T effector <strong>cells</strong> with<br />

memory phenotype and persist <strong>for</strong> extended periods of time<br />

(A) CFSE-labeled CD45.1 + OT-II <strong>cells</strong> were transferred into nontransgenic and CD11c-DTRtg mice<br />

(CD45.2 + ). 1 d later, mice were treated s.c. with DTx and 8 h later, they were immunized in hind<br />

footpads with OVA and CpG. 3 d after immunization, INF-γ production in the popliteal LNs w<strong>as</strong><br />

<strong>as</strong>sessed after in vivo restimulation with OVA peptide, 2 h be<strong>for</strong>e removal of popliteal LNs. Data<br />

represent INF-γ profiles after gating on CD4 + CD45.1 + lymphocytes. Results are representative of two<br />

independent experiments.<br />

(B) CFSE-labeled CD45.1 + OT-II <strong>cells</strong> were transferred into mixed [wt>wt] or [CD11c-DTRtg>wt]<br />

BM chimer<strong>as</strong> (CD45.2 + ). The mice were injected s.c. with DTx 1 d after the transfer and were<br />

immunized 8 h later with OVA into hind footpads. Sequential s.c. DTx injections were per<strong>for</strong>med<br />

every second day until day 21. Blood w<strong>as</strong> collected on days 4, 11 and 21 and evaluated <strong>for</strong> presence of<br />

OT-II <strong>cells</strong> (CD4 + CD45.1 + ), and expression of the memory T cell marker CD45RB. Bar graph depicts<br />

mean fluorescence intensity (MFI) values of blood staining <strong>for</strong> CD45RB on day 4, 11 and 21. Results<br />

are representative of two independent experiments.<br />

4.2.5 PDC-specific antigen targeting and PDC-mediated in vivo and in vitro CD4 +<br />

T cell <strong>priming</strong><br />

The above results suggest that LN PDCs efficiently ingest exogenous antigen and are<br />

able to initiate productive CD4 + T cell responses. To obtain independent evidence that<br />

PDCs can act <strong>as</strong> APC, we decided to adopt a strategy involving specific, antibodymediated<br />

antigen targeting to APC [176] and conjugate the PDC-specific mPDCA-1<br />

antibody to OVA. To avoid complement-dependent lysis [160], we used a nondepleting<br />

F(ab') 2 fragment. Functional integrity of the construct w<strong>as</strong> tested by<br />

Western blotting. As depicted in Fig. 25A, the conjugates were readily detected both<br />

74


via the kappa light chain and the OVA-fraction, where<strong>as</strong> free OVA or unconjugated<br />

mPDCA-1-F(ab’) 2 fragments were only detected with the anti-rat IgG or the anti-<br />

OVA antibody, respectively. The OVA-conjugated antibody fragment specifically<br />

targeted PDCs in splenic or LN single cell suspensions in vitro (data not shown). To<br />

demonstrate in vivo specificity, we took advantage of a FITC-conjugate of the<br />

mPDCA-1-F(ab’) 2 -OVA construct. Intravenous or s.c. injection of FITC-mPDCA-1-<br />

F(ab’) 2 -OVA resulted in specific labeling of CD11c low PDCs in spleens and LNs and<br />

spared CD11c high cDCs (Fig. 25B).<br />

Figure 25. Characterization of selective APC targeting via the anti-mPDCA-1-F(ab’) 2 -OVA construct<br />

(A) Western blot analysis of the OVA-conjugated antibody construct. Free OVA, unconjugated or OVAconjugated<br />

anti-mPDCA-1 antibody constructs were resolved by SDS-PAGE (4-12% gradient Tris-glycine<br />

gel) and after immunoblotting detected with anti-rat and anti-OVA antibody, respectively. Lanes 1 and 5<br />

contain free OVA, lane 2 and 6 contain the unconjugated anti-mPDCA-1 F(ab’) 2 antibody fragment, and<br />

lanes 3, 4 and 7, 8 contain two fractions of the anti-mPDCA-1-F(ab’) 2 -OVA conjugate.<br />

(B) Specific in vivo targeting of PDCs with FITC-labeled anti-mPDCA-1-F(ab’) 2 -OVA. Conjugates<br />

were injected i.v. or s.c. and after 3 h spleens and popliteal and inguinal LNs were isolated.<br />

Counterstaining with CD11c w<strong>as</strong> per<strong>for</strong>med on single cell preparations from untreated (left dot plots)<br />

or in vivo targeted <strong>cells</strong> (middle and right dot plots). Note that the staining in the FL-1 channel is b<strong>as</strong>ed<br />

on the in vivo injected FITC-coupled mPDCA-1-F(ab’) 2 -OVA construct.<br />

To direct the OVA antigen to both PDCs and cDCs we used the mPDCA-1-F(ab’) 2 -<br />

OVA conjugate (mPDCA-1-OVA) and an OVA conjugate that targets a subset of<br />

CD11c high cDCs via the CD205 receptor (DEC205-OVA) [176,180]. We first<br />

investigated the response of OVA-specific CD8 + T <strong>cells</strong> (OT-I) to the conjugate<br />

challenge. As seen in Fig. 26A, s.c. immunization of untreated CD11c-DTRtg mice<br />

with the mPDCA-1-OVA and DEC205-OVA conjugates resulted in comparable<br />

responses of the naïve CD8 + T <strong>cells</strong> in the draining popliteal LNs. Importantly,<br />

depletion of cDCs abrogated both the DEC205-OVA and the mPDCA-1-OVA-<br />

75


induced CD8 + T cell expansion. Thus, The cDC-driven CD8 + T cell response to the<br />

mPDCA-1-OVA conjugate w<strong>as</strong> likely due to antibody-independent antigen uptake by<br />

cDCs or secondary cross-presentation of cellular PDC debris by cDCs. Conventional<br />

DC ablation thus revealed that, in contr<strong>as</strong>t to a recent study [164], even when directly<br />

targeted with exogenous antigen, PDCs seem unable to trigger CD8 + T cell responses.<br />

Instead, our results support the notion of the general inability of PDCs to channel<br />

exogenous antigen into the MHC cl<strong>as</strong>s I presentation pathway and cross-present<br />

[149].<br />

Next we analyzed the response of OVA-specific LN CD4 + T <strong>cells</strong> (OT-II) to<br />

conjugate challenge. Like the immunization with OVA, challenge of untreated<br />

CD11c-DTRtg mice with the antibody-conjugates resulted in vigorous expansion of<br />

the CD4 + T cell grafts. In contr<strong>as</strong>t to the CD8 + T cell responses and confirming our<br />

previous results (Fig. 19), the CD4 + T cell responses to the OVA and mPDCA-1-<br />

F(ab’) 2 -OVA challenge resisted the DTx treatment in CD11c-DTRtg mice (Fig. 26B).<br />

Importantly, however, the CD4 + T cell response to the DEC205-OVA challenge w<strong>as</strong><br />

impaired by depletion of CD11c high cDCs. These results provide further evidence that<br />

the CD4 + T cell response we observe in the absence of CD11c high cDCs is driven by<br />

PDCs. Notably, the antibodies to DEC205 and mPDCA-1 target distinct surface<br />

receptors [14,181], which likely differ in their potential to promote endocytosis. This<br />

precludes any quantitative conclusion on the <strong>priming</strong> potential of cDCs vs. PDCs.<br />

76


Figure 26. In vivo antigen targeting of PDCs leads to <strong>priming</strong> of LN CD4 + T <strong>cells</strong>, but not CD8 + T<br />

<strong>cells</strong> in absence of cDCs<br />

(A) Flow cytometry of transferred CFSE-labeled CD45.1 + OT-I <strong>cells</strong> into s.c. DTx treated CD11c-<br />

DTRtg mice (CD45.2), 4 d after footpad immunization with 8 μg mPDCA-1-OVA (OVA content ~<br />

7.5%) or 7 ng DEC205-OVA (OVA content ~10%). Histograms represent CFSE profiles of <strong>cells</strong> gated<br />

according to CD45.1 and CD8 expression, <strong>as</strong> indicated in the dot plot. Data show a representative<br />

result out of three experiments.<br />

(B) Flow cytometry of transferred CFSE-labeled CD45.1 + OT-II <strong>cells</strong> into DTx treated CD11c-DTRtg,<br />

4 d after footpad immunization with F(ab) 2 , OVA, control F(ab) 2 -OVA, mPDCA-1- F(ab) 2 -OVA or<br />

DEC20-OVA. Histograms represent CFSE profiles of <strong>cells</strong> gated according to CD45.1 and CD4<br />

expression, <strong>as</strong> indicated in the dot plot. Data show one representative experiment out of three.<br />

To obtain final proof <strong>for</strong> the CD4 + T cell <strong>priming</strong> potential of PDCs we decided to<br />

employ an antibody-mediated depletion protocol to test T cell responses in the<br />

combined absence of both cDCs and PDCs. However, in our hands, even repetitive<br />

injection of mPDCA-1 and anti-Gr1 antibodies [36,160] resulted in incomplete LN<br />

PDC depletion (data not shown). There<strong>for</strong>e, we resorted to an in vivo loading / in vitro<br />

read-out system. Wt BALB/c mice were immunized with OVA or mPDCA-1-OVA.<br />

One day later, PDCs, cDCs and B <strong>cells</strong> were isolated from LNs and spleens, sorted to<br />

purity and tested in vitro <strong>for</strong> their ability to prime naïve OVA-specific CD4 + T <strong>cells</strong><br />

(DO11.10). As seen in Fig. 27, cDCs isolated from LNs or spleens of OVAimmunized<br />

mice readily stimulated T cell proliferation, where<strong>as</strong> cDCs and B <strong>cells</strong><br />

isolated from mPDCA-1-OVA immunized mice failed to do so. This confirms the<br />

specificity of the in vivo antigen targeting strategy (Fig. 25B). Importantly, PDCs<br />

77


isolated from LNs of mice immunized with both OVA and mPDCA-1-OVA were able<br />

to stimulate the naïve CD4 + T <strong>cells</strong>. Substantiating our in vivo data on the inability of<br />

splenic PDCs to prime CD4 + T <strong>cells</strong>, also in the in vitro <strong>as</strong>say PDCs isolated from<br />

spleens of immunized mice failed to stimulate the T <strong>cells</strong>. Collectively, our results<br />

show that LN PDCs can initiate unique CD4 + T helper cell-dominated primary<br />

immune responses.<br />

Figure 27. In vivo antigen targeting of PDCs leads to LN PDC-mediated in vitro CD4 + T cell<br />

proliferation<br />

CD11c high cDCs, PDCs and B <strong>cells</strong> were sorted from spleens and LNs of BALB/c mice that were i.v. or<br />

s.c. immunized with OVA or mPDCA-1-F(ab’) 2 -OVA, and cultured with OVA-specific CD4 + T <strong>cells</strong><br />

(DO11.10) <strong>for</strong> 72 h, after which thymidine incorporation w<strong>as</strong> me<strong>as</strong>ured. Results are plotted <strong>as</strong> the<br />

mean of cpm from triplicate cultures with <strong>cells</strong> isolated from immunized mice minus the mean cpm of<br />

cultures of the respective cell populations isolated from unimmunized mice. The positive fraction of<br />

PDCs contained less than 0.01% contaminating CD11c high cDCs. Note CD4 + T cell <strong>priming</strong> by LN<br />

PDCs isolated from OVA and PDCA-OVA immunized mice. (Each group n=4). Error bars represent<br />

s.d. Controls include responder T <strong>cells</strong> only and DO11.10 T <strong>cells</strong> with B <strong>cells</strong> isolated from PDCA-<br />

OVA immunized mice.<br />

78


4.3 Discussion<br />

Here we analyzed the responses of naïve CD4 + and CD8 + T <strong>cells</strong> to antigen challenge<br />

in mice that were depleted of conventional CD11c high DCs, but retain PDCs. In<br />

support of previous reports [42,133], we found that the capacity to prime naïve CD4 +<br />

and CD8 + T <strong>cells</strong> in the spleen is uniquely restricted to cDCs. Ablation of cDCs,<br />

however, revealed the surprising potential of PDCs to support CD4 + T cell responses<br />

in the LNs. Importantly, PDCs differed from cDCs in that they could not induce CD8 +<br />

T cell <strong>priming</strong>.<br />

Naïve T <strong>cells</strong> are stimulated in lymphoid organs by specialized, professional APC that<br />

induce clonal T cell expansion and differentiation. Efficient eradication of invading<br />

microbes requires distinct immune responses, involving both cellular and humoral<br />

defense mechanisms. Pathogen identification is believed to result from engagement of<br />

pathogen sensors (such <strong>as</strong> TLRs), which are differentially expressed on APC and may<br />

allow distinct APC subsets to trigger specific responses. Division of labor between<br />

APC remains however poorly understood. This holds in particular <strong>for</strong> IPC/ PDCs,<br />

which are critical <strong>for</strong> NK <strong>cells</strong> and cDC function, but whose in vivo capacity <strong>for</strong><br />

antigen presentation and T cell <strong>priming</strong> remains unclear. We re<strong>as</strong>oned that any<br />

potential APC functions of PDCs might be m<strong>as</strong>ked by the presence of cDCs and thus<br />

decided to test naïve T cell responses in mice that were conditionally depleted of<br />

cDCs.<br />

Surprisingly, PDCs could prime CD4 + T <strong>cells</strong> in the LNs, but not in cDC-depleted<br />

spleens. It remains unclear whether this discrepancy results from the distinct organ<br />

architecture or reflects an inherent difference between splenic and LN PDCs. In<br />

support of the <strong>for</strong>mer, PDCs are in naïve mice located in the red pulp and T cell area,<br />

but rarely in the marginal zone, which is the <strong>site</strong> of blood-borne antigen entry to the<br />

spleen [182]. Moreover, when challenged splenic PDCs <strong>for</strong>m clusters and, compared<br />

with cDCs, show a delayed translocation to the T cell zone [182]. This migration<br />

might be critical <strong>for</strong> the spleen, while it may not be required in the LNs. Recruitment<br />

of PDCs to the LNs occurs via high endothelial venules (HEVs), and thus the <strong>cells</strong><br />

enter directly into the T cell area [183]. Even be<strong>for</strong>e antigen-carrying DCs arrive from<br />

the periphery, resident LN APCs are known to take up soluble antigens [171], which<br />

enter the deeper LNs through an intricate network of conduits [174]. Accessibility of<br />

antigen to LN PDCs is also highlighted by the observation that these <strong>cells</strong> readily<br />

79


ingested injected DQ-OVA (Fig 23), although, interestingly, we failed to label splenic<br />

PDCs by intravenous DQ-OVA injection (data not shown). Alternatively, the unique<br />

ability of LN PDCs to prime CD4 + T <strong>cells</strong> might indicate that these <strong>cells</strong> differ from<br />

their splenic counterpart. Indeed, even when we targeted antigen directly to both LN<br />

and spleen PDCs and overrode the need <strong>for</strong> translocation (by an in vitro read-out, Fig.<br />

27), splenic PDCs failed to prime CD4 + T <strong>cells</strong> (Fig. 27). Interestingly, LN PDCs are<br />

characterized by a distinct expression pattern of activation markers, such <strong>as</strong> Sca-1,<br />

and are poor producers of IFN-α compared to Sca-1 + BM PDCs (J.A.A. Fischer and<br />

A. Dzionek, unpublished observation). However, a more detailed comparative study<br />

will be required to investigate qualitative differences between splenic and LN PDCs.<br />

Importantly, the antigen challenge results in a rapid PDC influx to the LNs and in our<br />

experimental set-up we do not discriminate between LN-resident and recruited PDCs.<br />

Although we would like to stress that we do not want to compare the efficiency of<br />

cDCs and PDCs in T cell <strong>priming</strong> potential, we establish that PDCs can prime naïve<br />

CD4 + T <strong>cells</strong>. Absence of cDCs is arguably an artificial situation unlikely to occur<br />

under physiological circumstances. However, we predict that pathogens that<br />

specifically target or activate PDCs will elicit exclusive CD4 + T cell responses<br />

supporting humoral, but not cytotoxic immunity. PDC-specific targeting could be<br />

mediated by endocytic receptors, such <strong>as</strong> the Siglec-H [18,164], which are expressed<br />

on murine PDCs, but not cDCs. So far, murine PDC-specific pathogen sensors have<br />

not been reported, although PDCs are characterized by higher expression of TLR7<br />

and TLR9 [184], which are also expressed by human PDCs, but absent from human<br />

interstitial DCs [25,185]. The unique <strong>priming</strong> of CD4 + , but not CD8 + T cell responses<br />

by PDCs could result from exclusive presentation of exogenous antigens on MHC II,<br />

but not MHC I molecules. As suggested earlier [149], PDCs might lack the<br />

phagosome-to-cytosol pathway required <strong>for</strong> cross-presentation. Importantly, this<br />

would spare the <strong>priming</strong> PDCs from a unique negative-feedback mechanism, through<br />

which CTLs kill APC and thereby terminate the <strong>priming</strong> process [186,187]. The<br />

inability to cross-present would render PDCs resistant to the CTLs elicited by cDCs.<br />

In the course of an ongoing response to a given antigen, the APC balance might<br />

there<strong>for</strong>e be shifted from cDCs to PDCs and, <strong>as</strong> a result, <strong>priming</strong> of CD4 + T <strong>cells</strong> and<br />

potentially humoral responses might be favored. As CD4 + T <strong>cells</strong> also critically<br />

contribute to the development of CD8 + T cell memory [188], the APC function of<br />

PDCs might have a general impact on the shape of immune responses.<br />

80


We show that in LNs, PDCs efficiently trigger productive naïve CD4 + T cell<br />

responses. Interestingly, and in contr<strong>as</strong>t to cDC-triggered responses, PDC-triggered<br />

CD4 + T cell responses are not <strong>as</strong>sociated with concomitant CD8 + T cell expansion.<br />

Instead, PDCs initiate unique CD4 + Th cell-dominated primary immune responses.<br />

81


5. Final Discussion<br />

Dendritic <strong>cells</strong> (DCs) are part of the mononuclear phagocyte system and <strong>for</strong>m a<br />

complex body-wide network comprised of multiple subsets, currently largely defined<br />

according to anatomic location and phenotype. The functionally definition of these<br />

DC subsets and detection of potential t<strong>as</strong>k division within the DC compartment,<br />

arguably requires the study of DCs in the in vivo context. In my PhD thesis I hence<br />

took advantage of two recently established transgenic mouse systems that allow (1)<br />

the visualization of DCs in physiological context and (2) the ablation of DCs from the<br />

otherwise intact organism. Using these novel approaches I focused on the role of DCs<br />

in the bone <strong>marrow</strong> and investigated the function of a unique population of DCs, the<br />

pl<strong>as</strong>macytoid dendritic <strong>cells</strong>.<br />

Characterization of BM-resident DCs: Flow cytometry analysis revealed the<br />

existence of a defined population of CD11c hi , MHC-II + DC in the BM of<br />

unchlallenged mice. BmDCs differ from splenic DCs in that they incre<strong>as</strong>ed levels of<br />

F4/80, Gr-1, CD103 and CD115, but also relatively high amounts of major MHC<br />

cl<strong>as</strong>s I and II, CD86, CD44 and CD40, suggesting that they have been activated.<br />

Two-photon imaging of cranial BM in vivo revealed characteristic spindle-shaped<br />

DCs in distinct periv<strong>as</strong>cular clusters surrounding the distinct v<strong>as</strong>cular sinuses.<br />

However, notably, the two-photon imaging shows that bmDCs are generally not in<br />

direct contact with the vessel walls. In this respect, it is interesting that Nag<strong>as</strong>awa and<br />

colleagues have reported that most v<strong>as</strong>cular sinuses are lined by a population of<br />

reticular <strong>cells</strong> with abundant expression of the chemokine CXCL12, and is essential<br />

<strong>for</strong> BM mature B <strong>cells</strong>. However, it remains to be shown whether these so-called<br />

CAR <strong>cells</strong> exists in the BM immune niches [190].<br />

Conditional ablation of CD11c high DCs resulted in a selectively loss of BM B <strong>cells</strong>.<br />

The ablation of DCs did not substantially affect the numbers of developing pro-B<br />

<strong>cells</strong>, pre-B <strong>cells</strong> or immature B <strong>cells</strong> in the BM, or the numbers of B <strong>cells</strong> in spleen<br />

or LNs, demonstrating a specific role <strong>for</strong> DCs in the maintenance of recirculating<br />

mature BM B <strong>cells</strong>. Two-photon imaging revealed that DCs in the niches organized in<br />

situ together with adoptively transferred B and T <strong>cells</strong>, suggesting that direct<br />

interactions between the DCs and B <strong>cells</strong> are required <strong>for</strong> maintaining mature B <strong>cells</strong><br />

in the BM. To understand how bmDCs maintain mature B <strong>cells</strong> in the BM we<br />

adoptively transferred B <strong>cells</strong> overexpressing a Bcl-2 transgene into the CD11c-<br />

82


DTRtg mice prior to DC ablation. The transferred Bcl-2 transgenic B <strong>cells</strong> were not<br />

affected by DC ablation, demonstrating that bmDCs are not required <strong>for</strong> mature B<br />

<strong>cells</strong> to home to BM, but more likely provide survival signals to B <strong>cells</strong> after they<br />

arrive. Given the strong preference of mature splenic B <strong>cells</strong> <strong>for</strong> BAFF, BAFF w<strong>as</strong> a<br />

likely candidate <strong>for</strong> mediating survival of mature BM B <strong>cells</strong>. We tested this by<br />

generating mixed BM chimer<strong>as</strong> using BAFF-deficient and CD11c-DTRtg BM, in<br />

which treatment with DTx selectively ablated BAFF-expressing DCs, so the mice<br />

contained only BAFF-deficient DCs. Surprisingly, BAFF expression by bmDCs w<strong>as</strong><br />

not required to retain recirculating BM B <strong>cells</strong>. T <strong>cells</strong> were also seen in periv<strong>as</strong>cular<br />

niches with bmDCs and B <strong>cells</strong>; however, the DC ablation did not affect their<br />

numbers, suggesting that bmDCs do not act via T <strong>cells</strong> to keep mature B <strong>cells</strong> in the<br />

BM. Another survival factor, MIF, turned out to be the B cell rescuer. MIF is a<br />

chemokine that activates phosphatidylinositol-3-OH kin<strong>as</strong>e-Akt signaling pathways<br />

and induces anti-apoptotic signals via binding CD44–CD74 receptor complexes<br />

expressed by B <strong>cells</strong>. The BM of MIF-deficient mice contained fewer recirculating<br />

mature B <strong>cells</strong>, and DC ablation in mixed BM chimer<strong>as</strong> generated with MIF-deficient<br />

and CD11c-DTRtg BM produced similar deficiencies in mature BM B-cell numbers<br />

<strong>as</strong> seen in CD11c-DTRtg mice. Importantly, although MIF expression w<strong>as</strong> detected<br />

by bmDCs, monocytes and macrophages, MIF expression by the latter <strong>cells</strong> w<strong>as</strong> not<br />

sufficient to rescue B-cell survival to BM following DC ablation, directly implicating<br />

bmDCs <strong>as</strong> the critical source of MIF. In addition, given that MIF is a noncognate<br />

ligand of CXCR4 [191], the interaction between MIF and CXCL12-CXCR4 signaling<br />

may be crucial <strong>for</strong> maintaining mature B <strong>cells</strong>.<br />

These experiments, thus, revealed a novel, previously unappreciated role of DC in<br />

lymphocyte homeost<strong>as</strong>is that is independent of their APC function. Moreover, this<br />

study provides a new b<strong>as</strong>is <strong>for</strong> understanding the spatio-temporal regulation of the<br />

development and function of lympho-hematopoietic <strong>cells</strong> in three-dimensional<br />

microenvironments in the BM.<br />

Although DCs in the BM are capable of maintaining the homeost<strong>as</strong>is of mature B<br />

<strong>cells</strong>, a t<strong>as</strong>k that in the spleen is carried out by nonhematopoietic <strong>cells</strong>, we<br />

unexpectedly found that the origin of DCs in both lymphoid organs is similar. BmDCs<br />

originated in the steady state exclusively from MDPs, rather than from recirculating<br />

monocytes. This similarity raised the question, whether bmDCs can act <strong>as</strong> APCs and<br />

initiate primary T-cell responses – a function thought to be restricted to secondary<br />

83


lymphoid organs. Our focus on primary BM T-cell responses to blood-borne antigen<br />

revealed clusters of specific T <strong>cells</strong> in are<strong>as</strong> enriched with DCs, in contr<strong>as</strong>t to<br />

unresponsiveness of polyclonal T <strong>cells</strong> in the same niches. Antigen-specific T-cell<br />

arrest w<strong>as</strong> accompanied by prompt early cell activation, me<strong>as</strong>ured by CD69. To<br />

further establish if bmDCs are critical APCs that initiate BM T-cell responses, we<br />

analyzed the responses of naïve CD8 + T <strong>cells</strong> to antigen challenge in mice that were<br />

depleted of DCs. Surprisingly, ablation of bmDCs did not impair the capacity of<br />

antigen-specific T <strong>cells</strong> to cluster. In addition, in the BM of these mice, T <strong>cells</strong><br />

showed CD69 upregulation, in vivo proliferation after antigen challenge and IFN-γ<br />

production. To identify the enigmatic cell type that prime BM CD8 + T <strong>cells</strong> in<br />

absence of DCs, we took advantage of chimer<strong>as</strong>, in which H-2K b -deficient recipients<br />

were reconstituted with Cx3cr1 gfp/+ BM. Surprisingly, preliminary results indicate that<br />

MHC cl<strong>as</strong>s I-proficient hematopoetic <strong>cells</strong> that are in the presence of MHC cl<strong>as</strong>s I-<br />

deprived stroma incapable of sustaining primary CD8 + T-cell response. These data<br />

suggest a novel unique role of nonhematopoietic <strong>cells</strong> in the BM.<br />

In contr<strong>as</strong>t to LN responses that function in preventing local spread of a dise<strong>as</strong>e, BM<br />

could control a systemic dise<strong>as</strong>e, similarly to the spleen. Further investigation is<br />

required to determine to which antigenic challenges and pathogens the BM responds.<br />

Indeed, previous research h<strong>as</strong> shown that pathogens can enter the BM; <strong>for</strong> example,<br />

Staphylococcus aureus can enter the BM and cause osteomyelitis [192], and<br />

intracellular pathogens like Listeria monocytogens induce cytotoxic T cell responses<br />

in the BM [193]. It remains unclear what distinguishes the immune responses initiated<br />

in the BM from those in the spleen. One hypothesis is that the BM <strong>as</strong>sumes an<br />

important role against systemic infections only when the spleen is not functional.<br />

Another alternative is that the BM functions alongside the spleen to stave off systemic<br />

infections. A third option is that the BM and spleen specialize in defense against<br />

different pathogens, which are more accessible to one organ or the other.<br />

Characterization of Pl<strong>as</strong>macytoid dendritic <strong>cells</strong> functions: Pl<strong>as</strong>macytoid dendritic<br />

<strong>cells</strong> (PDCs) play a pivotal role in anti-viral immune defense. Type I IFN secretion by<br />

PDC orchestrates both innate and adaptive immune responses highlighting them <strong>as</strong><br />

critical cytokine-secreting accessory <strong>cells</strong>. We studied the involvement of PDCs in T<br />

cell stimulation and their capacity to act <strong>as</strong> APCs in naïve T cell <strong>priming</strong>. In most<br />

84


experimental systems any putative in vivo T cell <strong>priming</strong> potential of PDCs could be<br />

m<strong>as</strong>ked by the presence of cDCs. To this end we took advantage of mice that were<br />

conditionally depleted of cDCs, but retain PDCs. When mice were challenged with<br />

s.c. antigen injection, PDCs in draining LNs efficiently ingested and processed<br />

antigen. Moreover, we found that in cDC-depleted LNs, PDCs readily stimulated<br />

antigen-specific TCR-transgenic CD4 + T <strong>cells</strong>. CD4 + T <strong>cells</strong> in the LNs proliferated<br />

and produced IFN-γ after antigen injection, even when the cDC ablation w<strong>as</strong><br />

combined with the removal of B <strong>cells</strong> or LCs. Importantly, PDCs were, however,<br />

unable to concomitantly activate TCR-transgenic CD8 + T <strong>cells</strong>, a fact that served <strong>as</strong> a<br />

functional confirmation and internal control <strong>for</strong> the absence of cDCs from the nodes.<br />

The <strong>priming</strong> potential of PDCs w<strong>as</strong> subsequently confirmed by an antigen-targeting<br />

strategy employing an mPDCA1-OVA conjugate. PDCs, but not cDCs sorted from<br />

LNs of the conjugate-immunized mice, were able to promote the proliferative<br />

expansion in an in vitro T-cell proliferation <strong>as</strong>say. The above results established that<br />

LN PDCs are bona fide DCs that can prime naive CD4 + T-cell responses. The<br />

inability of PDCs to stimulate CD8 + T-cell responses by PDCs could result from<br />

exclusive presentation of exogenous antigens on MHC cl<strong>as</strong>s II not MHC cl<strong>as</strong>s I that<br />

is, a lack the phagosome-to-cytosol pathway required <strong>for</strong> cross-presentation. This<br />

would be supported by the notion that even CpG-matured PDCs can only elicit<br />

responses of naive CD8 + T <strong>cells</strong> to endogenous, but not exogenous antigens.<br />

However, the cross-presentation potential of PDCs remains under debate.<br />

Interestingly, we found PDCs to be incapable to promote CD4 + T-cell <strong>priming</strong> in the<br />

spleens of DTx-treated CD11c-DTRtg mice. Thus, cDC ablation abrogated the<br />

<strong>priming</strong> of both TCR transgenic CD4 + and CD8 + T <strong>cells</strong>. It remains unclear, whether<br />

these results from the distinct tissue architecture of LNs and the spleen. Alternatively,<br />

it could indicate the existence of functionally distinct PDC subsets in different organs.<br />

In addition, the molecular b<strong>as</strong>is of the apparent T cell lineage-specific and organspecific<br />

T cell-stimulatory capacity of PDCs remains to be identified.<br />

Taken together, we believe our finding is an important addition to our understanding<br />

of the enigmatic cell type – PDC. Furthermore, given the emerging role of PDCs in<br />

the control of inflammation, it might be of critical importance <strong>for</strong> the clinic and<br />

vaccination, in particular the targeting of vaccine antigens to specific DC subsets,<br />

allo-graft rejection and auto-immunity.<br />

85


6. Materials and Methods<br />

6.1 Periv<strong>as</strong>cular clusters of dendritic <strong>cells</strong> provide critical survival<br />

signals to B <strong>cells</strong> in bone <strong>marrow</strong> niches<br />

Mice. The following mice were used: 8 - to 12-week-old C57BL/6 (CD45.2,<br />

CD45.1), BALB/c, CD11c-DTR transgenic (B6.FVB-Tg Itgax-DTR/GFP 57Lan/J)<br />

mice carrying a transgene encoding a human DTR-GFP fusion protein under the<br />

control of the murine CD11c promoter [42]; Cx3cr1 gfp/+ mice (B6.129P-<br />

Cx3cr1tm1Litt/J) harboring a targeted replacement of the Cx3cr1 gene by an<br />

enhanced GFP reporter gene [81]; RAG2 - / - mice (B6.129S6-Rag2tm1Fwa) [194],<br />

BAFF - / - mice [94], which (kindly provided by Biogen Idec., Inc., Cambridge, MA),<br />

MIF - / - mice [195] and Bcl-2 transgenic mice carrying a human Bcl-2 transgene driven<br />

by the Eμ immunoglobulin heavy chain enhancer [88]. Cx3cr1 gfp/+ mice were crossed<br />

with CD11c-DTR transgenic mice, generating CD11c-DTRtg Cx3cr1 gfp/+ mice. Bcl-2<br />

transgenic mice were crossed with mice bearing the CD45.1 allotype (B6.SJL-Ptprca<br />

Pep3b/BoyJ). Mixed [wt > wt], [CD11c-DTRtg > wt], [95% CD11c-DTRtg RAG1 - / - /<br />

5% wt > wt], [BAFF - / - > wt], [50% BAFF - / - / 50% CD11c-DTRtg > wt] and [50%<br />

MIF - / - / 50% CD11c-DTRtg / > wt] BM chimer<strong>as</strong> were generated <strong>as</strong> reported. For DC<br />

ablation, mice were inoculated intraperitoneally every day <strong>for</strong> 5 d with 16 ng DTx/g<br />

body weight. DC depletion w<strong>as</strong> analyzed 24 h later. All animals were maintained in<br />

specific pathogen-free (SPF) conditions and handled according to protocols approved<br />

by the <strong>Weizmann</strong> Institute Animal Care Committee <strong>as</strong> per international guidelines.<br />

Flow cytometry analysis. Staining reagents used in this study included the PEcoupled<br />

antibodies anti-B220, CD11b, CD62L, CD103, F4/80 (Serotec), CD4, CD8,<br />

CD86, CD80, CD44, CD40, Gr1 (Ly6C/G), CD115, H-2K b , I-A/I-E, IgD, CD93<br />

(AA4.1, early B lineage antigen), IgM; the biotinylated antibodies anti-B220 and<br />

anti- CD45.1; the APC-coupled antibodies anti-CD11c and anti-SA; and the FITCcoupled<br />

antibodies anti-IgD and anti-CD23. Unless indicated otherwise, the reagents<br />

were obtained from eBioscience. The <strong>cells</strong> were analyzed on a FACSCalibur<br />

cytometer using CellQuest software (Becton-Dickinson).<br />

Mixed-Leukocyte Reaction. Stimulator monocytes, MΦ and DCs were isolated from<br />

spleens and BM of CD11c-DTRtg mice by high speed sorting using the FACSAria<br />

(Becton-Dickinson) and were irradiated with 2500 rads. Responder T <strong>cells</strong> were<br />

enriched from spleens of BALB/c mice by positive selection with anti-CD4<br />

86


microbeads (Miltenyi Biotec). Stimulator DCs in indicated numbers were cultured<br />

with 1x10 5 responder CD4 + T <strong>cells</strong> (BALB/c). After 72 h, cultures were pulsed after<br />

with 1μCi of [H 3 ] thymidine, and incorporation w<strong>as</strong> me<strong>as</strong>ured 16 h later.<br />

Two-photon microscopy. Syngeneic splenic B <strong>cells</strong> were purified by depletion with<br />

immunomagnetic anti-CD43 beads (Miltenyi Biotec), and were labeled with Hoechst<br />

33342 (Molecular Probes), then 3-5x10 7 <strong>cells</strong> were injected intravenously into<br />

CD11c-DTRtg Cx3cr1 gfp/+ mice. CD4 + and CD8 + T cell grafts were isolated from<br />

spleens of naïve wt donor mice by magnetic-activated cell sorting (MACS), then were<br />

labeled with Hoechst 33342 and injected alone or in combination with B <strong>cells</strong> labeled<br />

with CMTMR (5-(and-6)-(((4-chloromethyl)benzoyl)amino)-tetramethylrhodamine;<br />

Molecular Probes) (1:1 mixture) into recipients, followed by analysis 5 – 24 h after<br />

engraftment. Immediately be<strong>for</strong>e imaging, tetramethylrhodamine isothiocyanatedextran<br />

TRITC-dextran (150 kDa; Sigma) w<strong>as</strong> injected into the mice <strong>for</strong> visualization<br />

of BM v<strong>as</strong>culature. Mice were anesthetized and a small incision made in the scalp to<br />

expose the underlying dorsal skull surface [52]. Two-photon imaging w<strong>as</strong> per<strong>for</strong>med<br />

on skull BM with a Zeiss LSM 510META-NLO two-photon fluorescence microscope<br />

equipped with a x20 or x40 objective. A MaiTai HP Ti:S one-box tunable l<strong>as</strong>er<br />

(Spectra Physics, USA) w<strong>as</strong> used <strong>for</strong> two-photon excitation. Image acquisition w<strong>as</strong><br />

per<strong>for</strong>med using LSM 510 acquisition software. For two-photon excitation of Hoechst<br />

and CMTMR, the l<strong>as</strong>er w<strong>as</strong> tuned to 800 nm, and the emission filter sets were 435-<br />

485 and 535-590 nm, respectively. For excitation of GFP, the l<strong>as</strong>er w<strong>as</strong> tuned to 920<br />

nm and the emission filter set w<strong>as</strong> 500-550 nm.<br />

ELISPOT <strong>as</strong>say. Mice were intraperitoneally immunized with 120 μg of NP-Ficoll<br />

(Biosearchtech, CA). Four days later, BM w<strong>as</strong> isolated and plated on 96-well plates<br />

coated with 5 μg/ml NP 30 -BSA (Biosearchtech, CA). To detect NP-specific IgMsecreting<br />

AFCs, biotinylated anti-IgM (Biotec) w<strong>as</strong> applied, followed by<br />

EXTRAvidin-AP (Sigma).<br />

RT-PCR <strong>as</strong>say. Standard methods were used to prepare total cellular RNA and firststrand<br />

cDNA from sorted monocytes, macrophages and DCs isolated from the BM of<br />

wt C57BL/6 mice. The following primer pairs were used (5’ – 3’; <strong>for</strong>ward / reverse):<br />

MIF, GAACACCAATGTTCCCCGCGC / GCGAAGGTGGAACCGTTCCAG<br />

(327bp product); HPRT1: GGGGGCTATAAGTTCTTTGC /<br />

TCCAACACTTCGAGAGGTCC (320bp product). BM RNA of MIF - / - mice 42 served<br />

<strong>as</strong> negative control.<br />

87


Statistical analysis. All statistics were generated using a Students’s t-test. All error<br />

bars in diagrams and numbers following a ± sign are standard deviations (s.d.).<br />

6.2 The bone <strong>marrow</strong> immune niche: <strong>Bone</strong> <strong>marrow</strong> <strong>as</strong> a <strong>priming</strong> <strong>site</strong><br />

<strong>for</strong> naïve <strong>CD8+</strong> T <strong>cells</strong><br />

Mice. The following mice were used: 8 - to 12-week-old C57BL/6 (CD45.2,<br />

CD45.1), CD11c-DTR transgenic (B6.FVB-Tg Itgax-DTR/GFP 57Lan/J) mice<br />

carrying a transgene encoding a human DTR-GFP fusion protein under the control of<br />

the murine CD11c promoter; Cx3cr1 gfp/+ mice (B6.129P-Cx3cr1tm1Litt/J) harboring a<br />

targeted replacement of the Cx3cr1 gene by an enhanced GFP reporter gene; OT-I<br />

(C57BL/6) TCR transgenic mice harboring OVA-specific CD8 + T <strong>cells</strong> [166]; OT-II<br />

(C57BL/6) TCR transgenic mice harboring OVA-specific CD4 + T <strong>cells</strong> [162]; β-<br />

actin-Enahnced Cyan Fluorescent Protein (β-actin-ECFP) (stock #004218 Jackson<br />

laboratories); Cx3cr1 gfp/+ mice which were crossed with CD11c-DTRtg mice creating<br />

CD11c-DTRtg Cx3cr1 gfp/+ mice. The following chimer<strong>as</strong> were created: C57BL6<br />

reconstituted with CD11c-DTRtg Cx3cr1 gfp/+ BM [CD11c-DTRtg Cx3cr1 gfp/+ > wt];<br />

or with CD11c-DTRtg BM [CD11c-DTRtg > wt]; H-2K b -deficient mice [130]<br />

reconstituted with Cx3cr1 gfp/+ BM [Cx3cr1 gfp/+ > H-2K b- / - ].<br />

Fluorescent reagents. Fluorescent reagents used in this study included: non-targeted<br />

quantum dots (2 µM Qtracker® 655, Molecular Probes), CellTracker Orange<br />

CMTMR (Molecular Probes), Fluorescin isothicyanate (FITC)-dextran average<br />

molelcular weight 500,000 (Sigma).<br />

Live two-photon microscopy of the BM. The imaging system is the Ultima<br />

Multiphoton Microscope from Prairie Technologies®. It incorporates a pulsed Mai<br />

Tai Ti-sapphire l<strong>as</strong>er (Newport Corp.) that produces an intense 3 Watt beam,<br />

allowing deep imaging with minimal light scattering. Highly sensitive Gallium-<br />

Arsenide photo-multiplier tubes and optimized optical elements minimize pulse<br />

dispersion and maximize light collection. The system is equipped with a setup <strong>for</strong><br />

intravital imaging (warming elements, respiration and anesthesia).<br />

For intravital imaging, mice are anesthetized with 100 mg ketamine, 15 mg xylazine<br />

and 2.5 mg acepromazine per gram and are kept anesthetized with hourly injections of<br />

half the dose. The hair on the skullcap is removed using lotion containing urea<br />

88


(ORNAT 19) and trimmer, the scalp is incised at the midline, the frontoparietal bone<br />

is exposed and an imaging chamber is glued to the skull using cyanoacrylate-b<strong>as</strong>ed<br />

glue. During imaging, mice are supplied with oxygen and their core temperature is<br />

maintained around 37°C with a warming plate. Mineralized bone is identified b<strong>as</strong>ed<br />

on the second harmonic signal. To visualize blood vessels, mice are i.v. injected with<br />

a contr<strong>as</strong>ting agent – non-targeted quantum dots. To create time-lapse sequences, we<br />

typically scan a 50-200 μm-thick volume of tissue at 5 μm Z-steps and 20-second<br />

intervals. The excitation wavelength used is between 870-910 nm. Cell movements<br />

were analyzed using Volocity® software (Improvision Ltd.) to determine cell<br />

coordinates, tracking over time and speed.<br />

Adoptive cell transfers. Syngeneic splenic B <strong>cells</strong> were purified from β-actin-ECFP<br />

by depletion with immunomagnetic anti-CD43 beads (Miltenyi Biotec CD4 + and<br />

CD8 + T cell grafts were isolated by MACS from spleens and LNs of OT-II and OT-I<br />

donor mice, respectively, then were labeled with 5 μM CMTMR. Polyclonal CD8 + T<br />

<strong>cells</strong> were isolated from spleens and LNs of naïve wild-type donor mice and labeled<br />

with 5 μM CMTMR. Then, 5-7x10 7 B <strong>cells</strong> and T <strong>cells</strong> (1:1 mixture) were i.v.<br />

injected into recipients, followed by analysis 4 h after engraftment.<br />

Immunizations. For peptide immunization, approximately 15 min after beginning the<br />

imaging, 50 μg of SIINFEKL peptide and 0.5 mg of dextran-FITC were i.v. injected.<br />

For analysis of response to intact protein, Cx3cr1 gfp/+ recipient mice were injected<br />

with 0.1-1 mg OVA protein (Sigma). Then, 24 h after the immunization, purified β-<br />

actin ECFP polyclonal CD8 + <strong>cells</strong> and CMTMR-labeled OT-I CD8 + T <strong>cells</strong> were<br />

transferred and 2 h later mice were prepared <strong>for</strong> intravital imaging. For analysis of<br />

CD69 expression, mice were adoptively transferred with 1x10 7 OT-I CD8 + T <strong>cells</strong>, 16<br />

h later mice were challenged with 0.5 mg OVA and 2 h later were sacrificed <strong>for</strong> flow<br />

cytometry analysis.<br />

Analysis of T cell proliferation. OT-I CD8 + T <strong>cells</strong> were isolated from spleens and<br />

LNs of respective mice, enriched by MACS cell sorting with anti-CD8 antibodies<br />

according to the manufacturer's protocol (Miltenyi Biotec) and labeled with CFSE<br />

(Molecular Probes, C-1157) (57). CFSE-labeled T <strong>cells</strong> (1x10 7 / mouse) were injected<br />

into the tail veins of the recipient mice.<br />

Intracellular cytokine staining. Mice were i.v. immunized with 0.1 mg OVA in 35<br />

μg CpG nucleotide 1668 (TIB MOLBIOL). Two days later, mice were in vivo<br />

89


estimulated by i.v. injection of 100 μg OVA peptide (residues 323-329) 2 h be<strong>for</strong>e<br />

the removal of the spleen and BM. The tissue w<strong>as</strong> minced and incubated in 1 mg/ ml<br />

of collagen<strong>as</strong>e D (Roche Biochemicals) <strong>for</strong> 20 min at 37 °C in medium supplemented<br />

with 10 μg/ml brefeldin A (Calbiochem). Cells were fixed in 2% <strong>for</strong>maldehyde,<br />

permeabilized with 0.3% saponin, and stained <strong>for</strong> anti-CD45.1, anti-CD8 and anti-<br />

IFN-γ antibodies.<br />

Flow Cytometry. Staining reagents used in this study included: biotinylated anti-<br />

CD45.1; APC-coupled anti CD8 and IFN-γ; FITC-coupled anti-CD8, CD69 and<br />

CD44, PE-coupled streptavidin (eBioscience). Cells were analyzed on a FACSCalibur<br />

with CellQuest software (Becton Dickinson).<br />

Analysis of cell motility. Cell movement w<strong>as</strong> analyzed with Volocity® software<br />

(Elmer Perkin©). To calculate cell speed, the coordinates of each cell were<br />

determined and tracked over time and motility parameters were calculated. To<br />

eliminate motion artifacts, displacements smaller than 2µm were filtered out of the<br />

cell tracks.<br />

Statistics. Student’s t-test w<strong>as</strong> used <strong>for</strong> comparison of two groups.<br />

6.3 Organ-dependent in vivo <strong>priming</strong> of naïve CD4 + , but not CD8 + T<br />

<strong>cells</strong> by pl<strong>as</strong>macytoid dendritic <strong>cells</strong><br />

Mice. CD11c-DTR transgenic (B6.FVB-Tg Itgax-DTR/GFP 57Lan/J) mice; OT-I<br />

(C57BL/6) TCR transgenic mice harboring OVA-specific CD8 + T <strong>cells</strong>; DO11.10<br />

(BALB/c) and OT-II (C57BL/6) TCR transgenic mice harboring OVA-specific CD4 +<br />

T <strong>cells</strong>; B cell deficient mice (C57BL/10-IgH-6tm1Cgn (μMt) [172] and LanDTRtg<br />

mice carrying the DTR-GFP gene under the Langerin promoter [173]. CD11c-DTRtg<br />

LanDTRtg and CD11c-DTRtg μMT mice were obtained by intercrossing of the<br />

respective lines. Mixed [CD11c-DTRtg > wt] BM chimer<strong>as</strong> were generated. For<br />

systemic cDC depletion mice were injected i.p. with 4 ng /g body weight DTx (Sigma<br />

D-2918). For local cDC depletion in popliteal LNs, animals were injected with 20 ng<br />

DTx s.c. into hind footpads. All mice were used at the age of 6 to 12 wks, maintained<br />

under specific pathogen-free conditions and handled according to protocols approved<br />

by the <strong>Weizmann</strong> Institute Animal Care Committee <strong>as</strong> per international guidelines.<br />

Isolation of CD11c high DCs and Pl<strong>as</strong>macytoid DCs. Peripheral LNs or spleens were<br />

digested with 1 mg/ ml of collagen<strong>as</strong>e D (Roche Biochemicals) <strong>for</strong> 45 min at 37° C.<br />

90


Tissues were mechanically disrupted in PBS, and <strong>for</strong> spleens, RBC lysis w<strong>as</strong><br />

per<strong>for</strong>med by incubation with 1 ml ACK buffer (0.15 M NH 4 Cl, 0.1 M KHCO 3 , 1<br />

mM EDTA and PBS) <strong>for</strong> 1 min at RT. PDCs were enriched by MACS cell sorting<br />

according to the manufacturer's protocol (Miltenyi Biotec).<br />

In vitro Proliferation <strong>as</strong>say. BALB/c mice were immunized i.v. (20 μg OVA or 10<br />

μg mPDCA-1-OVA) and s.c. to hind footpads (10 μg OVA or mPDCA-1-OVA.<br />

Twenty four hours later spleens, popliteal and inguinal LNs were harvested and cDCs<br />

and PDCs were purified by high-speed cell sorting according to CD11c and mPDCA-<br />

1 expression using a FACSAria (Beckton-Dickinson). Positive fraction of PDCs<br />

contained less than 0.01% contaminating CD11c high cDCs. DCs (10 4 ) or PDCs (2-<br />

3*10 4 ) were cultured with 10 5 DO11.10 CD4 + T <strong>cells</strong>. Cultures were pulsed after 72 h<br />

with 1μCi of [H 3 ] thymidine, and incorporation w<strong>as</strong> me<strong>as</strong>ured 16 h later.<br />

INF-α secretion <strong>as</strong>say. MACS-enriched splenic PDCs were cultured at 2 x 10 6 <strong>cells</strong>/<br />

ml in complete RPMI-1640 <strong>for</strong> 20 h with 400 HAU/ml influenza virus A/Tex<strong>as</strong>/1/77<br />

(kindly provided by R. Arnon, The <strong>Weizmann</strong> Institute, Rehovot). Supernatants were<br />

<strong>as</strong>sayed using an INF-α ELISA kit (Per<strong>for</strong>mance Biomedical Laboratories).<br />

Conjugation of OVA to anti-mPDCA-1 F(ab’) 2 fragments and specificity test. AntimPDCA-1<br />

monoclonal antibody (rat IgG2b, κ; Miltenyi Biotec GmbH) w<strong>as</strong> digested with<br />

Pepsin A (Sigma-Aldrich). After size fractionation (Superdex-200 16/60 gel filtration<br />

column; GE Healthcare), F(ab’) 2 fragments were conjugated to OVA (Sigma-Aldrich)<br />

activated with succinimidyl 4-(N-maleimidomethyl) cyclohexane-1-carboxylate (SMCC,<br />

Pierce Chemicals Co.) according to the manufacturer’s protocol. F(ab’) 2 -OVA conjugates<br />

were size fractionated and sterile filtrated (Millex GV filter unit 0.22 μm; Millipore) <strong>for</strong> in<br />

vivo application. Construct integrity w<strong>as</strong> evaluated by Western blot analysis with HRPconjugated<br />

polyclonal rabbit anti-rat Ig (H+L; Jackson ImmunoResearch Laboratories)<br />

and HRP-coupled rabbit anti-OVA antibody (Research Diagnostics Inc.) respectively.<br />

OVA content w<strong>as</strong> determined by ELISA to be around 75 ng OVA / μg mPDCA-1 F(ab’) 2 -<br />

OVA conjugate. To demonstrate in vivo specificity, we injected 10-25 μg FITCconjugated<br />

anti-mPDCA-1-F(ab’) 2 -OVA i.v. or into the hind footpads. Three hours later,<br />

spleens or pooled popliteal and inguinal LNs were analyzed <strong>for</strong> PDC-specific targeting.<br />

Immunohistochemistry. Paraffin sections (4μm) of popliteal LNs were<br />

deparaffinized and treated with 2 ml HCl, H 2 O 2 in 50% methanol and 50% PBS <strong>for</strong> 15<br />

min at RT. For antigen exposure, sections were microwaved in 10 mM sodium citrate<br />

91


pH 6.0. After blocking with 20% heat-inactivated normal goat serum and 0.1% Triton<br />

X-100 in PBS, sections were incubated overnight at RT with polyclonal biotinylated<br />

goat anti-GFP antibody (ab6658, Abcam). Staining w<strong>as</strong> revealed with an avidin-biotin<br />

peroxid<strong>as</strong>e complex (Vect<strong>as</strong>tain Elite ABC kit, Vector Laboratories). Finally, the<br />

sections were stained with haematoxylin-Mayer (Finkelman).<br />

Analysis of T cell proliferation. TCR transgenic T <strong>cells</strong> were isolated from spleens<br />

and LNs of respective mice, enriched by MACS cell sorting with anti-CD8 or anti-<br />

CD4 antibodies according to the manufacturer's protocol (Miltenyi Biotec) and<br />

labeled with CFSE (Molecular Probes, C-1157) (57). CFSE-labeled T <strong>cells</strong> (1 - 2 x<br />

10 6 / mouse) were injected into the tail veins of the recipient mice.<br />

Immunization. Mice were immunized with cell-<strong>as</strong>sociated antigen by i.v. injection of<br />

3 x 10 7 Ova-loaded β2m -/- splenocytes [8]. Soluble Ova w<strong>as</strong> injected i.v. (10μg/<br />

mouse) or s.c. to hind footpads (10μg/ footpad). Keyhole Limpet Hemocyanin (KLH)<br />

(Sigma) w<strong>as</strong> injected s.c. to hind footpads (10μg/ footpad). The anti-DEC205-OVA<br />

conjugate (kindly provided by K. Mahnke, University<br />

Hospital Heidelberg, Heidelberg,Germany) w<strong>as</strong> used at a dose of 7ng/ footpad.<br />

F(ab’) 2 and anti-mPDCA-1-F(ab’) 2 -OVA were injected at a dose of 8 μg/ footpad. For<br />

me<strong>as</strong>urement of Ag uptake mice were immunized with DQ-OVA (Invitrogen) by<br />

hind footpad injection of 10μg/ footpad.<br />

Intracellular cytokine staining. Mice were immunized with 25 μg OVA in 5 nmol/<br />

footpad CpG nucleotide 1668 (TIB MOLBIOL). Two days later, mice were in vivo<br />

restimulated by i.v. injection of 100 μg OVA peptide (residues 323-329) 2 h be<strong>for</strong>e<br />

the removal of popliteal LNs. The tissue w<strong>as</strong> minced and incubated in 1 mg/ ml of<br />

collagen<strong>as</strong>e D (Roche Biochemicals) <strong>for</strong> 20 min at 37 °C in medium supplemented<br />

with 10 μg/ml brefeldin A (Calbiochem). Cells were fixed in 2% <strong>for</strong>maldehyde,<br />

permeabilized with 0.3% saponin, and stained <strong>for</strong> anti-CD45.1, anti-CD4 and anti-<br />

IFN-γ antibodies.<br />

Flow cytometry analysis. The staining reagents used in this study included the PEcoupled<br />

anti-CD4, CD19, CD45.1, CD11c, CD45RB, CD8 and KJ126 (DO11.10<br />

clonotype); biotinylated anti-CD45.1 and mPDCA-1; APC-coupled CD11c, CD4,<br />

IFN-γ, and CD8; the FITC- coupled anti-CD4; PE-Cy5.5-coupled anti-B220 (Caltag<br />

laboratories) and PerCP- coupled anti-CD4 and CD8 antibodies. Unless indicated<br />

otherwise, the reagents were obtained from Pharmingen or eBioscience. The anti-<br />

92


Siglec-H antibody (440c) w<strong>as</strong> provided by M.Colonna (W<strong>as</strong>hington University<br />

School of Medicine, St. Louis, MO). Cells were analyzed on FACSCalibur cytometer<br />

using CellQuest software (Becton-Dickinson).<br />

93


7. References<br />

1. Banchereau, J. & Steinman, R.M. Dendritic <strong>cells</strong> and the control of immunity.<br />

Nature 392, 245-52 (1998).<br />

2. Steinman RM, Hawiger D, Nussenzweig MC. Tolerogenic dendritic <strong>cells</strong>. Annu<br />

Rev Immunol. 21, 685-711 (2003).<br />

3. Liu, Y.J. IPC: professional type 1 interferon-producing <strong>cells</strong> and pl<strong>as</strong>macytoid<br />

dendritic cell precursors. Annu Rev Immunol 23, 275-306 (2005).<br />

4. Ruedl, C., P. Koebel, M. Bachmann, M. Hess, and K. Karjalainen. Anatomical<br />

origin of dendritic <strong>cells</strong> determines their life span in peripheral lymph nodes. J<br />

Immunol. 165, 4910-4916 (2000).<br />

5. Liu, K., Victora, GD., Schwickert, TA., Guermonprez, P., Meredith, MM., Yao,<br />

K., Chu, FF., Randolph, GJ., Rudensky, AY., Nussenzweig, M. In vivo analysis of<br />

dendritic cell development and homeost<strong>as</strong>is. Science 324, 392-397 (2009).<br />

6. Shortman K, Liu YJ. Mouse and human dendritic cell subtypes. Nat Rev Immunol.<br />

2002, 151-161 (2002).<br />

7. Dudziak D, Kamphorst AO, Heidkamp GF, Buchholz VR, Trumpfheller C,<br />

Yamazaki S, Cheong C, Liu K, Lee HW, Park CG, Steinman RM, Nussenzweig MC.<br />

Differential antigen processing by dendritic cell subsets in vivo. Science 315, 107-11<br />

(2007).<br />

8. den Haan JM, Lehar SM, Bevan MJ. CD8(+) but not CD8(-) dendritic <strong>cells</strong> crossprime<br />

cytotoxic T <strong>cells</strong> in vivo. J Exp Med. 192, 1685-1696 (2000).<br />

9. Lin ML, Zhan Y, Proietto AI, Prato S, Wu L, Heath WR, Villadangos JA, Lew AM.<br />

Selective suicide of cross-presenting <strong>CD8+</strong> dendritic <strong>cells</strong> by cytochrome c injection<br />

shows functional heterogeneity within this subset. Proc Natl Acad Sci. U S A 105,<br />

3029-34 (2008).<br />

10. Randolph GJ, Ochando J, Partida-Sanchez S. Migration of dendritic cell subsets<br />

and their precursors. Annu Rev Immunol. 26, 293-316 (2008).<br />

11. O’Keeffe M, Hochrein H, Vremec D, et al. Mouse pl<strong>as</strong>macytoid <strong>cells</strong>: long-lived<br />

<strong>cells</strong>, heterogeneous in surface phenotype and function, that differentiate into CD8(+)<br />

dendritic <strong>cells</strong> only after microbial stimulus. J Exp Med. 196, 1307-1319 (2002).<br />

12. Asselin-Paturel C, Brizard G, Pin JJ, Briere F, Trinchieri G. Mouse strain<br />

differences in pl<strong>as</strong>macytoid dendritic cell frequency and function revealed by a novel<br />

monoclonal antibody. J Immunol. 171, 6466–6477 (2003).<br />

13. Krug A, Luker GD, Barchet W, Leib DA, Akira S, Colonna M. Herpes simplex<br />

virus type 1 activates murine natural interferon-producing <strong>cells</strong> through Toll-like<br />

receptor 9. Blood 103, 1433–1437 (2004).<br />

14. Bl<strong>as</strong>ius AL, Giurisato E, Cella M, Schreiber RD, Shaw AS, Colonna M. <strong>Bone</strong><br />

<strong>marrow</strong> stromal cell antigen 2 is a specific marker of type I IFN-producing <strong>cells</strong> in the<br />

naïve mouse, but a promiscuous cell surface antigen following IFN stimulation. J<br />

Immunol. 177, 3260–3265 (2006).<br />

15. Krug A, French AR, Barchet W, Fischer JA, Dzionek A, Pingel JT et al. TLR9-<br />

dependent recognition of MCMV by IPC and DC generates coordinated cytokine<br />

responses that activate antiviral NK cell function. Immunity 21, 107–119 (2004).<br />

16. Barchet W, Krug A, Cella M, Newby C, Fischer JA, Dzionek A et al. Dendritic<br />

<strong>cells</strong> respond to influenza virus through TLR7- and PKR-independent pathways. Eur<br />

J Immunol. 35, 236–242 (2005).<br />

17. Neil, S.J., Zang, T., and Bieni<strong>as</strong>z, P.D. Tetherin inhibits retrovirus rele<strong>as</strong>e and is<br />

antagonized by HIV-1 Vpu. Nature 451, 425–430 (2008).<br />

94


18. Bl<strong>as</strong>ius AL, Colonna M. Sampling and signaling in pl<strong>as</strong>macytoid dendritic <strong>cells</strong>:<br />

the potential roles of Siglec-H. Trends Immunol. 27, 255–260 (2006).<br />

19. Colonna M, Trinchieri G, Liu YJ. Pl<strong>as</strong>macytoid dendritic <strong>cells</strong> in immunity. Nat<br />

Immunol. 5, 1219-1226 (2004).<br />

20. Fitzgerald-Bocarsly, P., Dai, J., and Singh, S. Pl<strong>as</strong>macytoid dendritic<br />

<strong>cells</strong> and type I IFN: 50 years of convergent history. Cytokine Growth Factor<br />

Rev. 19, 3–19 (2008).<br />

21. Cisse B. et al. Transcription factor E2-2 is an essential and specific regulator of<br />

pl<strong>as</strong>macytoid dendritic cell development. Cell 135, 37-48 (2008).<br />

22. Asselin-Paturel, C., Boonstra, A., Dalod, M., Durand, I., Yessaad, N., Dezutter-<br />

Dambuyant, C., Vicari, A., O’Garra, A., Biron, C., Briere, F., and Trinchieri, G.<br />

Mouse type I IFN-producing <strong>cells</strong> are immature APCs with pl<strong>as</strong>macytoid<br />

morphology. Nat Immunol. 2, 1144–1150 (2001).<br />

23. Bjorck, P. Isolation and characterization of pl<strong>as</strong>macytoid dendritic <strong>cells</strong><br />

from Flt3 ligand and granulocyte-macrophage colony-stimulating factortreated<br />

mice. Blood 98, 3520–3526 (2001).<br />

24. Grouard, G., Rissoan, M.C., Filgueira, L., Durand, I., Banchereau, J., and Liu,<br />

Y.J. The enigmatic pl<strong>as</strong>macytoid T <strong>cells</strong> develop into dendritic <strong>cells</strong> with interleukin<br />

(IL)-3 and CD40-ligand. J Exp Med. 185, 1101–1111 (1997).<br />

25. Kadowaki, N., Ho, S., Antonenko, S., de Waal Malefyt, R., K<strong>as</strong>telein, R.A.,<br />

Bazan, F., and Liu, Y.J. Subsets of human dendritic cell precursors express<br />

different toll-like receptors and respond to different microbial antigens. J Exp Med.<br />

194, 863–870 (2001).<br />

26. Nakano, H., Yanagita, M., and Gunn, M.D. CD11c(+)B220(+)Gr-1(+) <strong>cells</strong> in<br />

mouse lymph nodes and spleen display characteristics of pl<strong>as</strong>macytoid dendritic <strong>cells</strong>.<br />

J Exp Med. 194, 1171–1178 (2001).<br />

27. Reis e Sousa, C. Dendritic <strong>cells</strong> in a mature age. Nat Rev Immunol. 6, 476–483<br />

(2006).<br />

28. Salio, M., Palmowski, M.J., Atzberger, A., Hermans, I.F., and Cerundolo, V.<br />

CpG-matured murine pl<strong>as</strong>macytoid dendritic <strong>cells</strong> are capable of in vivo <strong>priming</strong> of<br />

functional CD8 T cell responses to endogenous but not exogenous antigens. J Exp<br />

Med. 199, 567–579 (2004).<br />

29. Schlecht, G., Garcia, S., Escriou, N., Freit<strong>as</strong>, A.A., Leclerc, C., and Dadaglio,<br />

G. Murine pl<strong>as</strong>macytoid dendritic <strong>cells</strong> induce effector/memory <strong>CD8+</strong> T-cell<br />

responses in vivo after viral stimulation. Blood 104, 1808–1815 (2004).<br />

30. McGill, J., Van Rooijen, N., and Legge, K.L. Protective influenz<strong>as</strong>pecific<br />

CD8 T cell responses require interactions with dendritic <strong>cells</strong> in the lungs. J Exp Med.<br />

205, 1635–1646 (2008).<br />

31. Gilliet, M., and Liu, Y.J. Generation of human CD8 T regulatory <strong>cells</strong> by CD40<br />

ligand-activated pl<strong>as</strong>macytoid dendritic <strong>cells</strong>. J Exp Med. 195, 695–704 (2002).<br />

32. Ito, T., Yang, M., Wang, Y.H., Lande, R., Gregorio, J., Perng, O.A., Qin, X.F.,<br />

Liu, Y.J., and Gilliet, M. Pl<strong>as</strong>macytoid dendritic <strong>cells</strong> prime IL-10-producing<br />

T regulatory <strong>cells</strong> by inducible costimulator ligand. J Exp Med 204, 105–115 (2007).<br />

33. Martin, P., Del Hoyo, G.M., Anjuere, F., Ari<strong>as</strong>, C.F., Varg<strong>as</strong>, H.H., Fernandez,<br />

L.A., Parrill<strong>as</strong>, V., and Ardavin, C. Characterization of a new subpopulation<br />

of mouse CD8alpha+ B220+ dendritic <strong>cells</strong> endowed with type 1 interferon<br />

production capacity and tolerogenic potential. Blood 100, 383–390 (2002).<br />

34. Ochando, J.C., Homma, C., Yang, Y., Hidalgo, A., Garin, A., Tacke, F., Angeli,<br />

V., Li, Y., Boros, P., Ding, Y., et al. Alloantigen-presenting pl<strong>as</strong>macytoid<br />

95


dendritic <strong>cells</strong> mediate tolerance to v<strong>as</strong>cularized grafts. Nat Immunol. 7, 652–662<br />

(2006).<br />

35. Sharma, M.D., Baban, B., Chandler, P., Hou, D.Y., Singh, N., Yagita, H.,<br />

Azuma, M., Blazar, B.R., Mellor, A.L., and Munn, D.H. Pl<strong>as</strong>macytoid dendritic <strong>cells</strong><br />

from mouse tumor-draining lymph nodes directly activate mature Tregs via<br />

indoleamine 2,3-dioxygen<strong>as</strong>e. J Clin Invest. 117, 2570–2582 (2007).<br />

36. De Heer, H.J., Hammad, H., Soullie, T., Hijdra, D., Vos, N., Willart, M.A.,<br />

Hoogsteden, H.C., and Lambrecht, B.N. Essential role of lung pl<strong>as</strong>macytoid dendritic<br />

<strong>cells</strong> in preventing <strong>as</strong>thmatic reactions to harmless inhaled antigen. J Exp Med. 200,<br />

89–98 (2004).<br />

37. Smit, J.J., Rudd, B.D., and Lukacs, N.W. Pl<strong>as</strong>macytoid dendritic <strong>cells</strong> inhibit<br />

pulmonary immunopathology and promote clearance of respiratory syncytial virus. J<br />

Exp Med. 203, 1153–1159 (2006).<br />

38. Wang, H., Peters, N., and Schwarze, J. Pl<strong>as</strong>macytoid dendritic <strong>cells</strong> limit viral<br />

replication, pulmonary inflammation, and airway hyperresponsiveness in respiratory<br />

syncytial virus infection. J Immunol. 177, 6263–6270 (2006).<br />

39. Wilson NS, Villadangos JA. Regulation of antigen presentation and crosspresentation<br />

in the dendritic cell network: facts, hypothesis, and immunological<br />

implications. Adv Immunol. 86, 241-305 (2005).<br />

40. Strawbridge AB, Blum JS. Autophagy in MHC cl<strong>as</strong>s II antigen processing. Curr<br />

Opin Immunol. 19, 87-92. (2007).<br />

41. Rock KL, Shen L. Cross-presentation: underlying mechanisms and role in<br />

immune surveillance. Immunol Rev. 207, 166-83 (2005).<br />

42. Jung, S. et al. In vivo depletion of CD11c(+) dendritic <strong>cells</strong> abrogates <strong>priming</strong> of<br />

CD8(+) T <strong>cells</strong> by exogenous cell-<strong>as</strong>sociated antigens. Immunity 17, 211-20 (2002).<br />

43. Melief CJ. Regulation of cytotoxic T lymphocyte responses by dendritic <strong>cells</strong>:<br />

peaceful coexistence of cross-<strong>priming</strong> and direct <strong>priming</strong>? Eur J Immunol. 33, 2645-<br />

54 (2003).<br />

44. Zinkernagel RM. On cross-<strong>priming</strong> of MHC cl<strong>as</strong>s I-specific CTL: rule or<br />

exception? Eur J Immunol. 32, 2385-92 (2002).<br />

45. Fogg DK, Sibon C, Miled C, et al. A clonogenic bone <strong>marrow</strong> progenitor specific<br />

<strong>for</strong> macrophages and dendritic <strong>cells</strong>. Science 311, 83-87 (2006).<br />

46. van Furth R, Raeburn JA, van Zwet TL. Characteristics of human mononuclear<br />

phagocytes. Blood 54, 485–500 (1979).<br />

47. Gordon S, Taylor PR. Monocyte and macrophage heterogeneity. Nat Rev<br />

Immunol. 5, 953–964 (2005).<br />

48. Palframan RT, Jung S, Cheng G, Weninger W, Luo Y, Dorf M et al. Inflammatory<br />

chemokine transport and presentation in HEV: a remote control mechanism <strong>for</strong><br />

monocyte recruitment to lymph nodes in inflamed tissues. J Exp Med. 194, 1361–<br />

1373 (2001).<br />

49. Geissmann F, Jung S, Littman DR. Blood monocytes consist of two principal<br />

subsets with distinct migratory properties. Immunity 19, 71–82 (2003).<br />

50. Varol C, Landsman L, Fogg DK, Greenshtein L, Gildor B, Margalit R et al.<br />

Monocytes give rise to mucosal, but not splenic, conventional dendritic <strong>cells</strong>. J Exp<br />

Med. 204, 171–180 (2007).<br />

51. Colmone A, Sipkins DA. Beyond angiogenesis: the role of endothelium in the<br />

bone <strong>marrow</strong> v<strong>as</strong>cular niche. Transl Res. 151, 1-9 (2008).<br />

52. Mazo IB., von Andrian UH. Adhesion and homing of blood-borne <strong>cells</strong> in bone <strong>marrow</strong><br />

microvessels. J Leukoc Biol. 66, 25-32 (1999).<br />

96


53. Nag<strong>as</strong>awa, T. et al. Microenvironmental niches in the bone <strong>marrow</strong> required <strong>for</strong><br />

B-cell development. Nat Rev Immunol 6, 107–116 (2006).<br />

54. Cavanagh, L. et al. Activation of bone <strong>marrow</strong>-resident memory T <strong>cells</strong> by<br />

circulating, antigen-bearing dendritic <strong>cells</strong>. Nat Immunol. 6, 1029-1037 (2005).<br />

55. Cariappa, A. et al. Perisinusoidal B <strong>cells</strong> in the bone <strong>marrow</strong> participate in T-<br />

independent responses to blood-borne microbes. Immunity 23, 397-407 (2005).<br />

56. Cancro, M. et al. Peripheral B-cell maturation: the intersection of selection and<br />

homeost<strong>as</strong>is. Immunol Rev. 197, 89-101 (2004).<br />

57. Sriv<strong>as</strong>tava, B. et al. Models <strong>for</strong> peripheral B cell development and homeost<strong>as</strong>is.<br />

Sem. Immunol. 17, 175-182 (2005).<br />

58. Manz, R. et al. Lifetime of pl<strong>as</strong>ma <strong>cells</strong> in the bone <strong>marrow</strong>. Nature 388, 133-134<br />

(1997).<br />

59. Tokoyoda K, Egawa T, Sugiyama T, Choi BI, Nag<strong>as</strong>awa T. Cellular niches<br />

controlling B lymphocyte behavior within bone <strong>marrow</strong> during development.<br />

Immunity 20, 707-18 (2004).<br />

60. Pabst R, Binns RM, Westermann J. What is the function of peripheral<br />

lymphocytes migrating to the thymus and of B lymphocytes proliferating in the<br />

thymus? Thymus 13, 149–156 (1989).<br />

61. Di Rosa F, Pabst R. The bone <strong>marrow</strong>: a nest <strong>for</strong> migratory memory T <strong>cells</strong>.<br />

Trends Immunol. 26, 360–366 (2005).<br />

62. Becker, TC., Coley, SM., Wherry, EJ., Ahmed, R. <strong>Bone</strong> <strong>marrow</strong> is a preferred <strong>site</strong><br />

<strong>for</strong> homeostatic proliferation of memory CD8 T <strong>cells</strong>. J Immunol. 174, 1269-1273<br />

(2005).<br />

63. Mazo IB, Honczarenko M, Leung H, Cavanagh LL, Bon<strong>as</strong>io R, Weninger W et al.<br />

<strong>Bone</strong> <strong>marrow</strong> is a major reservoir and <strong>site</strong> of recruitment <strong>for</strong> central memory <strong>CD8+</strong> T<br />

<strong>cells</strong>. Immunity 22, 259–270 (2005).<br />

64. Sipkins DA, Wei X, Wu JW, Runnels JM, Cote D, Means TK et al. In vivo<br />

imaging of specialized bone <strong>marrow</strong> endothelial microdomains <strong>for</strong> tumour<br />

engraftment. Nature 435, 969–973 (2005).<br />

65. Sumen C, Mempel TR, Mazo IB, von Andrian UH. Intravital microscopy:<br />

visualizing immunity in context. Immunity 21, 315-329 (2004).<br />

66. Bousso P. T-cell activation by dendritic <strong>cells</strong> in the lymph node: lessons from the<br />

movies. Nat Rev Immunol. 8, 1-10 (2008).<br />

67. Mazo I., Gutierrez-Ramos JC., Frenette PS., Hynes RO., Wagner DD., von<br />

Andrian UH. Hematopoietic progenitor cell rolling in bone <strong>marrow</strong> microvessels:<br />

parallel contributions by endothelial selectins and VCAM-1. J Exp Med. 188, 465-473<br />

(1998).<br />

68. Calvi, L.M. et al. Osteobl<strong>as</strong>tic <strong>cells</strong> regulate the haematopoietic stem cell niche.<br />

Nature 425, 841-6 (2003).<br />

69. Zhang, J. et al. Identification of the haematopoietic stem cell niche and control of<br />

the niche size. Nature 425, 836-41 (2003).<br />

70. Avecilla, S.T. et al. Chemokine-mediated interaction of hematopoietic progenitors<br />

with the bone <strong>marrow</strong> v<strong>as</strong>cular niche is required <strong>for</strong> thrombopoiesis. Nat Med. 10,<br />

64-71 (2004).<br />

71. Hardy, R.R., Carmack, C.E., Shinton, S.A., Kemp, J.D. & Hayakawa, K.<br />

Resolution and characterization of pro-B and pre-pro-B cell stages in normal mouse<br />

bone <strong>marrow</strong>. J Exp Med. 173, 1213-25 (1991).<br />

72. Loder, F. et al. B cell development in the spleen takes place in discrete steps and<br />

is determined by the quality of B cell receptor-derived signals. J Exp Med. 190, 75-89<br />

(1999).<br />

97


73. Moser, K., Tokoyoda, K., Radbruch, A., MacLennan, I. & Manz, R.A. Stromal<br />

niches, pl<strong>as</strong>ma cell differentiation and survival. Curr Opin Immunol. 18, 265-70<br />

(2006).<br />

74. M<strong>as</strong>opust, D., Vezys, V., Marzo, A.L. & Lefrancois, L. Preferential localization of<br />

effector memory <strong>cells</strong> in nonlymphoid tissue. Science 291, 2413-7 (2001).<br />

75. Mazo, I.B. et al. <strong>Bone</strong> <strong>marrow</strong> is a major reservoir and <strong>site</strong> of recruitment <strong>for</strong><br />

central memory <strong>CD8+</strong> T <strong>cells</strong>. Immunity 22, 259-70 (2005).<br />

76. Feuerer, M. et al. <strong>Bone</strong> <strong>marrow</strong> <strong>as</strong> a <strong>priming</strong> <strong>site</strong> <strong>for</strong> T-cell responses to blood<br />

borne antigen. Nat Med. 9, 1151-7 (2003).<br />

77. Feuerer, M. et al. <strong>Bone</strong> <strong>marrow</strong> microenvironment facilitating dendritic cell: CD4<br />

T cell interactions and maintenance of CD4 memory. Int J Oncol. 25, 867-76 (2004).<br />

78. Shortman, K. & Naik, S.H. Steady-state and inflammatory dendritic-cell<br />

development. Nat Rev Immunol. 7, 19-30 (2007).<br />

79. Shortman, K. & Liu, Y.J. Mouse and human dendritic cell subtypes. Nat Rev<br />

Immunol. 2, 151-61 (2002).<br />

80. Steinman, R.M. & Witmer, M.D. Lymphoid dendritic <strong>cells</strong> are potent stimulators<br />

of the primary mixed leukocyte reaction in mice. Proc Natl Acad Sci. U S A 75, 5132-<br />

6 (1978).<br />

81. Jung, S. et al. Analysis of fractalkine receptor CX(3)CR1 function by targeted<br />

deletion and green fluorescent protein reporter gene insertion. Mol Cell Biol. 20,<br />

4106-14 (2000).<br />

82. Niess, J.H. et al. CX3CR1-mediated dendritic cell access to the intestinal lumen<br />

and bacterial clearance. Science 307, 254-8 (2005).<br />

83. Soos, T.J. et al. CX3CR1+ interstitial dendritic <strong>cells</strong> <strong>for</strong>m a contiguous network<br />

throughout the entire kidney. Kidney Int. 70, 591-6 (2006).<br />

84. Landsman, L., Varol, C. & Jung, S. Distinct differentiation potential of blood<br />

monocyte subsets in the lung. J Immunol. 178, 2000-7 (2007).<br />

85. Zaft, T., Sapoznikov, A., Krauthgamer, R., Littman, D.R. & Jung, S. CD11chigh<br />

dendritic cell ablation impairs lymphopenia-driven proliferation of naive and memory<br />

<strong>CD8+</strong> T <strong>cells</strong>. J Immunol. 175, 6428-35 (2005).<br />

86. Allman, D. et al. Resolution of three nonproliferative immature splenic B cell<br />

subsets reveals multiple selection points during peripheral B cell maturation. J<br />

Immunol. 167, 6834-40 (2001).<br />

87. Hebel, K. et al. Pl<strong>as</strong>ma cell differentiation in T-independent type 2 immune<br />

responses is independent of CD11c(high) dendritic <strong>cells</strong>. Eur J Immunol. 36, 2912-9<br />

(2006).<br />

88. Str<strong>as</strong>ser, A. et al. En<strong>for</strong>ced BCL2 expression in B-lymphoid <strong>cells</strong> prolongs<br />

antibody responses and elicits autoimmune dise<strong>as</strong>e. Proc Natl Acad Sci. U S A 88,<br />

8661-5 (1991).<br />

89. Lesley, R., Kelly, L.M., Xu, Y. & Cyster, J.G. Naive CD4 T <strong>cells</strong> constitutively<br />

express CD40L and augment autoreactive B cell survival. Proc Natl Acad Sci. U S A<br />

103, 10717-22 (2006).<br />

90. Mackay, F. & Browning, J.L. BAFF: a fundamental survival factor <strong>for</strong> B <strong>cells</strong>.<br />

Nat Rev Immunol. 2, 465-75 (2002).<br />

91. Gorelik, L. et al. Normal B cell homeost<strong>as</strong>is requires B cell activation factor<br />

production by radiation-resistant <strong>cells</strong>. J Exp Med. 198, 937-45 (2003).<br />

92. Nardelli, B. et al. Synthesis and rele<strong>as</strong>e of B-lymphocyte stimulator from myeloid<br />

<strong>cells</strong>. Blood 97, 198-204 (2001).<br />

93. Litinskiy, M.B. et al. DCs induce CD40-independent immunoglobulin cl<strong>as</strong>s<br />

switching through BLyS and APRIL. Nat Immunol. 3, 822-9 (2002).<br />

98


94. Schiemann, B. et al. An essential role <strong>for</strong> BAFF in the normal development of B<br />

<strong>cells</strong> through a BCMA-independent pathway. Science 293, 2111-4 (2001).<br />

95. Leng, L. et al. MIF signal transduction initiated by binding to CD74. J Exp Med.<br />

197, 1467-76 (2003).<br />

96. Lue, H. et al. Macrophage migration inhibitory factor (MIF) promotes cell<br />

survival by activation of the Akt pathway and role <strong>for</strong> CSN5/JAB1 in the control of<br />

autocrine MIF activity. Oncogene 26, 5046-5059 (2007).<br />

97. Starlets, D. et al. Cell-surface CD74 initiates a signaling c<strong>as</strong>cade leading to cell<br />

proliferation and survival. Blood 107, 4807-16 (2006).<br />

98. Lantner, F. et al. CD74 induces TAp63 expression leading to B cell survival.<br />

Blood 110, 4303-4311 (2007).<br />

99. Calandra, T., Bernhagen, J., Mitchell, R.A. & Bucala, R. The macrophage is an<br />

important and previously unrecognized source of macrophage migration inhibitory<br />

factor. J Exp Med. 179, 1895-902 (1994).<br />

100. Qi, H., Egen, J.G., Huang, A.Y. & Germain, R.N. Extrafollicular activation of<br />

lymph node B <strong>cells</strong> by antigen-bearing dendritic <strong>cells</strong>. Science 312, 1672-6 (2006).<br />

101. Sipkins, D.A. et al. In vivo imaging of specialized bone <strong>marrow</strong> endothelial<br />

microdomains <strong>for</strong> tumour engraftment. Nature 435, 969-73 (2005).<br />

102. Bajenoff, M. et al. Stromal cell networks regulate lymphocyte entry, migration,<br />

and territoriality in lymph nodes. Immunity 25, 989-1001 (2006).<br />

103. Morand, E.F., Leech, M. & Bernhagen, J. MIF: a new cytokine link between<br />

rheumatoid arthritis and atherosclerosis. Nat Rev Drug Discov. 5, 399-410 (2006).<br />

104. Balazs, M., Martin, F., Zhou, T. & Kearney, J. Blood dendritic <strong>cells</strong> interact with<br />

splenic marginal zone B <strong>cells</strong> to initiate T-independent immune responses.<br />

Immunity 17, 341-52 (2002).<br />

105. Cariappa, A., Ch<strong>as</strong>e, C., Liu, H., Russell, P. & Pillai, S. Naive recirculating B<br />

<strong>cells</strong> mature simultaneously in the spleen and bone <strong>marrow</strong>. Blood 109, 2339-45<br />

(2007).<br />

106. Nishizumi, H. et al. Impaired proliferation of peripheral B <strong>cells</strong> and indication of<br />

autoimmune dise<strong>as</strong>e in lyn-deficient mice. Immunity 3, 549-60 (1995).<br />

107. Otipoby, K.L. et al. CD22 regulates thymus-independent responses and the<br />

lifespan of B <strong>cells</strong>. Nature 384, 634-7 (1996).<br />

108. O'Keefe, T.L., Williams, G.T., Davies, S.L. & Neuberger, M.S. Hyperresponsive<br />

B <strong>cells</strong> in CD22-deficient mice. Science 274, 798-801 (1996).<br />

109. Pan, C., Baumgarth, N. & Parnes, J.R. CD72-deficient mice reveal nonredundant<br />

roles of CD72 in B cell development and activation. Immunity 11, 495-506 (1999).<br />

110. Cariappa, A. et al. The follicular versus marginal zone B lymphocyte cell fate<br />

decision is regulated by Aiolos, Btk, and CD21. Immunity 14, 603-15 (2001).<br />

111. Floyd, H., Nitschke, L. & Crocker, P.R. A novel subset of murine B <strong>cells</strong> that<br />

expresses unm<strong>as</strong>ked <strong>for</strong>ms of CD22 is enriched in the bone <strong>marrow</strong>: implications<br />

<strong>for</strong> B-cell homing to the bone <strong>marrow</strong>. Immunology 101, 342-7 (2000).<br />

112. Fu, Y.-X. & Chaplin, D.D. Development and maturation of secondary lymphoid<br />

tissues. Annu Rev Immunol. 17, 399-458 (1999).<br />

113. Lanzavecchia, A. & Sallusto, F. The instructive role of dendritic <strong>cells</strong> on T cell<br />

responses: lineages, pl<strong>as</strong>ticity and kinetics. Curr Opin Immunol. 13, 291–296 (2001).<br />

114. Osmond, D.G. Production and selection of B lymphocytes in bone <strong>marrow</strong>:<br />

lymphostromal interactions and apoptosis in normal, mutant and transgenic mice. Adv<br />

Exp Med Biol. 355, 15–20 (1994).<br />

115. Koni, P.A. et al. Conditional v<strong>as</strong>cular cell adhesion molecule 1 deletion in mice:<br />

impaired lymphocyte migration to bone <strong>marrow</strong>. J Exp Med. 193, 741–753 (2001).<br />

99


116. Di Rosa, F. & Santoni, A. <strong>Bone</strong> <strong>marrow</strong> CD8 T <strong>cells</strong> are in a different activation<br />

state than those in lymphoid periphery. Eur J Immunol. 32, 1873-1880 (2002).<br />

117. Becker, T.C., et al. <strong>Bone</strong> <strong>marrow</strong> is a preferred <strong>site</strong> <strong>for</strong> homeostatic proliferation<br />

of memory CD8 T <strong>cells</strong>. J Immunol. 174, 1269-1273 (2005).<br />

118. M<strong>as</strong>opust D, Vezys V, Marzo AL, Lefrançois L Preferential localization of<br />

effector memory <strong>cells</strong> in nonlymphoid tissue. Science 291, 2413-7 (2001).<br />

119. Martinez, C., et al. Immune reconstitution following allogeneic peripheral blood<br />

progenitor cell transplantation: comparison of recipients of positive CD34+ selected<br />

grafts with recipients of unmanipulated grafts. Exp Hematol. 27, 561-568 (1999).<br />

120. Adams, G.B., et al. Heterologous <strong>cells</strong> cooperate to augment stem cell migration,<br />

homing, and engraftment. Blood 101, 45-51 (2003).<br />

121. Naik, S. H., et al. Intr<strong>as</strong>plenic steady-state dendritic cell precursors that are<br />

distinct from monocytes. Nat Immunol. 7, 663-671 (2006).<br />

122. Miller, M.J., et al. Two-photon imaging of lymphocyte motility and antigen<br />

response in intact lymph node. Science 296, 1869-1873 (2002).<br />

123. Mempel, T.R., et al. T-cell <strong>priming</strong> by dendritic <strong>cells</strong> in lymph nodes occurs in<br />

three distinct ph<strong>as</strong>es. Nature 427, 154-159 (2004).<br />

124. Celluzzi CM, Mayordomo JI, Storkus WJ, Lotze MT, Falo LD, Jr. Peptidepulsed<br />

dendritic <strong>cells</strong> induce antigen-specific CTL-mediated protective tumor<br />

immunity. J Exp Med. 183, 283-287 (1996).<br />

125. Sancho, D., et al. CD69 is a an immunoregulatory molecule induced following<br />

activation. Trends Immunol. 26, 136-140 (2005).<br />

126. Reinhardt, R.L., A. Khoruts, R. Merica, T. Zell, and M.K. Jenkins. Visualizing<br />

the generation of memory CD4 T <strong>cells</strong> in the whole body. Nature 410, 101-105<br />

(2001).<br />

127. Kurts, C., M. Cannarile, I. Klebba, and T. Brocker. Dendritic <strong>cells</strong> are sufficient<br />

to cross-present self-antigens to CD8 T <strong>cells</strong> in vivo. J Immunol. 166, 1439-1442<br />

(2001).<br />

128. Limmer, A., Ohl, J., Wingender, G., and Knolle, PA. et al. Cross-presentation of<br />

oral antigens by liver sinusoidal endothelial <strong>cells</strong> leads to CD8 T cell tolerance. Eur J<br />

Immunol. 35, 2970-2981 (2005).<br />

129. Kiesel, JR., Buchwald, ZS., Aurora, R. Cross-presentation by osteocl<strong>as</strong>ts induces<br />

FoxP3 in <strong>CD8+</strong> T <strong>cells</strong>. J Immunol. 182, 5477-5487 (2009).<br />

130. P ´erarnau, B., Saron, M.-F., Reina San Martin, B., Berv<strong>as</strong>, N., Ong, H., Soloski,<br />

M., Smith, A. G., Ure, J. M., Gairin, J.-E. and Lemonnier, F. A., Single H-2K b , H-2D b<br />

and double H-2K b D b knock-out mice: peripheral <strong>CD8+</strong> T cell repertoire and antilymphocytic<br />

choriomeningitis virus cytolytic responses. Eur J Immunol. 29, 1243–<br />

1252 (1999).<br />

131. Hall, JG., & Morris, B. The immediate effect of antigens on the cell output of a<br />

lymph node. Br J Exp Pathol. 46, 450-454 (1965).<br />

132. Shiow, LR., Rosen, DB., Brdickova, N., Xu, Y., An, J., Lanier, LL., Cyster, JC.,<br />

Matloubian, M. CD69 acts downstream of interferon-α/β to inhibit S1P 1 and<br />

lymphocyte egress from lymphoid organs. Nature 440, 540-544 (2006).<br />

133. Tian, T., J. Woodworth, M. Skold, and S.M. Behar. In vivo depletion of CD11c+<br />

<strong>cells</strong> delays the CD4+ T cell response to Mycobacterium tuberculosis and exacerbates<br />

the outcome of infection. J Immunol. 175, 3268-3272 (2005).<br />

134. Gretz, JE., Kaldjian, EP., Anderson, AO., Shaw, S. Sophisticated strategies <strong>for</strong><br />

in<strong>for</strong>mation encounter in the lymph node: the reticular network <strong>as</strong> a conduit of soluble<br />

in<strong>for</strong>mation and a highway <strong>for</strong> cell traffic. J Immunol. 157, 495-499 (1996).<br />

100


135. Bajenoff, M., Egen, JG., Koo, LY., Laugier, JP., Brau, F., Glaichenhaus, N.,<br />

Germain, RN. Stromal cell networks regulate lymphocyte entry, migration, and<br />

territoriality in lymph nodes. Immunity 25, 989-1001 (2006).<br />

136. Spiotto, M., Rowley, DA., Schreiber, H. Bystander elimination of antigen loss<br />

variants in established tumors. Nat Med. 10, 294-298 (2004).<br />

137. Kawai, T., S. Sato, K.J. Ishii, C. Coban, H. Hemmi, M. Yamamoto, K. Terai, M.<br />

Matsuda, J. Inoue, S. Uematsu, O. Takeuchi, and S. Akira. Interferon-alpha induction<br />

through Toll-like receptors involves a direct interaction of IRF7 with MyD88 and<br />

TRAF6. Nat Immunol. 5, 1061-1068 (2004).<br />

138. Lennert, K., and W. Remmele. [Karyometric research on lymph node <strong>cells</strong> in<br />

man. I. Germinobl<strong>as</strong>ts, lymphobl<strong>as</strong>ts & lymphocytes.]. Acta Haematol. 19, 99-113<br />

(1958).<br />

139. Trinchieri, G., and D. Santoli. Anti-viral activity induced by culturing<br />

lymphocytes with tumor-derived or virus-trans<strong>for</strong>med <strong>cells</strong>. Enhancement of human<br />

natural killer cell activity by interferon and antagonistic inhibition of susceptibility of<br />

target <strong>cells</strong> to lysis. J Exp Med. 147, 1314-1333 (1978).<br />

140. Bandyopadhyay, S., B. Perussia, G. Trinchieri, D.S. Miller, and S.E. Starr.<br />

Requirement <strong>for</strong> HLA-DR+ accessory <strong>cells</strong> in natural killing of cytomegalovirusinfected<br />

fibrobl<strong>as</strong>ts. J Exp Med. 164, 180-195 (1986).<br />

141. Yoneyama, H., K. Matsuno, E. Toda, T. Nishiwaki, N. Matsuo, A. Nakano, S.<br />

Narumi, B. Lu, C. Gerard, S. Ishikawa, and K. Matsushima. Pl<strong>as</strong>macytoid DCs help<br />

lymph node DCs to induce anti-HSV CTLs. J Exp Med. 202, 425-435 (2005).<br />

142. Le Bon, A., N. Etchart, C. Rossmann, M. Ashton, S. Hou, D. Gewert, P. Borrow,<br />

and D.F. Tough. Cross-<strong>priming</strong> of <strong>CD8+</strong> T <strong>cells</strong> stimulated by virus-induced type I<br />

interferon. Nat Immunol. 4, 1009-1015 (2003).<br />

143. Shen, H., and A. Iw<strong>as</strong>aki. A crucial role <strong>for</strong> pl<strong>as</strong>macytoid dendritic <strong>cells</strong> in<br />

antiviral protection by CpG ODN-b<strong>as</strong>ed vaginal microbicide. J Clin Invest. 116,<br />

2237-2243 (2006).<br />

144. Kuwajima, S., T. Sato, K. Ishida, H. Tada, H. Tezuka, and T. Ohteki. Interleukin<br />

15-dependent crosstalk between conventional and pl<strong>as</strong>macytoid dendritic <strong>cells</strong> is<br />

essential <strong>for</strong> CpG-induced immune activation. Nat Immunol. 7, 740-746 (2006).<br />

145. Probst, H.C., and M. van den Broek. Priming of CTLs by lymphocytic<br />

choriomeningitis virus depends on dendritic <strong>cells</strong>. J Immunol. 174, 3920-3924 (2005).<br />

146. Kadowaki, N., and Y.J. Liu. Natural type I interferon-producing <strong>cells</strong> <strong>as</strong> a link<br />

between innate and adaptive immunity. Hum Immunol. 63, 1126-1132 (2002).<br />

147. Krug, A., R. Veer<strong>as</strong>wamy, A. Pekosz, O. Kanagawa, E.R. Unanue, M. Colonna,<br />

and M. Cella. Interferon-producing <strong>cells</strong> fail to induce proliferation of naive T <strong>cells</strong><br />

but can promote expansion and T helper 1 differentiation of antigen-experienced<br />

unpolarized T <strong>cells</strong>. J Exp Med. 197, 899-906 (2003).<br />

148. Boonstra, A., C. Asselin-Paturel, M. Gilliet, C. Crain, G. Trinchieri, Y.J. Liu,<br />

and A. O'Garra. Flexibility of mouse cl<strong>as</strong>sical and pl<strong>as</strong>macytoid-derived dendritic<br />

<strong>cells</strong> in directing T helper type 1 and 2 cell development: dependency on antigen dose<br />

and differential toll-like receptor ligation. J Exp Med. 197, 101-109 (2003).<br />

149. Salio, M., M.J. Palmowski, A. Atzberger, I.F. Hermans, and V. Cerundolo. CpGmatured<br />

murine pl<strong>as</strong>macytoid dendritic <strong>cells</strong> are capable of in vivo <strong>priming</strong> of<br />

functional CD8 T cell responses to endogenous but not exogenous antigens. J Exp<br />

Med. 199, 567-579 (2004).<br />

150. Smit, J.J., B.D. Rudd, and N.W. Lukacs. Pl<strong>as</strong>macytoid dendritic <strong>cells</strong> inhibit<br />

pulmonary immunopathology and promote clearance of respiratory syncytial virus. J<br />

Exp Med. 203, 1153-1159 (2006).<br />

101


151. Abe, M., B.L. Colvin, and A.W. Thomson. Pl<strong>as</strong>macytoid dendritic <strong>cells</strong>: in vivo<br />

regulators of alloimmune reactivity? Transplant Proc. 37, 4119-4121 (2005).<br />

152. Ochando, J.C., C. Homma, Y. Yang, A. Hidalgo, A. Garin, F. Tacke, V. Angeli,<br />

Y. Li, P. Boros, Y. Ding, R. Jessberger, G. Trinchieri, S.A. Lira, G.J. Randolph, and<br />

J.S. Bromberg. Alloantigen-presenting pl<strong>as</strong>macytoid dendritic <strong>cells</strong> mediate tolerance<br />

to v<strong>as</strong>cularized grafts. Nat Immunol. 7, 652-662 (2006).<br />

153. Brocker, T., M. Riedinger, and K. Karjalainen. Targeted expression of major<br />

histocompatibility complex (MHC) cl<strong>as</strong>s II molecules demonstrates that dendritic<br />

<strong>cells</strong> can induce negative but not positive selection of thymocytes in vivo. J Exp Med.<br />

185, 541-550 (1997).<br />

154. Probst, H.C., K. Tschannen, B. Odermatt, R. Schwendener, R.M. Zinkernagel,<br />

and M. Van Den Broek. Histological analysis of CD11c-DTR/GFP mice after in vivo<br />

depletion of dendritic <strong>cells</strong>. Clin Exp Immunol. 141, 398-404 (2005).<br />

155. van Rijt, L.S., S. Jung, A. Kleinjan, N. Vos, M. Willart, C. Duez, H.C.<br />

Hoogsteden, and B.N. Lambrecht. In vivo depletion of lung CD11c+ dendritic <strong>cells</strong><br />

during allergen challenge abrogates the characteristic features of <strong>as</strong>thma. J Exp Med.<br />

201, 981-991 (2005).<br />

156. Vallon-Eberhard, A., L. Landsman, N. Yogev, B. Verrier, and S. Jung.<br />

Transepithelial pathogen uptake into the small intestinal lamina propria. J Immunol.<br />

176, 2465-2469 (2006).<br />

157. Huleatt, J.W., and L. Lefrancois. Antigen-driven induction of CD11c on<br />

intestinal intraepithelial lymphocytes and <strong>CD8+</strong> T <strong>cells</strong> in vivo. J Immunol. 154,<br />

5684-5693 (1995).<br />

158. Laouar, Y., F.S. Sutterwala, L. Gorelik, and R.A. Flavell. Trans<strong>for</strong>ming growth<br />

factor-beta controls T helper type 1 cell development through regulation of natural<br />

killer cell interferon-gamma. Nat Immunol. 6, 600-607 (2005).<br />

159. Bl<strong>as</strong>ius, A., W. Vermi, A. Krug, F. Facchetti, M. Cella, and M. Colonna. A <strong>cells</strong>urface<br />

molecule selectively expressed on murine natural interferon-producing <strong>cells</strong><br />

that blocks secretion of interferon-alpha. Blood 103, 4201-4206 (2004).<br />

161. Krug, A., A.R. French, W. Barchet, J.A. Fischer, A. Dzionek, J.T. Pingel, M.M.<br />

Orihuela, S. Akira, W.M. Yokoyama, and M. Colonna. TLR9-dependent recognition<br />

of MCMV by IPC and DC generates coordinated cytokine responses that activate<br />

antiviral NK cell function. Immunity 21:107-119 (2004).<br />

161. Clarke, S.R., M. Barnden, C. Kurts, F.R. Carbone, J.F. Miller, and W.R. Heath.<br />

Characterization of the ovalbumin-specific TCR transgenic line OT-I: MHC elements<br />

<strong>for</strong> positive and negative selection. Immunol Cell Biol. 78, 110-117 (2000).<br />

162. Robertson, J.M., P.E. Jensen, and B.D. Evavold. DO11.10 and OT-II T <strong>cells</strong><br />

recognize a C-terminal ovalbumin 323-339 epitope. J Immunol. 164, 4706-4712<br />

(2000).<br />

163. Parish, C.R. Fluorescent dyes <strong>for</strong> lymphocyte migration and proliferation studies.<br />

Immunol Cell Biol. 77, 499-508 (1999).<br />

164. Zhang, J., A. Raper, N. Sugita, R. Hingorani, M. Salio, M.J. Palmowski, V.<br />

Cerundolo, and P.R. Crocker. Characterization of Siglec-H <strong>as</strong> a novel endocytic<br />

receptor expressed on murine pl<strong>as</strong>macytoid dendritic cell precursors. Blood 107,<br />

3600-3608 (2006).<br />

165. Foulds, K.E., L.A. Zenewicz, D.J. Shedlock, J. Jiang, A.E. Troy, and H. Shen.<br />

Cutting edge: CD4 and CD8 T <strong>cells</strong> are intrinsically different in their proliferative<br />

responses. J Immunol. 168,1528-1532 (2002).<br />

102


166. Hogquist, K.A., S.C. Jameson, W.R. Heath, J.L. Howard, M.J. Bevan, and F.R.<br />

Carbone. T cell receptor antagonist peptides induce positive selection. Cell 76, 17-27<br />

(1994).<br />

167. Zhong, G., C. Reis e Sousa, and R.N. Germain. Antigen-unspecific B <strong>cells</strong> and<br />

lymphoid dendritic <strong>cells</strong> both show extensive surface expression of processed antigenmajor<br />

histocompatibility complex cl<strong>as</strong>s II complexes after soluble protein exposure in<br />

vivo or in vitro. J Exp Med. 186, 673-682 (1997).<br />

168. Williams, G.S., A. Oxenius, H. Hengartner, C. Benoist, and D. Mathis. CD4+ T<br />

cell responses in mice lacking MHC cl<strong>as</strong>s II molecules specifically on B <strong>cells</strong>. Eur J<br />

Immunol. 28, 3763-3772 (1998).<br />

169. Reis e Sousa, C., and R.N. Germain. Analysis of adjuvant function by direct<br />

visualization of antigen presentation in vivo: endotoxin promotes accumulation of<br />

antigen-bearing dendritic <strong>cells</strong> in the T cell are<strong>as</strong> of lymphoid tissue. J Immunol. 162,<br />

6552-6561 (1999).<br />

170. Attanavanich, K., and J.F. Kearney. 2004. Marginal zone, but not follicular B<br />

<strong>cells</strong>, are potent activators of naive CD4 T <strong>cells</strong>. J Immunol. 172, 803-811 (2004).<br />

171. Itano, A.A., S.J. McSorley, R.L. Reinhardt, B.D. Ehst, E. Ingulli, A.Y.<br />

Rudensky, and M.K. Jenkins. Distinct dendritic cell populations sequentially present<br />

antigen to CD4 T <strong>cells</strong> and stimulate different <strong>as</strong>pects of cell-mediated immunity.<br />

Immunity 19, 47-57 (2003).<br />

172. Kitamura, D., and K. Rajewsky. Targeted disruption of mu chain membrane<br />

exon causes loss of heavy-chain allelic exclusion. Nature 356, 154-156 (1992).<br />

173. Bennett, C.L., E. van Rijn, S. Jung, K. Inaba, R.M. Steinman, M.L. Kapsenberg,<br />

and B.E. Clausen. Inducible ablation of mouse Langerhans <strong>cells</strong> diminishes but fails<br />

to abrogate contact hypersensitivity. J Cell Biol. 169, 569-576(2005).<br />

174. Sixt, M., N. Kanazawa, M. Selg, T. Samson, G. Roos, D.P. Reinhardt, R. Pabst,<br />

M.B. Lutz, and L. Sorokin. The conduit system transports soluble antigens from the<br />

afferent lymph to resident dendritic <strong>cells</strong> in the T cell area of the lymph node.<br />

Immunity 22, 19-29 (2005).<br />

175. Benitez-Rib<strong>as</strong>, D., G.J. Adema, G. Winkels, I.S. Kl<strong>as</strong>en, C.J. Punt, C.G. Figdor,<br />

and I.J. de Vries. Pl<strong>as</strong>macytoid dendritic <strong>cells</strong> of melanoma patients present<br />

exogenous proteins to CD4+ T <strong>cells</strong> after Fc gamma RII-mediated uptake. J Exp Med.<br />

203, 1629-1635 (2006).<br />

176. Hawiger, D., K. Inaba, Y. Dorsett, M. Guo, K. Mahnke, M. Rivera, J.V. Ravetch,<br />

R.M. Steinman, and M.C. Nussenzweig. Dendritic <strong>cells</strong> induce peripheral T cell<br />

unresponsiveness under steady state conditions in vivo. J Exp Med. 194, 769-779<br />

(2001).<br />

177. Itano, A.A., and M.K. Jenkins. Antigen presentation to naive CD4 T <strong>cells</strong> in the<br />

lymph node. Nat Immunol 4, 733-739 (2003).<br />

178. Sporri, R., and C. Reis e Sousa. Inflammatory mediators are insufficient <strong>for</strong> full<br />

dendritic cell activation and promote expansion of CD4+ T cell populations lacking<br />

helper function. Nat Immunol. 6, 163-170 (2005).<br />

179. Yang, C.P., S.M. Sparshott, D. Duffy, P. Garside, and E.B. Bell. 2006. The<br />

phenotype and survival of antigen-stimulated transgenic CD4 T <strong>cells</strong> in vivo: the<br />

influence of persisting antigen. Int Immunol. 18, 515-523 (2006).<br />

180. Mahnke, K., Y. Qian, J. Knop, and A.H. Enk. Induction of CD4+/CD25+<br />

regulatory T <strong>cells</strong> by targeting of antigens to immature dendritic <strong>cells</strong>. Blood 101,<br />

4862-4869 (2003).<br />

181. Swiggard, W.J., A. Mirza, M.C. Nussenzweig, and R.M. Steinman. DEC-205, a<br />

205-kDa protein abundant on mouse dendritic <strong>cells</strong> and thymic epithelium that is<br />

103


detected by the monoclonal antibody NLDC-145: purification, characterization, and<br />

N-terminal amino acid sequence. Cell Immunol. 165, 302-311 (1995).<br />

182. Asselin-Paturel, C., G. Brizard, K. Chemin, A. Boonstra, A. O'Garra, A. Vicari,<br />

and G. Trinchieri. Type I interferon dependence of pl<strong>as</strong>macytoid dendritic cell<br />

activation and migration. J Exp Med. 201, 1157-1167 (2005).<br />

183. Yoneyama, H., K. Matsuno, Y. Zhang, T. Nishiwaki, M. Kitabatake, S. Ueha, S.<br />

Narumi, S. Morikawa, T. Ezaki, B. Lu, C. Gerard, S. Ishikawa, and K. Matsushima.<br />

Evidence <strong>for</strong> recruitment of pl<strong>as</strong>macytoid dendritic cell precursors to inflamed lymph<br />

nodes through high endothelial venules. Int Immunol. 16, 915-928 (2004).<br />

184. Iw<strong>as</strong>aki, A., and R. Medzhitov. Toll-like receptor control of the adaptive immune<br />

responses. Nat Immunol. 5, 987-995 (2004).<br />

185. Jarrossay, D., G. Napolitani, M. Colonna, F. Sallusto, and A. Lanzavecchia.<br />

Specialization and complementarity in microbial molecule recognition by human<br />

myeloid and pl<strong>as</strong>macytoid dendritic <strong>cells</strong>. Eur J Immunol. 31, 3388-3393 (2001).<br />

186. Hermans, I.F., D.S. Ritchie, J. Yang, J.M. Roberts, and F. Ronchese. <strong>CD8+</strong> T<br />

cell-dependent elimination of dendritic <strong>cells</strong> in vivo limits the induction of antitumor<br />

immunity. J Immunol. 164, 3095-3101 (2000).<br />

187. Wong, P., and E.G. Pamer. Feedback regulation of pathogen-specific T cell<br />

<strong>priming</strong>. Immunity 18, 499-511 (2003).<br />

188. Janssen, E.M., E.E. Lemmens, T. Wolfe, U. Christen, M.G. von Herrath, and<br />

S.P. Schoenberger. CD4+ T <strong>cells</strong> are required <strong>for</strong> secondary expansion and memory<br />

in <strong>CD8+</strong> T lymphocytes. Nature 421, 852-856 (2003).<br />

189. Webster, B., E.H. Ekland, L.M. Agle, S. Chyou, R. Ruggieri, and T.T. Lu.<br />

Regulation of lymph node v<strong>as</strong>cular growth by dendritic <strong>cells</strong>. J Exp Med. 203, 1903-<br />

1913 (2006).<br />

190. Nag<strong>as</strong>awa, T. New niches <strong>for</strong> B <strong>cells</strong>. Nat Immunol. 9, 345-346 (2008).<br />

191. Bernhagen J, Krohn R, Lue H, Gregory JL, Zernecke A, Koenen RR, Dewor M,<br />

Georgiev I, Schober A, Leng L, Kooistra T, Fingerle-Rowson G, Ghezzi P, Kleemann<br />

R, McColl SR, Bucala R, Hickey MJ, Weber C. MIF is a noncognate ligand of CXC<br />

chemokine receptors in inflammatory and atherogenic cell recruitment. Nat Med. 13,<br />

587-96 (2007).<br />

192. Matsushita K, Hamabe M, Matsuoka M, et al. Experimental hematogenous<br />

osteomyelitis by Staphylococcus aureus. Clin Orthop Relat Res. 334, 291-297 (1997).<br />

193. Lenz LL, Butz EA, Bevan MJ. Requirements <strong>for</strong> bone <strong>marrow</strong>-derived antigenpresenting<br />

<strong>cells</strong> in <strong>priming</strong> cytotoxic T cell responses to intracellular pathogens. J Exp<br />

Med. 192, 1135-1142 (2000).<br />

194. Shinkai, Y. et al. Restoration of T cell development in RAG-2-deficient mice by<br />

functional TCR transgenes. Science 259, 822-5 (1993).<br />

195. Fingerle-Rowson, G. et al. The p53-dependent effects of macrophage migration<br />

inhibitory factor revealed by gene targeting. Proc Natl Acad Sci. U S A 100, 9354-9<br />

(2003).<br />

196. Mackay, F., Schneider, P., Rennert, P. And Browning, J. BAFF and APRIL: a<br />

tutorial on B cell survival. Annu Rev Immunol. 21, 231-264 (2003).<br />

197. Schiemann, B et al. An essential role of BAFF in the normal development of B<br />

<strong>cells</strong> through a BCMA-independent pathway. Science 293, 2111-2114 (2001).<br />

198. Gross, J. et al. TACI-Ig neutrolizes molecules critical <strong>for</strong> B cell development and<br />

autoimmune dise<strong>as</strong>e: impaired B cell maturation in mice lacking BLys. Immunity 15,<br />

289-302 (2001).<br />

199. Stumptner-Cuvelette P and Benaroch P. Multiple roles of the invariant chain in<br />

MHC cl<strong>as</strong>s II function. Biochim Biophys Acta. 1542, 1–13 (2002).<br />

104


200. Naujok<strong>as</strong> MF, Morin M, Anderson MS, Peterson M, Miller J. The chondroitin<br />

sulfate <strong>for</strong>m of invariant chain can enhance stimulation of T cell responses through<br />

interaction with CD44. Cell 74, 257–268 (1993).<br />

201. Gore Y, Starlets D, Maharshak N, Becker-Herman S, Kaneyuki U, Leng L,<br />

Bucala R, Shachar I. Macrophage migration inhibitory factor induces B cell survival<br />

by activation of a CD74-CD44 receptor complex. J Biol Chem. 283, 2784-92 (2008).<br />

105


8. List of publications<br />

8.1 Articles published in refereed journals (included in my thesis).<br />

1. Sapoznikov A., Fischer, J.A.A., Zaft T., Krauthgamer R., Dzionek A. and S. Jung.<br />

Organ-dependent In Vivo Priming of Naïve CD4 + , but not CD8 + T <strong>cells</strong> by<br />

Pl<strong>as</strong>macytoid Dendritic <strong>cells</strong>. (2007) J Exp Med. 204(8):1923-33.<br />

2. Sapoznikov A. *, Pewzner-Jung Y. *, Kalchenko V., Krauthgamer R., Shachar I.,<br />

Jung S. Periv<strong>as</strong>cular clusters of dendritic <strong>cells</strong> provide critical survival signals to B<br />

<strong>cells</strong> in bone <strong>marrow</strong> niches. (2008) Nat Immunol. 9(4):388-95.<br />

* equal contribution.<br />

8.2 Articles in refereed journals (not included in my thesis).<br />

1. Zaft, T.*, Sapoznikov, A.*, Krauthgamer, R., Littman, D.R. and S. Jung. CD11c high<br />

dendritic cell ablation impairs Lymphopenia - driven proliferation of naïve and<br />

memory CD8 + T <strong>cells</strong>. (2005) J Immunol. 175(10): 6428-35.<br />

* equal contribution.<br />

2. Vingert B, Adotevi O, Patin D, Jung S, Shrikant P, Freyburger L, Eppolito C,<br />

Sapoznikov A, Amessou M, Quintin-Colonna F, Fridman WH, Johannes L, Tartour<br />

E. The Shiga toxin B-subunit targets antigen in vivo to dendritic <strong>cells</strong> and elicits antitumor<br />

immunity. (2006) Eur J Immunol. 36(5):1124-35.<br />

3. Birnberg T., Bar-On L., Sapoznikov A., Caton M., Cervantes-Barragán L., Makia<br />

D., Krauthgamer R., Brenner O., Ludewig B., Brockschnieder D., Riethmacher D.,<br />

Reizis B., Jung S. Lack of conventional dendritic <strong>cells</strong> is compatible with normal<br />

development and T cell homeost<strong>as</strong>is, but causes myeloid proliferative syndrome.<br />

(2008) Immunity 29(6):986-97.<br />

4. Hartmann TN., Grabovsky V., Wang W., Desch P., Rubenzer G., Wollner S.,<br />

Binsky I., Vallon-Eberhard A., Sapoznikov A., Burger M, Shachar I, Haran M,<br />

Honczarenko M, Greil R, Alon R. Circulating B-cell chronic lymphocytic leukemia<br />

<strong>cells</strong> display impaired migration to lymph nodes and bone <strong>marrow</strong>. (2009) Cancer<br />

Res. 69(7):3121-30.<br />

8.3 Reviews<br />

1. Sapoznikov A., Jung S. Probing in vivo dendritic cell functions by conditional cell<br />

ablation. Review (2008) Immunol Cell Biol. 86(5):409-15.<br />

106


8. Statement about independent collaboration<br />

The manuscript entitled "Periv<strong>as</strong>cular clusters of dendritic <strong>cells</strong> provide critical<br />

survival signals to B <strong>cells</strong> in bone <strong>marrow</strong> niches" summarizes a study initiated by<br />

Dr. Yael Pewzner-Jung, who first observed the reduction of the B <strong>cells</strong> upon DC<br />

ablation. I substantiated the preliminary results and subsequently per<strong>for</strong>med all the<br />

experiments that led to the characterization of the underlying molecular mechanism.<br />

The two-photon imaging w<strong>as</strong> per<strong>for</strong>med with the help of Dr. Vyacheslav Kalchenko.<br />

The project entitled "The bone <strong>marrow</strong> immune niche: <strong>Bone</strong> <strong>marrow</strong> <strong>as</strong> a <strong>priming</strong><br />

<strong>site</strong> <strong>for</strong> naïve CD8 + T <strong>cells</strong>" is carried out in full collaboration with the laboratory of<br />

Dr. Guy Shakhar, (Department of Immunology, WIS), specifically with the PhD<br />

student Idan Milo. We contributed equally to the design of the experiments and<br />

discussions. I per<strong>for</strong>med and analyzed the experiments done by flow cytometry; the<br />

purification of the <strong>cells</strong>, the adoptive transfers, the chimera preparation and<br />

preparations of the <strong>cells</strong> and the mice <strong>for</strong> in vivo imaging. Idan per<strong>for</strong>med the<br />

intravital imaging and analyzed the results of the movies.<br />

107


סיכום<br />

T, ותאי<br />

תאים דנדריטיים משחקים תפקיד מרכזי בשפעולם של תאי T ומבקרים את התגובה האוטוריאקטיבית.‏<br />

תת-האוכלוסיות השונות של התאים הדנדריטים הינן בעלות תפקידים שונים.‏ למרות שהפנוטיפ והמיקום<br />

האנטומי של תת-האוכלוסיות ידוע,‏ תפקידיהם אינם מובנים עד תום.‏ חקר מעורבותם של תאים דנדריטיים<br />

במערכת יחסי גומלין עם תאים שונים,‏ כמו שפעול של מערכת החיסון או השתקתה,‏ דורש לימוד<br />

תפקודיהם בתוך מערכת פיסיולוגית.‏ בנוסף,‏ אוכלוסיית התאים הדנדריטיים הינה דינאמית ביותר.‏<br />

מסיבות אלו בעבודה זו נחקר תפקודם של התאים הללו באמצעות החסרתם המותנית בגוף החיי.‏<br />

בחלק הראשון של העבודה,‏ התבצע אפיון פנוטיפי ותפקודי של תאים דנדריטיים המאכלסים את מח<br />

העצם.‏ התגלו גומחות מיוחדות,‏ שתוחמו בצורה מבנית על-ידי מקבצים של תאים דנדריטיים,‏ אשר<br />

ממוקמים בקרבה לדפנות כלי דם מסוימים במצב שיווי-משקל.‏ גומחות אלה מאוכלסות בתאי B בוגרים<br />

דבר אשר מצביע על תפקידם במערכת חיסון הנרכשת.‏ להפתעתנו,‏ החסרה של תאים<br />

דנדריטיים במח העצם גרמה לירידה ספציפית בתאי B בוגרים בגומחות.‏ באמצעות שימוש במודלים של<br />

עכברים טרנסגניים ועכברים בהם בוצעה החסרה של גן מסוים,‏ גילינו את המנגנון המולקולארי,‏ אשר<br />

גרם לירידה בתאי B. נמצא כי,‏ תאים דנדריטיים במח העצם מספקים לתאי B פקטור הישרדותי.‏ פקטור<br />

B<br />

,MIF<br />

זה הינו הציטוקין אשר הכרחי לחיוניותם של תאי במח העצם.‏ זיהוי גומחות אלה,‏ המדמות<br />

פוליקלים לימפואידים במח העצם מהווה פריצת דרך בהבנה,‏ כי מח עצם יכול לשמש מקום לתגובות<br />

חיסוניות ראשוניות כנגד פתוגנים,‏ שמקורם בדם.‏<br />

בחלק השני של העבודה,‏ גילינו את המקור של תאים דנדריטיים במח העצם,‏ אשר נוצרים מאב קדמון של<br />

מאקרופאגים ותאים דנדריטיים,‏ הנקרא<br />

תנועתיותם של לימפוציטים מסוג<br />

,MDP<br />

T<br />

ללא שלב ביניים בו מתמיין למונוציטים.‏ בנוסף,‏ נחקרה<br />

ו-‏B בגומחות,‏ המאוכלסות בתאים דנדריטיים במצב של שיווי-‏<br />

משקל.‏ שימוש בשיטה חדשנית של דימות דו-פוטוני,‏ אפשרה להראות כי בגומחות המאוכלסות בתאים<br />

דנדריטיים,‏<br />

תאי T<br />

נאיביים מסוג + CD8 יוצרים מקבצים רב-תאיים ומתמיינים לתאים אפקטוריים,‏ לאחר<br />

מתן אנטיגן שמקורו בדם.‏ פעולה זאת מצביעה על תפקודו של מח העצם כעל איבר לימפואידי שניוני.‏<br />

באופן בלתי צפוי,‏ נגלה כי תאי<br />

CD8 + T<br />

נאיבים שופעלו באופן יעיל גם לאחר החסרה של תאים<br />

דנדריטיים במח העצם,‏ דבר אשר מטיל בספק את היותם של האחרונים הכרחיים כתאים מציגי אנטיגן<br />

באיבר זה.‏ שימוש בכימרות,‏ אפשר להבין כי תאים,‏ אשר עמידים בפני קרינה,‏ הנקראים גם תאי סטרומה<br />

היו חיוניים להצגה של אנטיגן ושפעול תאי CD8 + T בהעדר תאים דנדריטיים.‏ תוצאות אלו,‏ מדגישות את<br />

התפקיד החדש של תאים שאינם המטופואטיים במח העצם.‏<br />

לבסוף,‏ נחקרה יכולתם של תאים דנדריטיים מסוג פלסמציטואידיים<br />

תאי<br />

(PDCs)<br />

T<br />

להציג אנטיגנים.‏ תפקידם<br />

זה אינו מובן עד תום,‏ עקב מחסור במערכת ניסויית מתאימה.‏ במערכות הקיימות,‏ תפקידם לשפעל את<br />

תפקודם של תאי<br />

ממוסך על-ידי נוכחותם של תאים דנדריטיים קונבנציונאליים<br />

.(cDCs)<br />

,PDCs<br />

cDCs באופן מותנה,‏ אך לא נפגעו תאי<br />

נחקרו תגובותיהם של תאי T מסוג<br />

CD8 + - ו CD4 +<br />

.PDCs<br />

על מנת לעמוד על<br />

בעכברים,‏ בהם הוחסרו<br />

יתר על כך,‏ נעשה שימוש באסטרטגיה חדשה,‏ של הובלת<br />

108


PDCs<br />

cDCs או<br />

אנטיגן באמצעות נוגדן ספציפי לתאי .PDCs שימוש בשיטות אלה,‏ אפשר להראות כי<br />

אינם מסוגלים לתמוך בתגובות תאי T מכל סוג בטחולים של עכברים,‏ בהם הוחסרו עם זאת,‏<br />

להפתעתנו,‏ נמצא כי בהעדר<br />

.cDCs<br />

,cDCs תאי<br />

הלימפה.‏ באופן מעניין,‏ ובניגוד לתגובות המתווכות על-ידי<br />

PDCs גרמו לשפעול יעיל של תאי<br />

T מסוג + CD4 בבלוטות<br />

,cDCs<br />

PDCs נכשלו בשפעול תאי<br />

T מסוג<br />

.CD8 +<br />

לאור תוצאותינו במחקר זה,‏ ניתן להגדיר את תאי<br />

מסוגלים לאתחל תגובה ראשונית של תאי T עוזרים מסוג<br />

,PDCs<br />

.CD4 +<br />

כתאים דנדריטיים טיפוסיים,‏ אשר<br />

109

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

Saved successfully!

Ooh no, something went wrong!