07.03.2013 Views

Quintero et al. 2006 - Medical University of South Carolina

Quintero et al. 2006 - Medical University of South Carolina

Quintero et al. 2006 - Medical University of South Carolina

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.

Research Report<br />

Behavior<strong>al</strong> and morphologic<strong>al</strong> effects <strong>of</strong> minocycline in the<br />

6-hydroxydopamine rat model <strong>of</strong> Parkinson's disease<br />

Elias Matthew <strong>Quintero</strong> a, ⁎, Lauren Willis a , Rachel Singl<strong>et</strong>on a , Naida Harris b , Peng Huang c ,<br />

Narayan Bhat a , Ann-Charlotte Granholm a<br />

a<br />

Department <strong>of</strong> Neurosciences, Medic<strong>al</strong> <strong>University</strong> <strong>of</strong> <strong>South</strong> <strong>Carolina</strong>, 173 Ashley Avenue, Suite 403, Charleston, SC 29425, USA<br />

b<br />

Benedict College, Columbia, SC 29204, USA<br />

c<br />

Department <strong>of</strong> Biostatistics, Bioinformatics and Epidemiology, Medic<strong>al</strong> <strong>University</strong> <strong>of</strong> <strong>South</strong> <strong>Carolina</strong>, Charleston, SC 29425, USA<br />

ARTICLE INFO ABSTRACT<br />

Article history:<br />

Accepted 20 March <strong>2006</strong><br />

Available online 18 May <strong>2006</strong><br />

Keywords:<br />

Parkinson's disease<br />

6-Hydroxydopamine<br />

Minocycline<br />

Apomorphine<br />

Inflammation<br />

Neuroglia<br />

1. Introduction<br />

Neuroinflammation mediated by activated gli<strong>al</strong> cells has<br />

recently been investigated as a causatory factor for Parkin-<br />

⁎ Corresponding author. Fax: +1 843 792 4423.<br />

E-mail address: quintero@musc.edu (E.M. <strong>Quintero</strong>).<br />

BRAIN RESEARCH 1093 (<strong>2006</strong>) 198– 207<br />

available at www.sciencedirect.com<br />

www.elsevier.com/locate/brainres<br />

0006-8993/$ – see front matter © <strong>2006</strong> Elsevier B.V. All rights reserved.<br />

doi:10.1016/j.brainres.<strong>2006</strong>.03.104<br />

The neuropathology in many neurodegenerative diseases is mediated by inflammatory<br />

cascades that influence neuron<strong>al</strong> dysfunction and death. Minocycline reduces the<br />

neurodegeneration observed in various models <strong>of</strong> Parkinson's. We exploited the unilater<strong>al</strong><br />

6-hydroxydopamine (6-OHDA) lesion model to assess the effect <strong>of</strong> minocycline on related<br />

neurodegeneration. Thirty Fisher 344 rats were divided into three daily treatment groups: (1)<br />

after: 45 mg/kg <strong>of</strong> minocycline beginning 24 h after lesioning; (2) before: 45 mg/kg <strong>of</strong><br />

minocycline beginning 3 days before 6-OHDA lesioning; (3) control: corresponding s<strong>al</strong>in<strong>et</strong>reated<br />

controls. Anim<strong>al</strong>s were assessed for apomorphine-induced rotations for 4 weeks. A<br />

longitudin<strong>al</strong> model for repeated measures showed that both after and before groups had<br />

significantly lower rotations than controls (P < 0.001 for both comparisons). Pair-wise group<br />

comparisons showed that the before anim<strong>al</strong>s rotated less compared to controls (mean<br />

rotations: 164 ± 38 versus 386 ± 49, respectively, P = 0.001). After anim<strong>al</strong>s <strong>al</strong>so rotated<br />

significantly less then controls (mean rotations: 125 ± 41 versus 386 ± 49, respectively,<br />

P < 0.001). Anim<strong>al</strong>s receiving minocycline displayed reduced tyrosine hydroxylase-positive<br />

cell loss in the lesioned nigra versus contr<strong>al</strong>ater<strong>al</strong> nonlesioned nigra, compared to controls<br />

(mean differences: 5065 for after, 3550 for before, and 6483 for controls; P = 0.158 for after<br />

versus controls, P = 0.019 for before versus controls). The remaining lesioned nigr<strong>al</strong> cells <strong>of</strong><br />

both minocycline-treated groups were larger than controls, with the most robust cell size<br />

and fiber density observed in the after group. These data suggest that the therapeutic<br />

potenti<strong>al</strong> <strong>of</strong> minocycline may depend on the time <strong>of</strong> drug administration relative to<br />

neuropathogenic event.<br />

© <strong>2006</strong> Elsevier B.V. All rights reserved.<br />

son's disease (PD), an age-related neurodegenerative disease<br />

(Barcia <strong>et</strong> <strong>al</strong>., 2003; Dawson and Dawson, 2003). Studies have<br />

shown that activated microgli<strong>al</strong> cells surround dopaminergic<br />

neurons in patients with this disease and produce damaging


cytokines that may be directly responsible for the demise <strong>of</strong><br />

the dopamine (DA) neurons (Hirsch <strong>et</strong> <strong>al</strong>., 2003; Hunot and<br />

Hirsch, 2003). Activated glia (astrocytes and microglia) <strong>al</strong>so<br />

contribute towards the rampant oxidative stress that is<br />

associated with the neurodegenerative process via excessive<br />

production <strong>of</strong> neurotoxic free radic<strong>al</strong>s (Abramov <strong>et</strong> <strong>al</strong>., 2004;<br />

Leblhuber <strong>et</strong> <strong>al</strong>., 2003). Although astrocytes norm<strong>al</strong>ly protect<br />

neurons through their production <strong>of</strong> growth and trophic<br />

molecules, “aging” astrocytes, because <strong>of</strong> their expression <strong>of</strong><br />

pro-oxidant enzymes such as MAO-B and a high content <strong>of</strong><br />

free iron, can promote enzymatic and nonenzymatic oxidation<br />

<strong>of</strong> DA, generating toxic levels <strong>of</strong> reactive oxygen species<br />

(Johnstone <strong>et</strong> <strong>al</strong>., 1999; Hirsch <strong>et</strong> <strong>al</strong>., 2003).<br />

Minocycline, a second generation t<strong>et</strong>racycline, has recently<br />

f<strong>al</strong>len under close scrutiny as having possible therapeutic v<strong>al</strong>ue<br />

for treating sever<strong>al</strong> neurodegenerative disorders, including<br />

Huntington's disease (Chen <strong>et</strong> <strong>al</strong>., 2000), amyotrophic later<strong>al</strong><br />

sclerosis (Zhu <strong>et</strong> <strong>al</strong>., 2002), and autoimmune enceph<strong>al</strong>omyelitis<br />

(Popovic <strong>et</strong> <strong>al</strong>., 2001). Studies in models <strong>of</strong> PD, including the 1m<strong>et</strong>hyl-4-phenyl-1,2,3,6-t<strong>et</strong>rahydropyridine<br />

(MPTP) model (Du<br />

<strong>et</strong> <strong>al</strong>., 2001; Wu <strong>et</strong> <strong>al</strong>., 2002) and the 6-hydroxydopamine (6-<br />

OHDA) model (He <strong>et</strong> <strong>al</strong>., 2001), have provided compelling<br />

evidence that minocycline possesses vast potenti<strong>al</strong> for attenuation<br />

<strong>of</strong> the neurodegenerative changes that occur in the brain.<br />

In particular, He <strong>et</strong> <strong>al</strong>. (2001) compared the effects <strong>of</strong><br />

minocycline treatment to s<strong>al</strong>ine controls on nigr<strong>al</strong> dopaminergic<br />

neuron<strong>al</strong> and microgli<strong>al</strong> status in m<strong>al</strong>e mice lesioned in the<br />

right striatum with 6-OHDA. Tyrosine hydroxylase (TH) immunohistochemistry<br />

reve<strong>al</strong>ed a significant sparing <strong>of</strong> dopamine<br />

neurons in anim<strong>al</strong>s receiving minocycline treatment. In<br />

addition, the number <strong>of</strong> activated microglia was significantly<br />

decreased in those anim<strong>al</strong>s that received minocycline compared<br />

to their s<strong>al</strong>ine counterparts. Lin <strong>et</strong> <strong>al</strong>. (2003) reported that<br />

minocycline reduced the susceptibility <strong>of</strong> cultured rat cerebellar<br />

granule neurons to 6-OHDA-induced death and prevented the<br />

production <strong>of</strong> free radic<strong>al</strong>s following 6-OHDA administration.<br />

These two studies clearly demonstrate that minocycline<br />

impacts the toxicity associated with 6-OHDA administration<br />

and establish the possibility <strong>of</strong> manipulating the degenerative<br />

cascade that follows upon introduction <strong>of</strong> this toxin. However,<br />

6-OHDA studies in the rat model combined with minocycline<br />

have been sparse. The <strong>al</strong>lure <strong>of</strong> this phenomenon becomes<br />

apparent when one considers that the biochemic<strong>al</strong> and cellular<br />

events associated with 6-OHDA toxicity may be, at least in part,<br />

similar to the molecular neuroinflammatory cascades that are<br />

engaged in the ons<strong>et</strong> <strong>of</strong> neurodegenerative diseases such as<br />

Parkinson's disease (Hirsch <strong>et</strong> <strong>al</strong>., 1998; Jenner and Olanow,<br />

1998).<br />

Since its first use by Ungerstedt in the late 1960s (Ungerstedt,<br />

1968), the utilization <strong>of</strong> the catecholaminergic neurotoxin<br />

6-hydroxydopamine (6-OHDA) to experiment<strong>al</strong>ly destroy<br />

dopaminergic neurons in the brain has been well documented<br />

and is regarded as the gold standard for studying mesenceph<strong>al</strong>ic<br />

dopamine (DA)-containing neurons (Schwarting and<br />

Huston, 1996a,b; Glinka <strong>et</strong> <strong>al</strong>., 1997; Blum <strong>et</strong> <strong>al</strong>., 2001; Deumens<br />

<strong>et</strong> <strong>al</strong>., 2002). By stereotaxic<strong>al</strong>ly introducing 6-OHDA into<br />

individu<strong>al</strong> components <strong>of</strong> the nigrostriat<strong>al</strong> pathway, we<br />

have gained enormous insight into the neuroanatomic<strong>al</strong>,<br />

neurochemic<strong>al</strong>, and electrophysiologic<strong>al</strong> param<strong>et</strong>ers <strong>of</strong> DAcontaining<br />

neurons and their interaction with other sys-<br />

BRAIN RESEARCH 1093 (<strong>2006</strong>) 198– 207<br />

tems. The most common application <strong>of</strong> this toxin is the<br />

creation <strong>of</strong> a unilater<strong>al</strong> lesion in mesenceph<strong>al</strong>ic DA neurons<br />

or their ascending fibers, which will result in a loss <strong>of</strong> targ<strong>et</strong><br />

striat<strong>al</strong> DA, or into the striatum directly. Consequently, the<br />

efficacy <strong>of</strong> a 6-OHDA unilater<strong>al</strong> lesion can be demonstrated<br />

by DAergic compounds, such as apomorphine or amph<strong>et</strong>amine<br />

(Ungerstedt and Arbuthnott, 1970). The number <strong>of</strong><br />

apomorphine-induced turns per hour has been directly<br />

correlated to the extent <strong>of</strong> DA loss when 6-OHDA is injected<br />

into the medi<strong>al</strong> forebrain bundle (Hudson <strong>et</strong> <strong>al</strong>., 1993). This<br />

strategy has been proven to be an inv<strong>al</strong>uable tool in<br />

studying how diseases, such as PD, may encroach upon<br />

the nigrostriat<strong>al</strong> pathway and how experiment<strong>al</strong> manipulation<br />

<strong>of</strong> this system may provide clues for possible clinic<strong>al</strong><br />

intervention.<br />

For this study, we hypothesize that minocycline will reduce<br />

the number <strong>of</strong> lesioned nigr<strong>al</strong> dopaminergic cells and will<br />

reduce the number <strong>of</strong> apomorphine-induced rotations in rats<br />

when administered prior to or after receiving a unilater<strong>al</strong> 6-<br />

OHDA lesion. In order to test the preventive effects <strong>of</strong> this<br />

drug, daily intraperitone<strong>al</strong> injections <strong>of</strong> s<strong>al</strong>ine or minocycline<br />

were initiated 3 days prior to neurotoxic lesioning and<br />

continued for 4 weeks postlesioning. We <strong>al</strong>so assessed the<br />

rescue effects <strong>of</strong> minocycline by administering the drug (as<br />

well as s<strong>al</strong>ine control counterparts) 24 h following 6-OHDA<br />

lesioning and continuing for 4 weeks after surgery. In order to<br />

obtain behavior<strong>al</strong> evidence <strong>of</strong> the neuroprotective properties<br />

<strong>of</strong> this t<strong>et</strong>racycline, weekly apomorphine-induced rotation<strong>al</strong><br />

behavior was assessed on <strong>al</strong>l subjects, and histologic<strong>al</strong><br />

an<strong>al</strong>ysis <strong>of</strong> surviving dopaminergic neurons was performed<br />

following sacrifice <strong>of</strong> <strong>al</strong>l anim<strong>al</strong>s.<br />

2. Results<br />

2.1. Effects <strong>of</strong> minocycline on apomorphine-induced<br />

rotations<br />

199<br />

Apomorphine-induced rotation<strong>al</strong> an<strong>al</strong>ysis was performed in<br />

order to examine the effect <strong>of</strong> minocycline treatment on the<br />

surviv<strong>al</strong> <strong>of</strong> dopaminergic neurons exposed to 6-OHDA. All<br />

anim<strong>al</strong>s were an<strong>al</strong>yzed for apomorphine-induced rotations for<br />

1 h, once a week, for 4 weeks following lesioning (previous<br />

studies indicate that 6-OHDA requires 4 weeks before the full<br />

rotation<strong>al</strong> potenti<strong>al</strong> <strong>of</strong> each anim<strong>al</strong> is re<strong>al</strong>ized (Ungerstedt and<br />

Arbuthnott, 1970; Sauer and Oertel, 1994; Gerlach and<br />

Riederer, 1996). Fig. 1 is a graphic<strong>al</strong> representation <strong>of</strong> the<br />

rotation<strong>al</strong> assessments for the s<strong>al</strong>ine-treated anim<strong>al</strong>s (control<br />

group), minocycline 3 days before lesioning (before group), and<br />

minocycline 24 h after lesioning (after group) over the 4-week<br />

testing period following 6-OHDA administration. The control<br />

group consistently rotated the most number <strong>of</strong> times at each<br />

weekly time point, compared to those anim<strong>al</strong>s that received<br />

minocycline. At 1 week, the mean rotation rate for the controls<br />

was 202 turns per hour compared to 140 turns in the before<br />

and 34 turns in the after groups (n = 10). At 2 weeks postlesion,<br />

the controls rotated 381 turns per hour, whereas the before<br />

and after anim<strong>al</strong>s rotated 138 and 114 turns per hour,<br />

respectively. Three weeks following toxin administration<br />

resulted in 359 turns in the controls, 246 turns in the before


200 BRAIN RESEARCH 1093 (<strong>2006</strong>) 198– 207<br />

Fig. 1 – Minocycline reduces the number <strong>of</strong><br />

apomorphine-induced rotations in the 6-OHDA-lesioned rat.<br />

All anim<strong>al</strong>s that received minocycline rotated significantly<br />

fewer times over the 4-week testing period compared to their<br />

s<strong>al</strong>ine-treated counterparts (P < 0.001). At week 2, anim<strong>al</strong>s<br />

treated before (Before) and after (After) 6-OHDA lesioning<br />

rotated significantly fewer times than the s<strong>al</strong>ine-treated<br />

(Control) anim<strong>al</strong>s (138, 114 and 381 turns, respectively.<br />

P < 0.05). At week 4, the difference in rotations in the after<br />

treated group approached significance (P = 0.07) compared to<br />

the s<strong>al</strong>ine control group.<br />

and 148 turns in the after. Fin<strong>al</strong>ly, the fourth week <strong>of</strong> rotation<strong>al</strong><br />

an<strong>al</strong>ysis reve<strong>al</strong>ed that the controls rotated 430 turns per hour,<br />

while the before rotated 394 turns and the after rotated 149<br />

turns.<br />

A one-way ANOVA showed that there was a significant<br />

difference among the 3 groups in over<strong>al</strong>l turns across <strong>al</strong>l<br />

time points (P < 0.0001). Post hoc two-sample t tests to<br />

compare the after group versus controls, before group versus<br />

controls, and combined after and before versus controls <strong>al</strong>l<br />

yielded P v<strong>al</strong>ues


minocycline treatment. Fig. 2 demonstrates the contrast<br />

b<strong>et</strong>ween lesioned and nonlesioned nigras <strong>of</strong> <strong>al</strong>l three<br />

groups. The dopaminergic neurons <strong>of</strong> the lesioned nigra<br />

readily succumbed to 6-OHDA when treated with s<strong>al</strong>ine<br />

only (Fig. 2A). These results were identic<strong>al</strong> wh<strong>et</strong>her s<strong>al</strong>ine<br />

treatment was initiated before or after 6-OHDA administration.<br />

When minocycline was administered 3 days prior<br />

to 6-OHDA administration and continuously thereafter,<br />

there was a marked sparing <strong>of</strong> dopaminergic neurons in<br />

the substantia nigra 4 weeks following toxin introduction<br />

(Fig. 2B). There was <strong>al</strong>so a similar demonstration <strong>of</strong> nigr<strong>al</strong><br />

cell surviv<strong>al</strong> when minocycline was administered continuously<br />

starting 24 h after 6-OHDA lesioning (Fig. 2C).<br />

Stereologic<strong>al</strong> cell counts <strong>of</strong> TH-positive lesioned and<br />

contr<strong>al</strong>ater<strong>al</strong> nonlesioned nigr<strong>al</strong> cells in each anim<strong>al</strong> was<br />

performed to d<strong>et</strong>ermine the number <strong>of</strong> surviving DA cells 4<br />

weeks after lesioning. To an<strong>al</strong>yze wh<strong>et</strong>her minocycline was<br />

efficient in reducing the number <strong>of</strong> nigr<strong>al</strong> cells that<br />

succumbed to 6-OHDA exposure, we first computed the<br />

difference in cell numbers by subtracting the nigr<strong>al</strong> cell<br />

count <strong>of</strong> the lesioned side from the nigr<strong>al</strong> cell count <strong>of</strong> the<br />

contr<strong>al</strong>ater<strong>al</strong> nonlesioned side in each anim<strong>al</strong> (Fig. 4A). We<br />

then compared the mean difference measures among the<br />

three groups (after, before and control). A one-way an<strong>al</strong>ysis<br />

<strong>of</strong> variance showed that the mean cell differences were<br />

not the same among the three groups (P = 0.034). The<br />

mean cell difference in the controls was 6483 ± 782. The<br />

mean cell difference in <strong>al</strong>l minocycline-treated anim<strong>al</strong>s<br />

(after and before groups combined) was 4388 ± 446. A twosided<br />

t test generated a P v<strong>al</strong>ue <strong>of</strong> 0.047 (P < 0.05), implying<br />

the effectiveness <strong>of</strong> the minocycline treatment. We next<br />

examined wh<strong>et</strong>her early minocycline treatment (before<br />

group) or late minocycline treatment (after group) gives<br />

the best protection for cells against 6-OHDA exposure. We<br />

BRAIN RESEARCH 1093 (<strong>2006</strong>) 198– 207<br />

compared the mean cell differences <strong>of</strong> each minocycline<br />

group (after or before) separately with the control group<br />

v<strong>al</strong>ues using a two-sided t test. The observed mean cell<br />

differences were 3550 ± 628 and 5065 ± 476 in before and<br />

after anim<strong>al</strong>s, respectively. The early minocycline (before)<br />

treated group demonstrated a significant reduction in cell<br />

count loss (P < 0.05) compared to the controls, while the<br />

difference b<strong>et</strong>ween the late (after) minocycline subjects<br />

and controls was not significant (P > 0.1). These data imply<br />

that fewer nigr<strong>al</strong> cells succumbed to 6-OHDA-induced<br />

toxicity in anim<strong>al</strong>s treated with minocycline, particularly<br />

those that began the t<strong>et</strong>racycline three days prior to<br />

lesioning.<br />

Cell size measurements on randomly selected neurons<br />

for each anim<strong>al</strong> (>50 neurons/anim<strong>al</strong>) reve<strong>al</strong>ed a significant<br />

difference in cell size in the lesioned nigra b<strong>et</strong>ween the<br />

three groups (Fig. 4B). The before group had significantly<br />

sm<strong>al</strong>ler neurons than the after group (746.87 ± 35.121<br />

versus 897.07 ± 27.447 square pixels, respectively). The<br />

sm<strong>al</strong>lest cell size measurements were obtained in the<br />

controls (700.62 ± 185.240 square pixels). Significant cell<br />

size differences were observed b<strong>et</strong>ween the before and<br />

after group (P < 0.001) as well as b<strong>et</strong>ween the after and<br />

controls (P < 0.0001). In addition, remaining nigr<strong>al</strong> cells in<br />

the after group appeared more robust with thick neuritic<br />

processes, suggesting intact neuron<strong>al</strong> viability (Fig. 3C).<br />

Thus, it appeared that the cell size, but not the cell<br />

numbers, reflected the <strong>al</strong>terations seen in apomorphineinduced<br />

behavior (see Fig. 3). Subsequent densitom<strong>et</strong>ric<br />

measurements <strong>of</strong> TH-positive nigr<strong>al</strong> fibers <strong>of</strong> <strong>al</strong>l three<br />

groups reve<strong>al</strong>ed that anim<strong>al</strong>s treated with minocycline<br />

24 h after lesioning displayed more numerous thick<br />

sprouting processes compared to those treated with<br />

minocycline 3 days before or s<strong>al</strong>ine only (Fig. 4C). This<br />

Fig. 3 – Minocycline increases TH-immunoreactive cell size and fiber density in lesioned nigras. S<strong>al</strong>ine-treated anim<strong>al</strong>s display<br />

typic<strong>al</strong> degenerative pr<strong>of</strong>iles including sm<strong>al</strong>l, weakly stained cell bodies (A) and sparse dendritic processes (D). However,<br />

minocycline treatment before lesioning (B) and after lesioning (C) appears to enhance neuron<strong>al</strong> cell size and fiber density,<br />

particularly in the after group. Panels (D), (E), and (F) illustrate later<strong>al</strong> nigr<strong>al</strong> fibers <strong>of</strong> s<strong>al</strong>ine-, minocycline before-, and minocycline<br />

after-treated groups, respectively. Note the robust fiber sprouting and density in the minocycline-treated groups, particularly in<br />

the after lesion group (F). (A, B, C: 60× magnification, sc<strong>al</strong>e bar = 20 μm; D, E, F: 40× magnification, sc<strong>al</strong>e bar = 15 μm).<br />

201


202 BRAIN RESEARCH 1093 (<strong>2006</strong>) 198– 207<br />

Fig. 4 – Minocycline reduces TH-positive cell loss and<br />

increases the size and fiber density <strong>of</strong> remaining nigr<strong>al</strong> cells.<br />

The degree <strong>of</strong> cell loss is more attenuated in the minocycline<br />

before group than the after group, compared to the s<strong>al</strong>ine<br />

controls (A, *P < 0.05). However, close inspection reve<strong>al</strong>s that<br />

anim<strong>al</strong>s receiving minocycline after lesioning contained<br />

larger, more he<strong>al</strong>thy looking cells than either the minocycline<br />

before or s<strong>al</strong>ine controls (B, *P < 0.001, **P < 0.0001). In<br />

addition, the lesioned nigras <strong>of</strong> <strong>al</strong>l minocycline-treated<br />

anim<strong>al</strong>s contained more dense sprouting fibers compared to<br />

controls, especi<strong>al</strong>ly in the after group (C). See text for d<strong>et</strong>ails.<br />

was particularly evident in the later<strong>al</strong> nigra <strong>of</strong> <strong>al</strong>l anim<strong>al</strong>s<br />

an<strong>al</strong>yzed.<br />

3. Discussion<br />

Apomorphine-induced rotation<strong>al</strong> results and TH immunohistochemistry<br />

an<strong>al</strong>yses support our hypothesis that minocycline<br />

reduces the number <strong>of</strong> rotations and spares DA-containing<br />

nigr<strong>al</strong> neurons in 6-OHDA unilater<strong>al</strong>ly lesioned rats. We<br />

utilized apomorphine to ev<strong>al</strong>uate the extent <strong>of</strong> DA depl<strong>et</strong>ion<br />

because it serves as a more reliable predictor <strong>of</strong> lesion efficacy<br />

following 6-OHDA administration and avoids the possibility <strong>of</strong><br />

sensitization associated with D-amph<strong>et</strong>amine (Hudson <strong>et</strong> <strong>al</strong>.,<br />

1993; Carman <strong>et</strong> <strong>al</strong>., 1991; Casas <strong>et</strong> <strong>al</strong>., 1988). Anim<strong>al</strong>s receiving<br />

daily minocycline beginning 3 days prior to lesioning rotated<br />

<strong>al</strong>most h<strong>al</strong>f as many times as their s<strong>al</strong>ine-treated counterparts.<br />

Surprisingly, those anim<strong>al</strong>s that began their regimen <strong>of</strong><br />

minocycline 24 h after lesioning displayed over a 50%<br />

reduction in apomorphine-induced rotations compared to<br />

their corresponding s<strong>al</strong>ine-treated lesioned counterparts,<br />

suggesting that minocycline optim<strong>al</strong>ly spared DA depl<strong>et</strong>ion<br />

when administered after surgic<strong>al</strong> lesioning. Stereologic counts<br />

and cell size measurements demonstrated a greater sparing <strong>of</strong><br />

neurons on the lesioned side <strong>of</strong> anim<strong>al</strong>s that received<br />

minocycline before the lesion, <strong>al</strong>though these cells displayed<br />

a more atrophied pr<strong>of</strong>ile. In contrast, fewer nigr<strong>al</strong> cells<br />

remained in anim<strong>al</strong>s receiving minocycline after the lesion.<br />

Interestingly, the remaining nigr<strong>al</strong> cells in anim<strong>al</strong>s treated<br />

with minocycline after lesioning appeared much larger than<br />

the lesioned nigr<strong>al</strong> cells in the other two groups and contained<br />

numerous thick, darkly stained neuron<strong>al</strong> fibers, particularly in<br />

the later<strong>al</strong> nigr<strong>al</strong> area. It is possible that minocycline administered<br />

24 h after lesioning promoted he<strong>al</strong>thier surviving THpositive<br />

nigr<strong>al</strong> cells, compared to anim<strong>al</strong>s that began their<br />

treatment three days prior to lesioning.<br />

When 6-OHDA injections are administered into the MFB,<br />

there is minim<strong>al</strong> variability, and there is >90% success in<br />

obtaining anim<strong>al</strong>s with more than 90% reduction in striat<strong>al</strong> DA<br />

levels using this m<strong>et</strong>hod (Hudson <strong>et</strong> <strong>al</strong>., 1993; Bowenkamp <strong>et</strong><br />

<strong>al</strong>., 1995; Granholm <strong>et</strong> <strong>al</strong>., 1997). Thus, the technique is reliable<br />

and reproducible. Extensive use <strong>of</strong> 6-OHDA in our laboratory<br />

and from other investigators has clearly demonstrated that<br />

sham-operated controls do not display neurodegenerative<br />

characteristics typic<strong>al</strong>ly present in toxin-treated subjects. In<br />

recent studies in our laboratory utilizing minocycline, Hunter<br />

<strong>et</strong> <strong>al</strong>. reported that the number <strong>of</strong> remaining cells in shamlesioned<br />

anim<strong>al</strong>s treated with minocycline did not differ from<br />

that in sham-lesioned anim<strong>al</strong>s receiving s<strong>al</strong>ine (Hunter <strong>et</strong> <strong>al</strong>.,<br />

2004b), suggesting that minocycline did not <strong>al</strong>ter the bas<strong>al</strong><br />

histologic<strong>al</strong> pr<strong>of</strong>ile observed in host brains. Based on the fact<br />

that we found no effects <strong>of</strong> minocycline in control anim<strong>al</strong>s<br />

after up to 3 months <strong>of</strong> treatment (Hunter <strong>et</strong> <strong>al</strong>., 2004b), we<br />

opted to not include sham-lesioned controls in the present<br />

study. In addition, inspection <strong>of</strong> the contr<strong>al</strong>ater<strong>al</strong> histologic<strong>al</strong><br />

data among <strong>al</strong>l groups in this study corroborates the ration<strong>al</strong>e<br />

to exclude sham-lesioned anim<strong>al</strong>s in that there are no<br />

observable effects <strong>of</strong> minocycline on the unlesioned side <strong>of</strong><br />

the brain.<br />

In order to interpr<strong>et</strong> the data presented here, one must<br />

consider the time course <strong>of</strong> degeneration in nigr<strong>al</strong> dopaminergic<br />

neurons following a 6-OHDA lesion. Earlier studies<br />

using 6-OHDA have d<strong>et</strong>ermined that the behavior<strong>al</strong> phenotype<br />

begins 1 week following toxin administration, when there<br />

is a greater than 60–80% depl<strong>et</strong>ion <strong>of</strong> striat<strong>al</strong> DA; this<br />

behavior<strong>al</strong> phenotype is fully evident at about 4 weeks<br />

(Ungerstedt and Arbuthnott, 1970; Sauer and Oertel, 1994;<br />

Gerlach and Riederer, 1996). In a previous study to d<strong>et</strong>ermine<br />

the tempor<strong>al</strong> pattern <strong>of</strong> acute phenotypic and degenerative<br />

<strong>al</strong>terations following 6-OHDA lesions in the MFB <strong>of</strong> rats, Zuch<br />

<strong>et</strong> <strong>al</strong>. (Zuch <strong>et</strong> <strong>al</strong>., 2000) demonstrated that events leading to<br />

dopaminergic neuron<strong>al</strong> loss begin <strong>al</strong>most immediately


following introduction <strong>of</strong> the toxin. FluoroJade staining, a<br />

marker for cell death, was evident in the ipsilater<strong>al</strong> substantia<br />

nigra pars compacta as early as 6 h postlesion, peaking at 48 h<br />

following toxin administration. Tyrosine hydroxylase immunohistochemistry<br />

<strong>of</strong> corresponding sections reve<strong>al</strong>ed that<br />

degenerative <strong>al</strong>terations to DA neurons were evident at 6 h<br />

postlesion and progressed to a dramatic loss <strong>of</strong> TH immunoreactivity<br />

in the ventromedi<strong>al</strong> substantia nigra pars compacta<br />

by 96 h postlesion. These data clearly demonstrate that the<br />

degenerative changes following 6-OHDA administration begin<br />

within the first 24 h after exposure.<br />

Early characterizations <strong>of</strong> the mechanisms <strong>of</strong> 6-OHDAinduced<br />

degeneration have established that the toxin is<br />

oxidized to reactive oxygen species, affects apoptosis (Choi<br />

<strong>et</strong> <strong>al</strong>., 1999; Lotharius <strong>et</strong> <strong>al</strong>., 1999) and promotes the<br />

production <strong>of</strong> proinflammatory cytokines (Mladenovic <strong>et</strong> <strong>al</strong>.,<br />

2004; Mogi <strong>et</strong> <strong>al</strong>., 1999). There is well documented evidence<br />

that minocycline imparts its anti-inflammatory properties by<br />

inhibiting the activation <strong>of</strong> microglia and attenuating the p38<br />

MAPK cascade with resultant reduction <strong>of</strong> inflammatory<br />

cytokine synthesis (Yrjänheikki <strong>et</strong> <strong>al</strong>., 1998; Tikka <strong>et</strong> <strong>al</strong>.,<br />

2001; Wu <strong>et</strong> <strong>al</strong>., 2002). These anti-inflammatory properties<br />

were origin<strong>al</strong>ly identified in studies <strong>of</strong> brain ischemia<br />

(Yrjänheikki <strong>et</strong> <strong>al</strong>., 1998; 1999). In addition, previous reports<br />

reve<strong>al</strong> that minocycline is capable <strong>of</strong> blocking components <strong>of</strong><br />

pro-apoptotic pathways, including a reduction in the formation<br />

<strong>of</strong> caspase-3 and caspase-1 (Zhu <strong>et</strong> <strong>al</strong>., 2002; Wang <strong>et</strong> <strong>al</strong>.,<br />

2003; Arvin <strong>et</strong> <strong>al</strong>., 2002; Chen <strong>et</strong> <strong>al</strong>., 2000). It is likely that these<br />

events are being influenced by the minocycline administration<br />

in our study. Therefore, this drug may be an effective tool<br />

in d<strong>et</strong>erring both caspase-dependent and -independent<br />

components <strong>of</strong> neurodegenerative cascades.<br />

It is possible that the differences in toxin susceptibility<br />

observed in this study could be due to an interaction b<strong>et</strong>ween<br />

minocycline and 6-OHDA such that the full effect <strong>of</strong> the toxin<br />

may not have been re<strong>al</strong>ized. For instance, it is evident that<br />

the reduction in number <strong>of</strong> lesioned nigr<strong>al</strong> cells by 6-OHDA<br />

was more attenuated by minocycline in the before than the<br />

after group. However, an interaction b<strong>et</strong>ween minocycline<br />

and 6-OHDA is unlikely. T<strong>et</strong>racyclines such as minocycline<br />

impart their bacteriostatic effects by binding to a single high<br />

affinity site on the bacteri<strong>al</strong> 30S ribosome within the cell,<br />

effectively blocking the entry <strong>of</strong> aminoacyl transfer RNA into<br />

the A site <strong>of</strong> the ribosome, preventing elongation <strong>of</strong> amino<br />

acid residues into polypeptide chains, and inhibiting protein<br />

synthesis (Zhanel <strong>et</strong> <strong>al</strong>., 2004). As for its anti-inflammatory<br />

properties, the literature fully supports two mechanisms <strong>of</strong><br />

cell death prevention by t<strong>et</strong>racyclines: potent inhibition <strong>of</strong><br />

innate and adaptive immunity components and obstruction<br />

<strong>of</strong> apoptotic cascades (Domercg and Matute, 2004). Fin<strong>al</strong>ly, a<br />

comprehensive consultation with the Medic<strong>al</strong> <strong>University</strong> <strong>of</strong><br />

<strong>South</strong> <strong>Carolina</strong> Drug Information Center has reve<strong>al</strong>ed no<br />

reported documentation or theor<strong>et</strong>ic<strong>al</strong> interaction b<strong>et</strong>ween<br />

minocycline (or other similar t<strong>et</strong>racyclines) and DA or DA<br />

an<strong>al</strong>ogs, such as L-DOPA and hydroxydopamine, based on 12<br />

major drug references, including many primary literature<br />

sources (person<strong>al</strong> communication).<br />

Current publications have proposed that the proinflammatory<br />

response <strong>of</strong> microgli<strong>al</strong> cells in the brain may consist <strong>of</strong><br />

both del<strong>et</strong>erious and benefici<strong>al</strong> responses (V<strong>al</strong>lat-Decouve-<br />

BRAIN RESEARCH 1093 (<strong>2006</strong>) 198– 207<br />

203<br />

laere <strong>et</strong> <strong>al</strong>., 2003; Teismann <strong>et</strong> <strong>al</strong>., 2003). Most recently, a study<br />

examining the effects <strong>of</strong> blueberry- and spirulina-enriched<br />

di<strong>et</strong>s on striat<strong>al</strong> dopamine recovery following 6-OHDA lesioning<br />

reve<strong>al</strong>ed that an early phase <strong>of</strong> activated microgli<strong>al</strong><br />

response is associated with successful dopaminergic protection<br />

(Strömberg <strong>et</strong> <strong>al</strong>., 2005). When the activated microgliaassociated<br />

OX-6 expression was suppressed using antisense to<br />

TNFα, more severe dopamine depl<strong>et</strong>ion was observed. Alternatively,<br />

others have reported that delayed inhibition <strong>of</strong><br />

cytokine release and microgli<strong>al</strong> activation following 6-OHDA<br />

lesioning will result in less severe dopamine destruction<br />

(Gemma <strong>et</strong> <strong>al</strong>., 2003). Collectively, these data support the<br />

possibility <strong>of</strong> an initi<strong>al</strong> benefici<strong>al</strong> microgli<strong>al</strong> response to<br />

neuroinflammation and, if left unabated, subsequent d<strong>et</strong>riment<strong>al</strong><br />

effect. If this phenomenon holds true, it may explain<br />

why we observed a greater loss <strong>of</strong> TH-positive nigr<strong>al</strong> cells but<br />

b<strong>et</strong>ter rotation<strong>al</strong> phenotype in the minocycline after group,<br />

compared to the minocycline before group. Because the early<br />

benefici<strong>al</strong> wave was reduced in the before group but was<br />

present in the after group, the remaining nigr<strong>al</strong> cells may be<br />

more he<strong>al</strong>thy with enhanced fiber arborization in the latter,<br />

suggesting continued nigr<strong>al</strong> destruction but more viable<br />

dopaminergic function because this first wave <strong>of</strong> gli<strong>al</strong><br />

response is preserved. Indeed, upon examination <strong>of</strong> <strong>al</strong>l<br />

histologic<strong>al</strong> preparations for this study, we observed a marked<br />

increase in size and fiber density <strong>of</strong> the remaining TH-positive<br />

cells in anim<strong>al</strong>s treated with minocycline 24 h after 6-OHDA<br />

lesioning, compared to the other two groups. Neuron<strong>al</strong> cell<br />

size measurements reve<strong>al</strong>ed that, <strong>al</strong>though stereology identified<br />

more TH-positive cells in the lesioned nigra <strong>of</strong> anim<strong>al</strong>s<br />

exposed to minocycline before neurotoxin exposure, these<br />

cells displayed more classic apoptotic pr<strong>of</strong>iles (e.g., sm<strong>al</strong>ler,<br />

ruptured cells with dense, clumped vesicular bodies in the<br />

neuropil, <strong>et</strong>c.) than cells in the after group. Addition<strong>al</strong>ly, many<br />

<strong>of</strong> the lesioned nigr<strong>al</strong> neurons in the before group displayed<br />

fewer or absent neuritic processes when compared to the after<br />

group. We propose that the b<strong>et</strong>ter outcome in apomorphine<br />

rotation<strong>al</strong> an<strong>al</strong>yses in the after group may be due to fewer but<br />

he<strong>al</strong>thier TH-containing neurons remaining because the<br />

initi<strong>al</strong> benefici<strong>al</strong> wave <strong>of</strong> microgli<strong>al</strong> response is preserved. A<br />

more in-depth examination <strong>of</strong> the timing <strong>of</strong> minocycline<br />

administration on neuron<strong>al</strong> viability relative to toxin exposure<br />

is currently underway in our laboratory, using markers for cell<br />

death and degeneration.<br />

Previous work demonstrates that inflammation is a key<br />

component <strong>of</strong> the degenerative cascade associated with<br />

Parkinson's disease (McGeer <strong>et</strong> <strong>al</strong>., 2001; Cicch<strong>et</strong>ti <strong>et</strong> <strong>al</strong>.,<br />

2002). Microgli<strong>al</strong> activation, in particular, has been previously<br />

implicated as a key mediator <strong>of</strong> the inflammatory response in<br />

the brain and has been observed in the brains <strong>of</strong> PD patients<br />

(McGeer <strong>et</strong> <strong>al</strong>., 1988). Similar observations have been reported<br />

in PD anim<strong>al</strong> models, such as the 6-OHDA-induced Parkinsonian<br />

rat (Akiyama and McGeer, 1989; He <strong>et</strong> <strong>al</strong>., 1999). Indeed,<br />

we <strong>al</strong>so observe this phenomenon in our 6-OHDA-lesioned<br />

anim<strong>al</strong>s following CD11b immunohistochemistry for microgli<strong>al</strong><br />

status (data not shown). Although the activation <strong>of</strong><br />

resting microglia is a norm<strong>al</strong> event necessary for maintaining<br />

neuron<strong>al</strong> homeostasis, unabated gliosis associated with a<br />

pathologic<strong>al</strong> state such as PD may exacerbate the neurodegenerative<br />

processes <strong>al</strong>ready established (Hirsch <strong>et</strong> <strong>al</strong>., 1998,


204 BRAIN RESEARCH 1093 (<strong>2006</strong>) 198– 207<br />

2003; Stoll <strong>et</strong> <strong>al</strong>., 2002). Because activated microglia are<br />

associated with phagocytosis and the liberation <strong>of</strong> cytotoxic<br />

molecules (e.g., reactive oxygen species) and proinflammatory<br />

cytokines (e.g., tumor necrosis factor α, interleukin-1β,<br />

interferon-γ), it is probable that chronic microgli<strong>al</strong> activation<br />

will significantly contribute to the destruction <strong>of</strong> resident<br />

cells. Because minocycline easily passes through the blood–<br />

brain barrier, it may be indicated as a d<strong>et</strong>errent to the<br />

inflammatory environment observed in diseases like Parkinson's,<br />

Alzheimer's and others.<br />

These data are concordant with previous literature that<br />

supports the therapeutic potenti<strong>al</strong> <strong>of</strong> minocycline against<br />

neurodegenerative events. Our findings suggest that the time<br />

<strong>of</strong> minocycline delivery relative to neuropathogenic event<br />

may d<strong>et</strong>ermine the effectiveness <strong>of</strong> this drug for therapy.<br />

However, recent studies suggest that minocycline treatment<br />

may lead to adverse effects clinic<strong>al</strong>ly, especi<strong>al</strong>ly after longterm<br />

use (Porter and Harrison, 2003; Wolthuis <strong>et</strong> <strong>al</strong>., 2003; Abe<br />

<strong>et</strong> <strong>al</strong>., 2003; Bradfield <strong>et</strong> <strong>al</strong>., 2003). Although these reports<br />

have been generated in the clinic<strong>al</strong> s<strong>et</strong>ting, it is readily<br />

apparent that further, comprehensive ev<strong>al</strong>uations <strong>of</strong> longterm<br />

minocycline use must be pursued before this t<strong>et</strong>racycline<br />

can gain univers<strong>al</strong> acceptance to treat neurodegenerative<br />

diseases.<br />

4. Experiment<strong>al</strong> procedures<br />

4.1. Minocycline protocol<br />

Thirty m<strong>al</strong>e Fisher 344 rats (200 grams) were lesioned with 6-<br />

OHDA (see d<strong>et</strong>ails below) and randomly divided into three<br />

groups (n = 10). One group received daily IP injections <strong>of</strong><br />

45 mg/kg <strong>of</strong> minocycline (ICN Biomedic<strong>al</strong>s, Inc.) beginning 3<br />

days prior to lesioning. Another group received daily IP<br />

injections <strong>of</strong> 45 mg/kg <strong>of</strong> minocycline beginning 24 h after 6-<br />

OHDA lesioning. The remaining control group received daily<br />

IP s<strong>al</strong>ine injections either 3 days before or 24 h after lesioning.<br />

All anim<strong>al</strong>s continued to receive daily injections <strong>of</strong> 45 mg/kg<br />

<strong>of</strong> minocycline or s<strong>al</strong>ine for 1 week following surgic<strong>al</strong><br />

procedures. After this period, <strong>al</strong>l injections were reduced to<br />

every other day for the remainder <strong>of</strong> the 4-week rotation<strong>al</strong><br />

an<strong>al</strong>ysis phase. All experiments were carried out in accordance<br />

with the Nation<strong>al</strong> Institutes <strong>of</strong> He<strong>al</strong>th Guide for the<br />

Care and Use <strong>of</strong> Laboratory Anim<strong>al</strong>s (NIH Publications No. 80-<br />

23; revised 1996).<br />

4.2. 6-Hydroxydopamine lesioning<br />

All anim<strong>al</strong>s were anesth<strong>et</strong>ized with chlor<strong>al</strong> hydrate (400 mg/kg<br />

in 0.9% NaCl, IP) and stereotaxic<strong>al</strong>ly lesioned in the right<br />

medi<strong>al</strong> forebrain bundle with 9 μg/4 μl <strong>of</strong> 6-OHDA in s<strong>al</strong>ine<br />

containing 0.02% ascorbate (coordinates −4.4 mm AP, +1.3 mm<br />

ML, and −7.8 mm DV from the dur<strong>al</strong> surface; see Granholm <strong>et</strong><br />

<strong>al</strong>., 1997). The 6-OHDA neurotoxin was administered over<br />

4 min followed by a 2-min period delay prior to needle<br />

withdraw<strong>al</strong> to <strong>al</strong>low for toxin absorption into the brain<br />

tissue. Anim<strong>al</strong>s were provided with ac<strong>et</strong>aminophen in their<br />

drinking water for 24 h to <strong>al</strong>leviate potenti<strong>al</strong> postsurgic<strong>al</strong><br />

discomfort.<br />

4.3. Rotation<strong>al</strong> ev<strong>al</strong>uation<br />

All anim<strong>al</strong>s included in this study were ev<strong>al</strong>uated for<br />

apomorphine induced rotation<strong>al</strong> behavior beginning 1 week<br />

following 6-OHDA administration and tested once a week<br />

thereafter for a tot<strong>al</strong> <strong>of</strong> 4 weeks. Briefly, anim<strong>al</strong>s were placed in<br />

rotom<strong>et</strong>er bowls (Hudson <strong>et</strong> <strong>al</strong>., 1993) and secured to the<br />

counting sensor by a thoracic harness. Subjects were <strong>al</strong>lowed<br />

to acclimate to the environment for 10 min, then administered<br />

a subcutaneous injection <strong>of</strong> apomorphine (0.05 mg/kg).<br />

Subsequent rotation<strong>al</strong> behavior was then recorded for 1 h.<br />

Following rotation<strong>al</strong> testing, anim<strong>al</strong>s were released from their<br />

harnesses, removed from the rotom<strong>et</strong>er bowls, and r<strong>et</strong>urned<br />

to their cages.<br />

4.4. Immunohistochemistry<br />

After 4 weeks, anim<strong>al</strong>s were deeply anesth<strong>et</strong>ized with<br />

H<strong>al</strong>othane (H<strong>al</strong>ocarbon Laboratories). Rats were then perfused<br />

through the aorta with 0.9% s<strong>al</strong>ine followed by 4% paraform<strong>al</strong>dehyde<br />

and transferred to 30% sucrose in phosphatebuffered<br />

s<strong>al</strong>ine the next day. Sections (45 μm) were obtained<br />

from the nigr<strong>al</strong> region <strong>of</strong> each anim<strong>al</strong> using a sliding<br />

microtome (Microm). Sections were processed for free-floating<br />

immunohistochemistry using antibodies against tyrosine<br />

hydroxylase (1:1000, Pel-Freeze Biologic<strong>al</strong>s). Briefly, the sections<br />

were washed and incubated for 48 h with TH antibodies.<br />

The sections were washed again in Tris-buffered s<strong>al</strong>ine (TBS)<br />

and incubated with the secondary antibody reacted with the<br />

ABC solution (Vectastain, Vector Laboratories) and diaminobenzidine<br />

(Sigma) (DAB, 0.0300 g/100 ml imidazole-ac<strong>et</strong>ate<br />

buffer solution, Fisher Scientific). Sections were mounted on<br />

glass slides and cover slipped. Control sections were processed<br />

without primary or secondary antibodies. Sections<br />

from <strong>al</strong>l groups were processed simultaneously in the same<br />

bath, using mesh-bottom incubation wells, to avoid intersection<br />

staining variability (for further d<strong>et</strong>ails, see Granholm<br />

<strong>et</strong> <strong>al</strong>., 2000).<br />

4.5. Stereologic an<strong>al</strong>ysis<br />

Estimated numbers <strong>of</strong> TH-positive cells in the substantia nigra<br />

<strong>of</strong> <strong>al</strong>l anim<strong>al</strong>s were obtained using the optic<strong>al</strong> fractionator<br />

stereologic<strong>al</strong> m<strong>et</strong>hod (StereoInvestigator, MicroBrightfield,<br />

Inc.) according to previously published protocols (Hunter <strong>et</strong><br />

<strong>al</strong>., 2004a). Briefly, the optic<strong>al</strong> fractionator system consisted <strong>of</strong><br />

a computer-assisted image an<strong>al</strong>ysis system including a Nikon<br />

Eclipse E-600 microscope hard coupled to a Prior H128<br />

computer-controlled x–y–z motorized stage, an Olympus 750<br />

video camera system, a Micron Pentium III computer and the<br />

Stereoinvestigator stereologic<strong>al</strong> s<strong>of</strong>tware (Microbrightfield<br />

Inc.). The region <strong>of</strong> interest was outlined under low magnification<br />

and the number <strong>of</strong> neurons in the outlined region<br />

measured with a systematic random design <strong>of</strong> disector<br />

counting frames. A 60× objective lens with a 1.4 numeric<strong>al</strong><br />

aperture was used to count and measure individu<strong>al</strong> cells<br />

within the counting frames. The counting brick was approximately<br />

20 μm thick, and upper and lower gard zones were<br />

excluded from counting. Means and standard error <strong>of</strong> the<br />

mean (SEM) <strong>of</strong> estimated cell counts were c<strong>al</strong>culated from the


aw data using the stereology s<strong>of</strong>tware system. The tot<strong>al</strong> cell<br />

number for each lesioned nigra was compared to the cell<br />

number <strong>of</strong> the contr<strong>al</strong>ater<strong>al</strong> nonlesioned nigra. These comparisons<br />

were expressed as a percent decrease <strong>of</strong> cell number<br />

in the lesioned nigra compared to the respective nonlesioned<br />

nigra in the same anim<strong>al</strong>.<br />

4.6. Cell size measurements<br />

In order to d<strong>et</strong>ermine if the surviving neurons were he<strong>al</strong>thy<br />

in <strong>al</strong>l groups or had undergone atrophic <strong>al</strong>terations, we<br />

characterized the size <strong>of</strong> lesioned nigr<strong>al</strong> neurons using Scion<br />

Imaging s<strong>of</strong>tware. Fifty neurons from the lesioned side <strong>of</strong><br />

each anim<strong>al</strong> randomly picked from 5 sections for each<br />

anim<strong>al</strong> were measured. Only cells displaying a clearly<br />

defined nucleus and/or at least two neuritic processes were<br />

considered for measurement. The NIH Image s<strong>of</strong>tware<br />

system is written using Think Pasc<strong>al</strong> from Symantec<br />

Corporation. The perim<strong>et</strong>er <strong>of</strong> each neuron was measured<br />

using a sc<strong>al</strong>e c<strong>al</strong>ibration feature. In addition, fiber density<br />

measurements were made <strong>of</strong> the remaining lesioned nigr<strong>al</strong><br />

neurons in <strong>al</strong>l groups using the Scion Imaging s<strong>of</strong>tware<br />

package.<br />

4.7. Statistic<strong>al</strong> an<strong>al</strong>ysis<br />

The apomorphine-induced rotation data were an<strong>al</strong>yzed using<br />

a one-way ANOVA for <strong>al</strong>l 3 groups followed by sever<strong>al</strong> post hoc<br />

two-sample t tests <strong>of</strong> the mean rotations for different group<br />

pairs. We further an<strong>al</strong>yzed time (week) effect and treatment<br />

effects jointly by using a two-way ANOVA followed by a<br />

sever<strong>al</strong> post hoc paired t tests to find which group demonstrated<br />

significant increase in rotation from week 1 to week 4.<br />

In addition, regression an<strong>al</strong>ysis was used to test for linear<br />

trends in rotations over time for each group. For the cell count<br />

results, the mean <strong>of</strong> the difference b<strong>et</strong>ween lesioned and<br />

nonlesioned nigras <strong>of</strong> each anim<strong>al</strong> for each group was<br />

an<strong>al</strong>yzed using a two-sided t test. Descriptive statistics,<br />

including the mean, median and mode, for each group's cell<br />

size and fiber density measurements were obtained. The<br />

an<strong>al</strong>ysis was done using SAS statistic<strong>al</strong> s<strong>of</strong>tware (SAS<br />

Institute, Inc.) and Statview for Windows.<br />

Acknowledgments<br />

This work was made possible by a grant from the Murray<br />

Center for Parkinson's Disease Research and Related Disorders.<br />

REFERENCES<br />

Abe, M., Furukawa, S., Takayama, S., Michitaka, K., Minami, H.,<br />

Yamamoto, K., Horiike, N., Onji, M., 2003. Drug-induced<br />

hepatitis with autoimmune features during minocycline<br />

therapy. Intern<strong>al</strong> Medicine 42, 48–52.<br />

Abramov, A.Y., Canevari, L., Duchen, M.R., 2004. B<strong>et</strong>a-amyloid<br />

peptides induce mitochondri<strong>al</strong> dysfunction and oxidative<br />

stress in astrocytes and death <strong>of</strong> neurons through activation <strong>of</strong><br />

NADPH oxidase. J. Neurosci. 24, 565–575.<br />

BRAIN RESEARCH 1093 (<strong>2006</strong>) 198– 207<br />

205<br />

Akiyama, H., McGeer, P., 1989. Microgli<strong>al</strong> response to 6hydroxydopamine-induced<br />

substantia nigra lesions. Brain Res.<br />

489, 247–253.<br />

Arvin, K.L., Han, B.H., Du, Y., Lin, S.Z., Paul, S.M., Holtzman, D.M.,<br />

2002. Minocycline markedly protects the neonat<strong>al</strong> brain<br />

against hypoxic–ischemic injury. Ann<strong>al</strong>s. Neurol. 52, 54–61.<br />

Barcia, C., Fernandez-Barreiro, A., Poza, M., Herrero, M.T., 2003.<br />

Parkinson's disease and inflammatory changes. Neurotoxicity<br />

Res. 5, 411–418.<br />

Blum, D., Torch, S., Lambeng, N., Nissou, M., Benabid, A.L., Sadoul,<br />

R., Verna, J.M., 2001. Molecular pathways involved in the<br />

neurotoxicity <strong>of</strong> 6-OHDA, dopamine and MPTP: contribution to<br />

the apoptotic theory in Parkinson's disease. Prog. Neurobiol.<br />

65, 135–172.<br />

Bowenkamp, K.E., H<strong>of</strong>fman, A.F., Gerhardt, G.A., Henry, M.A.,<br />

Biddle, P.T., H<strong>of</strong>fer, B.J., Granholm, A.-C., 1995. Gli<strong>al</strong> cell<br />

line-derived neurotrophic factor supports surviv<strong>al</strong> <strong>of</strong> injured<br />

midbrain dopaminergic neurons. J. Comp. Neurol. 355,<br />

479–489.<br />

Bradfield, Y.S., Robertson, D.M., S<strong>al</strong>omao, D.R., Link, T.P., Rostvold,<br />

J.A., 2003. Minocycline-induced ocular pigmentation. Archives<br />

<strong>of</strong> Ophthamology 121, 144–145.<br />

Carman, L.S., Gage, F.H., Shults, C.W., 1991. Parti<strong>al</strong> lesion <strong>of</strong> the<br />

substantia nigra: relation b<strong>et</strong>ween extent <strong>of</strong> lesion and<br />

rotation<strong>al</strong> behavior. Brain Res. 553, 275–283.<br />

Casas, M., Ferre, S., Cobos, A., Cadaf<strong>al</strong>ch, J., Grau, J.M., Jane, F.,<br />

1988. Comparison b<strong>et</strong>ween apomorphine and<br />

amph<strong>et</strong>amine-induced rotation<strong>al</strong> behaviour in rats with a<br />

unilater<strong>al</strong> nigrostriat<strong>al</strong> pathway lesion. Neuropharmacology<br />

27, 657–659.<br />

Chen, M., Ona, V.O., Li, M., Ferrante, R.J., Fink, K.B., Zhu, S., Bian, J.,<br />

Guo, L., Farrell, L.A., Hersch, S.M., Hobbs, W., Vonsattel, J.P.,<br />

Cha, J.H., Friedlander, R.M., 2000. Minocycline inhibits<br />

caspase-1 and caspase-3 expression and delays mort<strong>al</strong>ity in a<br />

transgenic mouse model <strong>of</strong> Huntington's disease. Nat. Med. 6,<br />

797–801.<br />

Choi, W.S., Yoon, S.Y., Oh, T.H., Choi, E.J., O'M<strong>al</strong>ley, K.L., Oh, Y.J.,<br />

1999. Two distinct mechanisms are involved in 6hydroxydopamine-<br />

and MPP-induced dopaminergic neuron<strong>al</strong><br />

cell death: role <strong>of</strong> caspases, ROS, and JNK. J. Neurosci. Res. 57,<br />

86–94.<br />

Cicch<strong>et</strong>ti, F., Brownell, A.L., Williams, K., Chen, Y.I., Livni, E.,<br />

Isacson, O., 2002. Neuroinflammation <strong>of</strong> the nigrostriat<strong>al</strong><br />

pathway during progressive 6-OHDA dopamine degeneration<br />

in rats monitored by immunohistochemistry and PET imaging.<br />

Eur. J. Neurosci. 15, 991–998.<br />

Dawson, T.M., Dawson, V.L., 2003. Molecular pathways <strong>of</strong><br />

neurodegeneration in Parkinson's disease. Science 302,<br />

819–822.<br />

Deumens, R., Blokland, A., Prickaerts, J., 2002. Modeling<br />

Parkinson's disease in rats: an ev<strong>al</strong>uation <strong>of</strong> 6-OHDA lesions <strong>of</strong><br />

the nigrostriat<strong>al</strong> pathway. Exp. Neurol. 175, 303–317.<br />

Domercg, M., Matute, C., 2004. Neuroprotection by t<strong>et</strong>racyclines.<br />

TIPS 25, 609–612.<br />

Du, Y., Ma, Z., Lin, S., Dodel, R.C., Gao, F., B<strong>al</strong>es, K.R., Triarhou, L.C.,<br />

Chern<strong>et</strong>, E., Perry, K.W., Nelson, D.L.G., Luecke, S., Phebus, L.A.,<br />

Bymaster, F.P., Paul, S., 2001. Minocycline prevents<br />

nigrostriat<strong>al</strong> dopaminergic neurodegeneration in the MPTP<br />

model <strong>of</strong> Parkinson's disease. PNAS 98, 14669–14674.<br />

Gemma, C., Catlow, B., Hudson, C., Bickford, P.C., 2003. Knockdown<br />

TNFa with antisense in 6-hydroxydopamine lesioned rats.<br />

Abstr. Am. 410–416.<br />

Gerlach, M., Riederer, P., 1996. Anim<strong>al</strong> models <strong>of</strong> Parkinson's<br />

disease: an empiric<strong>al</strong> comparison with the phenomenology <strong>of</strong><br />

the disease in man. J. Neur<strong>al</strong> Transm. 103, 987–1041.<br />

Glinka, Y., Gassen, M., Youdin, M.B.H., 1997. Mechanism <strong>of</strong><br />

6-hydroxydopamine neurotoxicity. J. Neur<strong>al</strong> Transm. 50, 55–66<br />

(Suppl.).<br />

Granholm, A.-C., Mott, J.L., Eken, S., van Horne, C., Bowenkamp, K.,


206 BRAIN RESEARCH 1093 (<strong>2006</strong>) 198– 207<br />

H<strong>of</strong>fer, B.J., Henry, S., Gerhardt, G.A., 1997. Gli<strong>al</strong> cell<br />

line-derived neurotrophic factor improves surviv<strong>al</strong> <strong>of</strong> ventr<strong>al</strong><br />

mesenceph<strong>al</strong>ic grafts to the 6-OHDA lesioned striatum. Exp.<br />

Brain Res. 116, 29–38.<br />

Granholm, A.-C., Reyland, M., Albeck, D.A., Sanders, L.A., Gerhardt,<br />

G.A., Hoernig, G., Shen, L., Westph<strong>al</strong>, H., H<strong>of</strong>fer, B.J., 2000. Gli<strong>al</strong><br />

cell line-derived neurotrophic factor is needed for postnat<strong>al</strong><br />

surviv<strong>al</strong> <strong>of</strong> midbrain dopaminergic neurons. J. Neurosci. 20,<br />

3182–3190.<br />

He, Y., Lee, T., Leong, S.K., 1999. Time course <strong>of</strong> dopaminergic cell<br />

death and changes in iron, ferritin and transferring levels in<br />

the rat substantia nigra after 6-hydroxydopamine (6-OHDA)<br />

lesioning. Free Radic<strong>al</strong> Res. 31, 103–112.<br />

He, Y., Appel, S., Le, W., 2001. Minocycline inhibits microgli<strong>al</strong><br />

activation and protects nigr<strong>al</strong> cells after 6hydroxydopamine<br />

injection into mouse striatum. Brain Res.<br />

909, 187–193.<br />

Hirsch, E.C., Hunot, S., Damier, P., Faucheux, B., 1998. Gli<strong>al</strong> cells<br />

and inflammation in Parkinson's disease: a role in<br />

neurodegeneration? Ann. Neurol. 44 (Suppl. 1), S115–S120.<br />

Hirsch, E.C., Breidert, T., Roussel<strong>et</strong>, E., Hunot, S., Hartmann, A.,<br />

Michel, P.P., 2003. The role <strong>of</strong> gli<strong>al</strong> reaction and inflammation<br />

in Parkinson's disease. Ann<strong>al</strong>s New York Acad. Sciences 991,<br />

214–228.<br />

Hudson, J.L., van Horne, C.G., Strömberg, I., Brock, S., Clayton, J.,<br />

Masserano, J., H<strong>of</strong>fer, B.J., Gerhardt, G.A., 1993. Correlation <strong>of</strong><br />

apomorphine- and amph<strong>et</strong>amine-induced turning with<br />

nigrostriat<strong>al</strong> dopamine content in unilater<strong>al</strong> 6hydroxydopamine<br />

lesioned rats. Brain Res. 626, 167–174.<br />

Hunot, S., Hirsch, E.C., 2003. Neuroinflammatory processes<br />

in Parkinson's disease. Ann<strong>al</strong>s Neurol. 53 (Suppl. 3),<br />

S49–S58.<br />

Hunter, C.L., Bachman, D., Granholm, A.-C., 2004a. Minocycline<br />

prevents cholinergic loss in a mouse model <strong>of</strong> Down's<br />

syndrome. Ann<strong>al</strong>s <strong>of</strong> Neurology 675–688.<br />

Hunter, C.L., <strong>Quintero</strong>, E.M., Gilstrap, L., Bhat, N.R., Granholm,<br />

A.-C., 2004b. Minocycline protects bas<strong>al</strong> forebrain cholinergic<br />

neurons from mu p75-saporin immunotoxic lesioning. Eur. J.<br />

Neurosci. 19, 3305–3316.<br />

Jenner, P., Olanow, C.W., 1998. Understanding cell death in<br />

Parkinson's disease. Ann. Neurol. 44 (Suppl. 1), S72–S84.<br />

Johnstone, M., Gearing, A.J., Miller, K.M., 1999. A centr<strong>al</strong> role for<br />

astrocytes in the inflammatory response to b<strong>et</strong>a-amyloid;<br />

chemokines, cytokines and reactive oxygen species are<br />

produced. J. Neuroimmunology 93, 182–193.<br />

Leblhuber, F., Neurauter, G., Fuchs, D., 2003. Oxidative damage and<br />

cytogenic an<strong>al</strong>ysis in leukocytes <strong>of</strong> Parkinson's disease<br />

patients. Neurology 60, 729 (Comment).<br />

Lin, S., Wei, X., Xu, Y., Yan, C., Dodel, R., Zhang, Y., Liu, J.,<br />

Klaunig, J.E., Farlow, M., Du, Y., 2003. Minocycline blocks<br />

6-hydroxydopamine-induced neurotoxicity and free radic<strong>al</strong><br />

production in rat cerebellar granule neurons. Life Sci. 72,<br />

1635–1641.<br />

Lotharius, J., Dugan, L.L., O'M<strong>al</strong>ley, K.L., 1999. Distinct mechanisms<br />

underlie neurotoxin-mediated cell death in cultured<br />

dopaminergic neurons. J. Neurosci. 19, 1284–1293.<br />

McGeer, P.L., Itagaki, S., Byes, B.E., McGeer, E.G., 1988. Reactive<br />

microglia are positive for HLA-DR in the substantia nigra <strong>of</strong><br />

Parkinson's and Alzheimer's disease brains. Neurology 38,<br />

1285–1291.<br />

McGeer, P.L., Yasojima, K., McGeer, E.G., 2001. Inflammation in<br />

Parkinson's disease. Adv. Neurol. 86, 83–89.<br />

Mladenovic, A., Perovic, M., Raicevic, N., Kanazir, S., Rakie, L.,<br />

Ruzdijic, S., 2004. 6-hydroxydopamine increases the level <strong>of</strong><br />

TNFα and bax mRNA in the striatum and induces apoptosis <strong>of</strong><br />

dopaminergic neurons in hemi-parkinsonian rats. Brain Res.<br />

996, 237–245.<br />

Mogi, M., Togari, A., Tanaka, K.-L., Ogawa, N., Ichinose, H.,<br />

Nagatsu, T., 1999. Increase in level <strong>of</strong> tumor necrosis factor<br />

(TNF)-α in 6-hydroxydopamine-lesioned striatum in rats<br />

without influence <strong>of</strong> systemic L-DOPA on the TNF-α induction.<br />

Neurosci. L<strong>et</strong>t. 268, 101–104.<br />

Popovic, N., Schubart, A., Go<strong>et</strong>z, B.D., Zhang, S.-C., Linington, C.,<br />

Duncan, I.D., 2001. Inhibition <strong>of</strong> autoimmune<br />

enceph<strong>al</strong>omyelitis by a t<strong>et</strong>racycline. Ann. Neurol. 51,<br />

215–223.<br />

Porter, D., Harrison, A., 2003. Minocycline-induced lupus: a case<br />

series. New Ze<strong>al</strong>and Medic<strong>al</strong> J. U384.<br />

Sauer, H., Oertel, W.H., 1994. Progressive degeneration <strong>of</strong><br />

nigrostriat<strong>al</strong> dopamine neurons following intrastriat<strong>al</strong><br />

termin<strong>al</strong> lesions with 6-hydroxydopamine: a combined<br />

r<strong>et</strong>rograde tracing and immunocytochemic<strong>al</strong> study in the rat.<br />

Neuroscience 59, 401–415.<br />

Schwarting, R.K.W., Huston, J.P., 1996a. Unilater<strong>al</strong> 6hydroxydopamine<br />

lesions <strong>of</strong> meso-striat<strong>al</strong> dopamine<br />

neurons and their physiologic<strong>al</strong> sequelae. Prog. Neurobiol. 49,<br />

215–266.<br />

Schwarting, R.K.W., Huston, J.P., 1996b. The unilater<strong>al</strong><br />

6-hydroxydopamine lesion model in behavior<strong>al</strong> brain research.<br />

An<strong>al</strong>ysis <strong>of</strong> function<strong>al</strong> deficits, recovery and treatments. Prog.<br />

Neurobiol. 50, 275–331.<br />

Stoll, G., Jander, S., Schro<strong>et</strong>er, M., 2002. D<strong>et</strong>riment<strong>al</strong> and benefici<strong>al</strong><br />

effects <strong>of</strong> injury-induced inflammation and cytokine<br />

expression in the nervous system. Advances Exp. Medicine<br />

Biol. 513, 87–113.<br />

Strömberg, I., Gemma, C., Vila, J., Bickford, P.C., 2005.<br />

Blueberry- and spirulina-enriched di<strong>et</strong>s enhance striat<strong>al</strong><br />

dopamine recovery and induce a rapid, transient microglia<br />

activation after injury <strong>of</strong> the rat nigrostriat<strong>al</strong> dopamine<br />

system. Exp. Neurol. 196, 298–307.<br />

Teismann, P., Tieu, K., Cohen, O., Choi, D.K., Wu du, C., Marks, D.,<br />

Vila, M., Jackson-Lewis, V., Przedborski, S., 2003. Pathogenic<br />

role <strong>of</strong> gli<strong>al</strong> cells in Parkinson's disease. Mov. Disord. 18,<br />

121–129.<br />

Tikka, T., Fiebich, B., Goldsteins, G., Keinanen, R., Koistinaho, J.,<br />

2001. Minocycline, a t<strong>et</strong>racycline derivative, is neuroprotective<br />

against excitotoxicity by inhibiting activation and proliferation<br />

<strong>of</strong> microglia. J. Neurosci. 21, 2580–2588.<br />

Ungerstedt, U., 1968. 6-Hydroxy-dopamine induced degeneration<br />

<strong>of</strong> centr<strong>al</strong> monoamine neurons. Eur. J. Pharmacol. 5, 107–110.<br />

Ungerstedt, U., Arbuthnott, G.W., 1970. Quantitation<strong>al</strong> recording<br />

<strong>of</strong> rotation<strong>al</strong> behavior in rats after 6-hydroxy-dopamine<br />

lesions <strong>of</strong> the nigrostriat<strong>al</strong> dopamine system. Brain Res. 24,<br />

485–493.<br />

V<strong>al</strong>lat-Decouvelaere, A.V., Chr<strong>et</strong>ien, F., Gras, G., Le Pavec, G.,<br />

Dormont, D., Gray, F., 2003. Expression <strong>of</strong> amino acid<br />

transporter-1 in brain macrophages and microglia <strong>of</strong><br />

HIV-infected patients. A neuroprotective role for activated<br />

microglia? J. Neuropathol. and Exp. Neurol. 62, 475–485.<br />

Wang, X., Zhu, S., Drozda, M., <strong>et</strong> <strong>al</strong>., 2003. Minocycline inhibits<br />

caspase-independent and dependent mitochondri<strong>al</strong> cell death<br />

pathways in models <strong>of</strong> Huntington's disease. Proc. Natl. Acad.<br />

Sci. U. S. A. 100, 10483–10487.<br />

Wolthuis, A., Nackaerts, K., Guffens, P., Jousten, P., Demedts, M.,<br />

2003. Minocycline-induced pulmonary infiltration and<br />

eosinophilia. Acta Clinica 50–53.<br />

Wu, D.C., Jackson-Lewis, V., Vila, M., Tieu, K., Teismann,<br />

P., Vads<strong>et</strong>h, C., Choi, D.-K., Ischiropoulos, H., Przedborski,<br />

S., 2002. Blockade <strong>of</strong> microgli<strong>al</strong> activation is<br />

neuroprotective in the 1-m<strong>et</strong>hyl-4-phenyl-1,2,3,6t<strong>et</strong>rahydropyridine<br />

mouse model <strong>of</strong> Parkinson disease.<br />

J. Neurosci. 22, 1763–1771.<br />

Yrjänheikki, J., Keinanen, R., Pellikka, M., Hokfelt, T., Koistinaho, J.,<br />

1998. T<strong>et</strong>racyclines inhibit microgli<strong>al</strong> activation and are<br />

neuroprotective in glob<strong>al</strong> brain ischemia. Proc. Natl. Acad. Sci.<br />

U. S. A. 95, 15769–15774.<br />

Yrjänheikki, J., Tikka, T., Keinänen, R., Goldsteins, G., Chan, P.H.,<br />

Koistinaho, J., 1999. A t<strong>et</strong>racycline derivative, minocycline,


educes inflammation and protects against foc<strong>al</strong> brain<br />

ischemia with a wide therapeutic window. PNAS 96,<br />

13496–13500.<br />

Zhanel, G.C., Homenuik, K., Nichol, K., Noreddin, A., Vercaigne, L.,<br />

Embil, J., Gin, A., Karlowsky, J.A., Hoban, D.J., 2004. The<br />

glycylcyclines: a comparative review with the t<strong>et</strong>racyclines.<br />

Drugs 64, 63–88.<br />

Zhu, S., Stavrovskaya, I.G., Drozda, M., Kim, B.Y.S., Ona, V., Li, M.,<br />

Sarang, S., Liu, A.S., Hartley, D.M., Wu, D.C., Gullans, S.,<br />

BRAIN RESEARCH 1093 (<strong>2006</strong>) 198– 207<br />

207<br />

Ferrante, R.J., Przedborski, S., Krist<strong>al</strong>, B.S., Friedlander, R.M.,<br />

2002. Minocycline inhibits cytochrome c release and delays<br />

progression <strong>of</strong> amyotrophic later<strong>al</strong> sclerosis in mice. Nature<br />

417, 74–78.<br />

Zuch, C.L., Nordstroem, V.K., Briedrick, L.A., Hoernig, G.R.,<br />

Granholm, A.-C., Bickford, P.C., 2000. Time course <strong>of</strong><br />

degenerative <strong>al</strong>terations in nigr<strong>al</strong> dopaminergic neurons<br />

following a 6-hydroxydopamine lesion. J. Comp. Neurol. 427,<br />

440–454.

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

Saved successfully!

Ooh no, something went wrong!