ischemic injury author manuscript nih public access c ... · (sigma) at 37°c for 20 minutes. after...

17
Methazolamide and Melatonin Inhibit Mitochondrial Cytochrome C Release and Are Neuroprotective in Experimental Models of Ischemic Injury Xin Wang, PhD, Bryan E. Figueroa, MD, Irina G. Stavrovskaya, PhD, Yi Zhang, PhD, Ana C. Sirianni, MS, Shan Zhu, PhD, Arthur L. Day, MD, Bruce S. Kristal, PhD, and Robert M. Friedlander, MD, MA Neuroapoptosis Laboratory (X.W., B.E.F., Y.Z., A.C.S., S.Z., R.M.F.), Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Mass; Department of Neurosurgery (I.G.S., A.L.D., B.S.K.), Brigham and Women’s Hospital, Harvard Medical School, Boston, Mass. Abstract Background and Purpose—The identification of a neuroprotective drug for stroke remains elusive. Given that mitochondria play a key role both in maintaining cellular energetic homeostasis and in triggering the activation of cell death pathways, we evaluated the efficacy of newly identified inhibitors of cytochrome c release in hypoxia/ischemia induced cell death. We demonstrate that methazolamide and melatonin are protective in cellular and in vivo models of neuronal hypoxia. Methods—The effects of methazolamide and melatonin were tested in oxygen/glucose deprivation —induced death of primary cerebrocortical neurons. Mitochondrial membrane potential, release of apoptogenic mitochondrial factors, pro—IL-1β processing, and activation of caspase -1 and -3 were evaluated. Methazolamide and melatonin were also studied in a middle cerebral artery occlusion mouse model. Infarct volume, neurological function, and biochemical events were examined in the absence or presence of the 2 drugs. Results—Methazolamide and melatonin inhibit oxygen/glucose deprivation—induced cell death, loss of mitochondrial membrane potential, release of mitochondrial factors, pro—IL-1β processing, and activation of caspase-1 and -3 in primary cerebrocortical neurons. Furthermore, they decrease infarct size and improve neurological scores after middle cerebral artery occlusion in mice. Conclusions—We demonstrate that methazolamide and melatonin are neuroprotective against cerebral ischemia and provide evidence of the effectiveness of a mitochondrial-based drug screen in identifying neuroprotective drugs. Given the proven human safety of melatonin and methazolamide, and their ability to cross the blood-brain-barrier, these drugs are attractive as potential novel therapies for ischemic injury. Keywords methazolamide; melatonin; neuroprotection; ischemic stroke Release of cytochrome c from the mitochondria triggers a set of events leading to the activation of programmed cell death. 1,2 Cytochrome c release occurs in a variety of neurological diseases, Correspondence to Robert M. Friedlander, MD, MA, Brigham and Women’s Hospital, Harvard Medical School, LMRC 111-113, Department of Neurosurgery, Boston, MA 02115. E-mail E-mail: [email protected]. Disclosures None. NIH Public Access Author Manuscript Stroke. Author manuscript; available in PMC 2009 May 1. Published in final edited form as: Stroke. 2009 May ; 40(5): 1877–1885. doi:10.1161/STROKEAHA.108.540765. NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript

Upload: others

Post on 15-Jul-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Ischemic Injury Author Manuscript NIH Public Access C ... · (Sigma) at 37°C for 20 minutes. After staining, each slice was scanned with an HP scanjet 4200C. The stained and unstained

Methazolamide and Melatonin Inhibit Mitochondrial CytochromeC Release and Are Neuroprotective in Experimental Models ofIschemic Injury

Xin Wang, PhD, Bryan E. Figueroa, MD, Irina G. Stavrovskaya, PhD, Yi Zhang, PhD, Ana C.Sirianni, MS, Shan Zhu, PhD, Arthur L. Day, MD, Bruce S. Kristal, PhD, and Robert M.Friedlander, MD, MANeuroapoptosis Laboratory (X.W., B.E.F., Y.Z., A.C.S., S.Z., R.M.F.), Department of Neurosurgery,Brigham and Women’s Hospital, Harvard Medical School, Boston, Mass; Department ofNeurosurgery (I.G.S., A.L.D., B.S.K.), Brigham and Women’s Hospital, Harvard Medical School,Boston, Mass.

AbstractBackground and Purpose—The identification of a neuroprotective drug for stroke remainselusive. Given that mitochondria play a key role both in maintaining cellular energetic homeostasisand in triggering the activation of cell death pathways, we evaluated the efficacy of newly identifiedinhibitors of cytochrome c release in hypoxia/ischemia induced cell death. We demonstrate thatmethazolamide and melatonin are protective in cellular and in vivo models of neuronal hypoxia.

Methods—The effects of methazolamide and melatonin were tested in oxygen/glucose deprivation—induced death of primary cerebrocortical neurons. Mitochondrial membrane potential, release ofapoptogenic mitochondrial factors, pro—IL-1β processing, and activation of caspase -1 and -3 wereevaluated. Methazolamide and melatonin were also studied in a middle cerebral artery occlusionmouse model. Infarct volume, neurological function, and biochemical events were examined in theabsence or presence of the 2 drugs.

Results—Methazolamide and melatonin inhibit oxygen/glucose deprivation—induced cell death,loss of mitochondrial membrane potential, release of mitochondrial factors, pro—IL-1β processing,and activation of caspase-1 and -3 in primary cerebrocortical neurons. Furthermore, they decreaseinfarct size and improve neurological scores after middle cerebral artery occlusion in mice.

Conclusions—We demonstrate that methazolamide and melatonin are neuroprotective againstcerebral ischemia and provide evidence of the effectiveness of a mitochondrial-based drug screen inidentifying neuroprotective drugs. Given the proven human safety of melatonin and methazolamide,and their ability to cross the blood-brain-barrier, these drugs are attractive as potential novel therapiesfor ischemic injury.

Keywordsmethazolamide; melatonin; neuroprotection; ischemic stroke

Release of cytochrome c from the mitochondria triggers a set of events leading to the activationof programmed cell death.1,2 Cytochrome c release occurs in a variety of neurological diseases,

Correspondence to Robert M. Friedlander, MD, MA, Brigham and Women’s Hospital, Harvard Medical School, LMRC 111-113,Department of Neurosurgery, Boston, MA 02115. E-mail E-mail: [email protected] None.

NIH Public AccessAuthor ManuscriptStroke. Author manuscript; available in PMC 2009 May 1.

Published in final edited form as:Stroke. 2009 May ; 40(5): 1877–1885. doi:10.1161/STROKEAHA.108.540765.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 2: Ischemic Injury Author Manuscript NIH Public Access C ... · (Sigma) at 37°C for 20 minutes. After staining, each slice was scanned with an HP scanjet 4200C. The stained and unstained

including stroke, Huntington disease, and amyotrophic lateral sclerosis.3–5 Inhibition ofcytochrome c is therefore a potential therapeutic target for neuroprotection. To identify novelneuroprotective agents, we screened a library of FDA-approved drugs for inhibition ofcytochrome c release. Cytochrome c release can be induced in purified mitochondria, providinga screening method for the identification of compounds which could inhibit this process.

We identified a number of drugs that inhibit cytochrome c release in purified mitochondria aswell as in mutant Huntington-expressing striatal cells.6 One of these drugs, methazolamide, isneuroprotective in vivo in a transgenic mouse model of Huntington disease.6 To determinewhether this screen identified protective drugs for acute neurodegenerative models, we tested2 drugs in an experimental model of cerebral ischemia. Here we provide evidence for theneuroprotective properties of 2 of the drugs identified in the screen, methazolamide andmelatonin, both in primary cerebrocortical neurons (PCNs) exposed to several cell deathstimuli, as well as in vivo in cerebral ischemia. Neuroprotection in vivo is associated withinhibition of cytochrome c release and of caspase-3 activation. This report provides evidenceof the efficacy of a mitochondrial-based screen in identifying drugs that are protective in anacute model of neurodegeneration and provides further support for the importance ofcytochrome c release in the pathogenesis of ischemic injury.

Materials and MethodsDrugs

Methazolamide and melatonin were obtained from Sigma.

Cell Lines and Induction of Cell DeathCulture of PCNs were isolated as previously described and subjected to oxygen-glucosedeprivation (OGD),4 1 mmol/L H2O2

,7 or 500 μmol/L NMDA7 for 18 hours. All cultures wereused between days 7 to 10 after harvest. Cell death was determined by lactate dehydrogenaseassay. Morphology of PCN cells was observed, chromatin condensation and nuclearfragmentation were analyzed with Hoechst 33342 staining (Molecular Probes; 1:5000 for 2 to5 minutes).

Lactate Dehydrogenase AssayThe assay was performed as previously described according to the manufacturer’s instructions(Roche).4

Terminal dUTP Nick-End Labeling AssayThe assay was performed using the DeadEnd Fluorometric terminal dUTP nick-end labeling(TUNEL) system (Promega) as specified by the manufacturer. Briefly, attached PCNs werefixed with 4% methanol-free formaldehyde, permeabilized by 0.2% Triton-X-100, andincubated with the TUNEL reaction mixture for 1 hour at 37°C. After thorough washes,chromatin condensation and nuclear fragmentation were analyzed using a Nikon ECLIPSETE-200 fluorescence microscope.

Western BlotMouse brain samples were lysed in RIPA buffer with protease inhibitors.5,8 Antibody tocaspase-3 was purchased from Cell Signaling Technology, antibody to caspase-1 from SantaCruz Biotechnology, and antibody to β-actin from Sigma.

Wang et al. Page 2

Stroke. Author manuscript; available in PMC 2009 May 1.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 3: Ischemic Injury Author Manuscript NIH Public Access C ... · (Sigma) at 37°C for 20 minutes. After staining, each slice was scanned with an HP scanjet 4200C. The stained and unstained

Cellular and Tissue FractionationPCNs and mouse brain cytosolic fractionations were performed as described.7 Releasedcytochrome c or AIF was analyzed by Western blot. Antibody to cytochrome c was purchasedfrom PharMingen, and to AIF from Sigma.

Mature IL-1β DeterminationMature IL-1β quantification was performed as previously described4 by using an ELISA kitspecific for the mature form of the cytokine (R&D Systems).

Caspase-1 Activity AssayCell extracts and enzyme assays were performed with the ApoAlert caspase fluorescent assaykit as previously described. Caspase-1-like substrate Ac-YVAD-AFC (7-amino-4-trifluoromethylcoumarin) was purchased from Calbiochem. Released AFC was quantified ina Bio-Rad Versa Fluorometer (excitation at 400 nm and emission at 505 nm).

Determination of Mitochondrial Transmembrane Potential (ΔΨm)PCNs were treated as indicated with or without methazolamide, or melatonin. Living cells werestained with Rhodamine 123 (Rh 123, Molecular Probes) as previously described.5,6

Measurement of Mitochondrial Permeability TransitionRat liver mitochondria were isolated and treated as previously described.6,9

Permanent Middle Cerebral Artery OcclusionFocal cerebral ischemia was produced as described previously.4,7 Briefly, anesthesia wasinduced in 6- to 8-week-old C57BL/6J mice (body weight 20 to 24 g, Jackson Laboratory, BarHarbor, Maine) with 2% (vol/vol) isoflurane (70% N2O/30% O2) and was maintained with a1 to 0.5% concentration. Rectal temperature was maintained between 37.0 and 37.5°C with aheating pad (Harvard Apparatus). Focal cerebral ischemia was induced by an intraluminal 7-0nylon thread with a silicone tip (180 μm diameter) introduced into the right cervical internalcarotid artery. The thread was inserted 9±1.0 mm into the internal carotid artery up to the middlecerebral artery. A laser Doppler perfusion monitor (Perimed AB) was adhered to the righttemporal aspect of the animal’s skull and used to confirm middle cerebral artery flowdisruption. Control animals underwent sham surgery consisting of only anesthesia and carotidartery dissection. Each animal was euthanized after surgery. The brain was removed, and thecerebral hemisphere was retained for either evaluation of infract volume or Western blotanalysis.

Drug Treatment in Middle Cerebral Artery Occlusion ModelControl mice were treated with a normal saline vehicle control containing 0.4% DMSO and3% Tween-20. Methazolamide and melatonin were respectively prepared in normal salinecontaining 0.4% DMSO or 3% Tween-20 and administered by intraperitoneal injection at adose of 20 mg/kg for methazolamide and 10 mg/kg for melatonin. Mice were treated either 1hour before 30 minutes after permanent middle cerebral artery occlusion (MCAO).

Determination of Infarct VolumeAfter 24 hours of MCAO, the mice were euthanized. Brains were rapidly removed and chilledfor 2 minutes. Coronal sections (1-mm thick; n=7) were cut with a mouse brain matrix. Eachslice was immersed in saline solution containing 2% 2,3,5-triphenyltetrazolium chloride(Sigma) at 37°C for 20 minutes. After staining, each slice was scanned with an HP scanjet4200C. The stained and unstained areas of the right hemisphere were quantified with imageJ

Wang et al. Page 3

Stroke. Author manuscript; available in PMC 2009 May 1.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 4: Ischemic Injury Author Manuscript NIH Public Access C ... · (Sigma) at 37°C for 20 minutes. After staining, each slice was scanned with an HP scanjet 4200C. The stained and unstained

1.32j (NIH), and these values were used to calculate the infarct volume expressed as apercentage of the lesioned hemisphere.

Neurobehavioral ExaminationMice were assigned a neurobehavioral score at 24 hours after MCAO. A previously reportedscale was used.7,10

Statistical AnalysisDensitometric quantification was performed with the Quantity One Program (Bio-Rad).Statistical significance was evaluated by t test: *probability values <0.05 were consideredsignificant; **probability values <0.001. Drug analysis, including IC50 and maximumprotection, were performed using the GraphPad Prism program.

ResultsMethazolamide and Melatonin Are Neuroprotective in Oxygen Glucose Deprivation—Mediated Primary Cerebrocortical Neuronal Death

Using a purified mitochondrial screen, we identified drugs that inhibit release of cytochromec.6 To determine whether this screen is effective at identifying drugs that are neuroprotectivein acute cell death, we evaluated two of the “hits,” methazolamide and melatonin, in an in vitromodel of cerebral ischemia. Exposing PCNs to OGD induces cytochrome c release andcaspase-3 activation, ultimately leading to cell death.4,11 Incubation with eithermethazolamide (100 nmol/L to 100 μmol/L) or melatonin (1 μmol/L to 100 μmol/L) resultedin statistically significant inhibition of OGD-mediated PCN cell death (Figure 1A and 1C). Todetermine the relative potencies of the 2 drugs, we measured the extent of cell death as afunction of drug concentration. The resulting curves (plotted semilogarithmically) define theIC50 and maximum protection afforded by methazolamide (250 nmol/L and 53.45%,respectively, in Figure 1B) and melatonin (490 nmol/L and 40.59%, respectively, in Figure1D).

Methazolamide and Melatonin Protect PCNs From a Variety of Cell Death InducersAs described above, methazolamide and melatonin inhibit OGD-induced PCN cell death. Wenext evaluated whether methazolamide- or melatonin-mediated neuroprotection could beextended to additional cell death paradigms. H2O2- and NMDA-induced cell death have beenused as in vitro models of oxidative damage and excitotoxic neuronal injury.7,8,12,13 Wetherefore evaluated the ability of methazol-amide and melatonin to protect PCNs challengedwith H2O2 or NMDA. Melatonin and methazolamide inhibited both H2O2 and NMDA-inducedPCN cell death in a dose dependant manner (Figure 2A, 2B, 2D, and 2E). Phase-contrastphotomicrographs demonstrate H2O2-mediated loss of PCN neuritic processes (Figure 2G,lower panel), cell death-associated nuclear fragmentation, and chromatin condensation (arrowsin Figure 2G, upper panel). Methazolamide (Figure 2A and 2B) and melatonin (Figure 2D and2E) significantly inhibited cell death induced by either stimulus. Phase-contrast micrographdemonstrates that both methazolamide and melatonin not only inhibited cell death, but alsopartially restored normal cellular morphology. Note that Figure 2 also includes data on thecytochrome c release inhibitor minocycline (Figure 2C and 2F). Minocycline is used here asa positive control, given its demonstrated neuroprotective properties as an inhibitor ofcytochrome c release.8,14,15 To provide further evidence of modulation of cell death, weperformed TUNEL labeling and evaluated the neurons using a fluorescence microscope (Figure2H). TUNEL staining revealed a clear increase of chromatin condensation and fragmentationin H2O2-treated apoptotic nuclei of PCNs as compared to control cells. Incubation ofmethazolamide and melatonin reduced the degree of TUNEL-positive cells. Having observed

Wang et al. Page 4

Stroke. Author manuscript; available in PMC 2009 May 1.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 5: Ischemic Injury Author Manuscript NIH Public Access C ... · (Sigma) at 37°C for 20 minutes. After staining, each slice was scanned with an HP scanjet 4200C. The stained and unstained

that methazolamide and melatonin prolong the survival of PCNs subjected to OGD, H2O2, andNMDA, we proceeded to study what cell death–associated molecular changes they affect.

Inhibition of Mitochondrial Cell Death Pathways Contributes to Neuroprotection byMethazolamide and Melatonin

Our observation that methazolamide is neuroprotective in PCNs is the first in the literature.Now it becomes of interest to determine the effects of the compound on the molecularphysiology of the cell. The ability of melatonin to inhibit neuronal cell death has beenpreviously demonstrated,16–21 but the signaling mechanisms mediating the neuroprotectiveactions of melatonin remain poorly understood. Given that methazolamide and melatonin wereidentified by their ability to inhibit cytochrome c release from purified mitochondria, we nextevaluated whether these drugs could inhibit OGD-mediated cytochrome c release andcaspase-3 activation.

The release of cytochrome c and apoptosis-inducing factor (AIF) from mitochondria triggerdownstream caspase-3 activation and additional caspase-independent cell death events.1,22Blocking the release of apoptogenic factors into the cytoplasm should inhibit cell death. Indeed,as demonstrated by Western blotting, OGD induces mitochondrial release of cytochrome c andAIF (Figure 3A and 3B). Methazolamide (Figure 3A) and melatonin (Figure 3B) effectivelyinhibited release of these cell death mediators. We next evaluated the activity of thedownstream effector caspase-3. Western blot analysis demonstrated that caspase-3 wasactivated on OGD induction, and both methazolamide and melatonin effectively inhibitedcaspase-3 activation (Figure 3C).

Methazolamide and Melatonin Slow Dissipation of the Mitochondrial Membrane PotentialGradient in Primary Cerebrocortical Neurons

Proper mitochondrial membrane potential (Δψm) is critical for appropriate cellularbioenergetic homeostasis, and its loss is an important event associated with progression ofmitochondrial dysfunction and leading to cellular demise.23 We therefore evaluated whether,given their neuroprotective properties, methazolamide and melatonin could inhibit thedissipation of Δψm. Comparing with the punctate Rh123 staining seen in normal Δψm incontrol PCNs, OGD results in a diffuse and lower intensity staining pattern as previouslydescribed4 (Figure 4A, upper panel). Both methazolamide and melatonin inhibited OGD-associated loss of mitochondrial Δψm (Figure 4A, lower panel). Hence methazolamide andmelatonin treated PCNs exposed to OGD maintain proper Δψm, therefore pointing to amechanism of action involving not only inhibition of release of apoptogenic factors, but alsopreservation of appropriate cellular energetics.

Melatonin Is Not a Mitochondrial Permeability Transition Inhibitor in Isolated MitochondriaThe mitochondrial permeability transition pore consists of a multimeric complex of proteinsspanning the inner and outer membranes.24,25 Its opening is a component of mitochondrialdysfunction, itself a result of such biochemical stresses as high concentrations of Ca2+ ion,oxidizing agents, thiol reactive compounds (ie, reactive aldehyde), and proapoptotic cytosolicproteins.26 The loss of ΔΨm is an early event in the cell death pathway and an important triggerof the caspase cascade.

There is no satisfactory method for directly determining the mitochondrial permeabilitytransition in living cells. Consequently, we investigated the effects of methazolamide on thismolecular event in purified mitochondria. In these experiments, mitochondrial permeabilitytransition (mPT) was induced by Ca2+ ions, the oxidating agent organic hydroperoxide tBH,the thiol cross-linking agent PhAsO, the thiol oxidant diamide, or a combination thereof (Figure4B, and 4C). Melatonin, similar to methazolamide,6 did not protect purified mitochondria from

Wang et al. Page 5

Stroke. Author manuscript; available in PMC 2009 May 1.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 6: Ischemic Injury Author Manuscript NIH Public Access C ... · (Sigma) at 37°C for 20 minutes. After staining, each slice was scanned with an HP scanjet 4200C. The stained and unstained

these chemical challenges. These observations contrast with the analogous ones using (1)minocycline, a compound that blocks the loss of the mitochondrial membrane potential bothin vitro and in vivo,5,8 or (2) the heterocyclic, tricyclic, or phenothiazine-derived agents.26

Neither inhibition of the mPT nor direct effects on mitochondrial physiology appear to underliethe protection imparted by melatonin. Initial studies in isolated liver mitochondrial focused onmonitoring swelling as a marker or mPT induction and tested the agents at a broad doseresponse range. No effects were seen, suggesting that these agents do not interfere with mPTinduction across a broad concentration range. Because work in other systems, such as culturedcerebellar granular neurons, had suggested that interference with mitochondrial physiologythrough uncoupling and respiratory inhibition could also appear protective, we examined theeffects of melatonin on basic physiological properties. Studies were done on a recently adaptedfluorescence system9 that allows simultaneous measurement of membrane potential (byfollowing TMRM fluorescence), Ca2+ flux (using Calcium Green 5N), NAD+/NADH redoxstatus (using autofluorescence of the NAD+/NADH couple), and swelling (via light scatter).The system is an improved fluorescence-based analog of the electrode-based system we havepreviously used and described.26

Melatonin did not have any significant effects on ΔΨm, calcium transport, NAD+/NADH redoxstatus, or swelling (Figure 4B). Thus, the data in Figure 3B and 3C suggest that melatonin doesnot mediate its protective effect by acting on basic mitochondrial physiology, includingmitochondrial redox status and substrate, proton, electron, or Ca2+ transport. These data aremost consistent with effects mediated outside the inner mitochondrial membrane/mitochondrial matrix, including potential targets such as bcl-2 family members (eg, bak/bax)and the elements involved in release of proapoptotic proteins. Studies are in progress to addressthese potential mechanisms.

Melatonin and Methazolamide Inhibit OGD-Induced Mature IL-1β Release and Caspase-1Activation

Caspase-1 plays a critical role as an apical activator of OGD-induced PCN death.4 The releaseof mature IL-1β serves as an indicator of caspase-1 activation.27 Endogenously producedIL-1β plays a role in a variety of cell death paradigms, and the inhibition of cleavage of pro-IL-1β and the secretion of mature IL-1β are associated with inhibition of cell death.4,27–29To further evaluate the mechanisms of action of methazolamide and melatonin, using an ELISAassay specific for the mature form of IL-1β, we quantified the release of the active cytokineinto conditioned medium of OGD-treated PCNs. Consistent with previous evidence that OGDtreatment induces caspase-1 activation and IL-1β processing in PCNs,4 we detected a greaterthan 4-fold increase in the concentration of mature IL-1β (Figure 5A and 5B) and caspase-1cleavage (p20; Figure 5C). Moreover, the release of mature IL-1β and activation of caspase-1were significantly blocked by methazolamide (Figure 5A and 5C) and by melatonin (Figure5B and 5C). In parallel, caspase-1 activity was measured by the hydrolysis of fluorogenictetrapeptide substrate. Again, treatment of PCNs with either methazolamide or melatonininhibited caspase-1 activation (Figure 5D). Taken together, our findings provide for the firsttime evidence demonstrating methazolamide- and melatonin-mediated inhibition of OGD-induced neuronal caspase-1 activation and mature IL-1β release.

Methazolamide and Melatonin Decrease Cerebral Ischemia-Induced InjuryJust as it rescued cultured neurons from a variety of cell death stimuli, minocycline decreasesthe volume of the infarct produced by focal ischemia.30 We found that methazolamide andmelatonin, like minocycline, forestall cell death attributable to the same stimuli H2O2 orNMDA, suggesting that they may be protective in cerebral ischemia. Indeed, methazolamideand melatonin-treated mice had significantly smaller infarcts after MCAO than mice injected

Wang et al. Page 6

Stroke. Author manuscript; available in PMC 2009 May 1.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 7: Ischemic Injury Author Manuscript NIH Public Access C ... · (Sigma) at 37°C for 20 minutes. After staining, each slice was scanned with an HP scanjet 4200C. The stained and unstained

with the vehicle (Figure 6A and 6B). Associated with a smaller infract, their postischemicbehavior was less severely impaired (Figure 6A and 6C). The drugs were effective ifadministered either 1 hour before or 30 minutes after the onset of ischemia. We note that eventhough pretreatment and posttreatment resulted in similar protection as evaluated by TTC,pretreatment resulted in a greater degree of protection as assessed by behavior (Figure 6B and6C). Considering that TTC is a gross marker of territorial infarction and does not distinguishfunctional versus dysfunctional brain that is alive, thus it is possible that even though the sizeof the injury is the same based on TTC criteria, there is a difference in the cerebral territorythat is dysfunctional, but not dead depending whether the mice were pre or post treated withmelatonin. These observations constitute the first report that methazolamide offersneuroprotection in an animal model of cerebral ischemia. They also confirm that melatonin isbeneficial in animal models of cerebral ischemia, this time in mice rather than in rats.31,32

Like mitochondrial dysfunction, the resulting caspase-mediated cell death pathways areimportant in a broad spectrum of acute and chronic neurodegenerative diseases.2,4,5,15 Inparticular, cytochrome c release and caspase-3 activation are observed in brains of mice thathave recently suffered ischemic damage.15,33,34 Because methazolamide and melatonin areneuroprotective in animal models of stroke, we evaluated whether they diminish cytochromec release and caspase-3 activation stimulated by acute insult. As observed in cultured cellschallenged with OGD, methazolamide and melatonin reduced cytochrome c release andcaspase-3 activation in ischemic tissue in vivo (Figure 6D and 6E).

DiscussionConsequences of cerebral ischemia result in significant human morbidity and mortality.Despite great efforts to identify protective drugs for cerebral ischemia, discovery of an effectiveagent remains elusive. Failure might be in part the result of selection of inappropriate targetsor the inappropriate executions and expectations of clinical trials. Given the prominent role ofmitochondria both in generating energy required for cell survival and in triggering cell deathpathways, we hypothesize that identifying drugs that target mitochondrial pathways may bebroadly neuroprotective. Using an isolated/cell-free mitochondrial screen, we have identifieda set of drugs that inhibit cytochrome c release in the isolated organelle.6 We demonstrate thatmost but not all the cytochrome c release inhibitors are protective in a number of neuronal celldeath models. The 2 drugs selected for further evaluation, methazolamide and melatonin, notonly inhibited cytochrome c release and cell death, but also inhibited the loss of mitochondrialmembrane potential, caspase-3 activation, caspase-1 activation, and IL-1β processingassociated with exposure to a cell death stimulus.

Drug-mediated preservation of membrane potential under circumstances that otherwise wouldlead to cellular demise point to the ability of these agents to protect/enhance mithochondrialfunction. Therefore the drugs not only inhibit the release of mitochondrial cytochrome c/AIFwith the subsequent activation of cell death pathways, but concomitantly improvemitochondrial function and as a result cellular health. Given that AIF is an important mediatorof caspase-independent cell death pathways, our observation with methazolamide andmelatonin inhibiting AIF release represents the first report demonstrating the ability to inhibitboth caspase-dependent (cytochrome c) and caspase-independent (AIF) mitochondrial celldeath pathways.

There are at least 2 major pathways for cytochrome c release in both in vitro and in vivo. Theseare the mPT and the proapoptotic pathways induced by Bcl-2 family members such as Bid/Bax/Bak/bad, etc. Given that these drugs do not act at the mPT, it is most likely that they actby a bcl-2 family—modulated pathway. Further refinement of the molecular target is a goal ofongoing and future research.

Wang et al. Page 7

Stroke. Author manuscript; available in PMC 2009 May 1.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 8: Ischemic Injury Author Manuscript NIH Public Access C ... · (Sigma) at 37°C for 20 minutes. After staining, each slice was scanned with an HP scanjet 4200C. The stained and unstained

We demonstrate that these 2 drugs are protective in an in vivo experimental model of cerebralischemia. This is the first demonstration that the carbonic anhydrase inhibitor methazolamideis neuroprotective in cerebral ischemia. The exogenous administration of methazolamide inthe experimental stroke model reduced infarct volume, improved the neurological score, andlowered cytochrome c release and caspase-3 activation. Even though melatonin has beenpreviously universally demonstrated to be neuroprotective in experimental models of cerebralischemia in several species including mice,17 adult18 and neonatal rats,19 gerbils,20 and cats,21 the mechanisms of action are not known understood. Hence we extend this finding bydemonstrating not only efficacy in a mouse model of cerebral ischemia but also inhibition ofcytochrome c release as a target of melatonin-mediated neuroprotection. An additional newfinding is the demonstration that melatonin inhibits hypoxia-induced caspase-1 activation.From a mechanistic and drug development standpoint, we demonstrate that a mitochondrial-based drug screen can identify effective neuroprotective drugs. Therefore, this work providesevidence of the efficacy of a mitochondrial screen in identifying drugs with neuroprotectiveproperties. Given that the drugs were identified as inhibitors of cytochrome c release, this workalso provides further support for the key role of this biological event in neurodegeneration. Atthe physiological level, it validates targeting the molecular process as a rational approach tothe design of therapies for cerebral ischemia. We demonstrate that the mitochondrial screen isefficacious at selecting protective drugs not only for acute but also for chronicneurodegeneration. It is important to note that these drugs are safe, have been in human usefor many years, and cross the blood— brain barrier, making them attractive agents for furtherhuman evaluation.

AcknowledgementsSources of Funding This work was supported by grants from the National Institutes of Health/National Institute ofNeurological Disorders and Stroke (to R.M.F., X.W.); the Huntington’s Disease Society of America (to R.M.F.);Departmental funds (to R.M.F.); the Hereditary Disease Foundation (to X.W., B.S.K.) and New York State COREgrants (to I.G.S., and B.S.K.).

References1. Li P, Nijhawan D, Budihardjo I, Srinivasula SM, Ahmad M, Alnemri ES, Wang X. Cytochrome c and

datp-dependent formation of apaf-1/caspase-9 complex initiates an apoptotic protease cascade. Cell1997;91:479–489. [PubMed: 9390557]

2. Chan PH. Mitochondria and neuronal death/survival signaling pathways in cerebral ischemia.Neurochem Res 2004;29:1943–1949. [PubMed: 15662830]

3. Namura S, Nagata I, Takami S, Masayasu H, Kikuchi H. Ebselen reduces cytochrome c release frommitochondria and subsequent DNA fragmen-tation after transient focal cerebral ischemia in mice.Stroke 2001;32:1906–1911. [PubMed: 11486124]

4. Zhang WH, Wang X, Narayanan M, Zhang Y, Huo C, Reed JC, Friedlander RM. Fundamental role ofthe rip2/caspase-1 pathway in hypoxia and ischemia-induced neuronal cell death. Proc Natl Acad SciU S A 2003;100:16012–16017. [PubMed: 14663141]

5. Wang X, Zhu S, Drozda M, Zhang W, Stavrovskaya IG, Cattaneo E, Ferrante RJ, Kristal BS,Friedlander RM. Minocycline inhibits caspase-independent and -dependent mitochondrial cell deathpathways in models of Huntington’s disease. Proc Natl Acad Sci U S A 2003;100:10483–10487.[PubMed: 12930891]

6. Wang X, Zhu S, Pei Z, Drozda M, Stavrovskaya IG, Del Signore SJ, Cormier K, Shimony EM, WangH, Ferrante RJ, Kristal BS, Friedlander RM. Inhibitors of cytochrome c release with therapeuticpotential for Huntington’s disease. J Neurosci 2008;28:9473–9485. [PubMed: 18799679]

7. Zhang WH, Wang H, Wang X, Narayanan MV, Stavrovskaya IG, Kristal BS, Friedlander RM.Nortriptyline protects mitochondria and reduces cerebral ischemia/hypoxia injury. Stroke2008;39:455–462. [PubMed: 18174477]

Wang et al. Page 8

Stroke. Author manuscript; available in PMC 2009 May 1.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 9: Ischemic Injury Author Manuscript NIH Public Access C ... · (Sigma) at 37°C for 20 minutes. After staining, each slice was scanned with an HP scanjet 4200C. The stained and unstained

8. Zhu S, Stavrovskaya IG, Drozda M, Kim BY, Ona V, Li M, Sarang S, Liu AS, Hartley DM, Wu duC, Gullans S, Ferrante RJ, Przedborski S, Kristal BS, Friedlander RM. Minocycline inhibitscytochrome c release and delays progression of amyotrophic lateral sclerosis in mice. Nature2002;417:74–78. [PubMed: 11986668]

9. Baranov SV, Stavrovskaya IG, Brown AM, Tyryshkin AM, Kristal BS. Kinetic model for Ca2+-induced permeability transition in energized liver mitochondria discriminates between inhibitormechanisms. J Biol Chem 2008;283:665–676. [PubMed: 17962193]

10. Friedlander RM, Gagliardini V, Hara H, Fink KB, Li W, MacDonald G, Fishman MC, GreenbergAH, Moskowitz MA, Yuan J. Expression of a dominant negative mutant of interleukin-1 betaconverting enzyme in transgenic mice prevents neuronal cell death induced by trophic factorwithdrawal and ischemic brain injury. J Exp Med 1997;185:933–940. [PubMed: 9120399]

11. Plesnila N, Zinkel S, Le DA, Amin-Hanjani S, Wu Y, Qiu J, Chiarugi A, Thomas SS, Kohane DS,Korsmeyer SJ, Moskowitz MA. Bid mediates neuronal cell death after oxygen/ glucose deprivationand focal cerebral ischemia. Proc Natl Acad Sci U S A 2001;98:15318–15323. [PubMed: 11742085]

12. Sarang SS, Yoshida T, Cadet R, Valeras AS, Jensen RV, Gullans SR. Discovery of molecularmechanisms of neuroprotection using cell-based bioassays and oligonucleotide arrays. PhysiolGenomics 2002;11:45–52. [PubMed: 12388792]

13. Tikka T, Fiebich BL, Goldsteins G, Keinanen R, Koistinaho J. Mino-cycline, a tetracycline derivative,is neuroprotective against excitotoxicity by inhibiting activation and proliferation of microglia. JNeurosci 2001;21:2580–2588. [PubMed: 11306611]

14. Mejia, RO Sanchez; Ona, VO.; Li, M.; Friedlander, RM. Minocycline reduces traumatic brain injury-mediated caspase-1 activation, tissue damage, and neurological dysfunction. Neurosurgery2001;48:1393–1399. [PubMed: 11383749]discussion 1399-1401

15. Friedlander RM. Apoptosis and caspases in neurodegenerative diseases. N Engl J Med2003;348:1365–1375. [PubMed: 12672865]

16. Cazevieille C, Safa R, Osborne NN. Melatonin protects primary cultures of rat cortical neurones fromNMDA excitotoxicity and hypoxia/reoxy-genation. Brain Res 1997;768:120–124. [PubMed:9369308]

17. Chen HY, Chen TY, Lee MY, Chen ST, Hsu YS, Kuo YL, Chang GL, Wu TS, Lee EJ. Melatonindecreases neurovascular oxidative/nitrosative damage and protects against early increases in theblood-brain barrier permeability after transient focal cerebral ischemia in mice. J Pineal Res2006;41:175–182. [PubMed: 16879324]

18. Koh PO. Melatonin attenuates the focal cerebral ischemic injury by inhibiting the dissociation of pbadfrom 14-3 3. J Pineal Res 2008;44:101–106. [PubMed: 18078455]

19. Carloni S, Perrone S, Buonocore G, Longini M, Proietti F, Balduini W. Melatonin protects from thelong-term consequences of a neonatal hypoxic-ischemic brain injury in rats. J Pineal Res2008;44:157–164. [PubMed: 18289167]

20. Rennie K, de Butte M, Frechette M, Pappas BA. Chronic and acute melatonin effects in gerbil globalforebrain ischemia: Long-term neural and behavioral outcome. J Pineal Res 2008;44:149–156.[PubMed: 18289166]

21. Letechipia-Vallejo G, Gonzalez-Burgos I, Cervantes M. Neuroprotective effect of melatonin on braindamage induced by acute global cerebral ischemia in cats. Arch Med Res 2001;32:186–192.[PubMed: 11395182]

22. Susin SA, Lorenzo HK, Zamzami N, Marzo I, Snow BE, Brothers GM, Mangion J, Jacotot E,Costantini P, Loeffler M, Larochette N, Goodlett DR, Aebersold R, Siderovski DP, Penninger JM,Kroemer G. Molecular characterization of mitochondrial apoptosis-inducing factor. Nature1999;397:441–446. [PubMed: 9989411]

23. Chang LK, Putcha GV, Deshmukh M, Johnson EM. Mitochondrial involvement in the point of noreturn in neuronal apoptosis. Biochimie 2002;84:223–231. [PubMed: 12022953]

24. Zoratti M, Szabo I, De Marchi U. Mitochondrial permeability transitions: How many doors to thehouse? Biochim Biophys Acta 2005;1706:40–52. [PubMed: 15620364]

25. Crompton M, Virji S, Doyle V, Johnson N, Ward JM. The mitochondrial permeability transition pore.Biochem Soc Symp 1999;66:167–179. [PubMed: 10989666]

Wang et al. Page 9

Stroke. Author manuscript; available in PMC 2009 May 1.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 10: Ischemic Injury Author Manuscript NIH Public Access C ... · (Sigma) at 37°C for 20 minutes. After staining, each slice was scanned with an HP scanjet 4200C. The stained and unstained

26. Stavrovskaya IG, Narayanan MV, Zhang W, Krasnikov BF, Heemskerk J, Young SS, Blass JP, BrownAM, Beal MF, Friedlander RM, Kristal BS. Clinically approved heterocyclics act on a mitochondrialtarget and reduce stroke-induced pathology. J Exp Med 2004;200:211–222. [PubMed: 15263028]

27. Li P, Allen H, Banerjee S, Franklin S, Herzog L, Johnston C, McDowell J, Paskind M, Rodman L,Salfeld J, et al. Mice deficient in Il-1 beta-converting enzyme are defective in production of matureil-1 beta and resistant to endotoxic shock. Cell 1995;80:401–411. [PubMed: 7859282]

28. Friedlander RM, Gagliardini V, Rotello RJ, Yuan J. Functional role of interleukin 1 beta (Il-1 beta)in Il-1 beta-converting enzyme-mediated apoptosis. J Exp Med 1996;184:717–724. [PubMed:8760825]

29. Wang X, Narayanan M, Bruey JM, Rigamonti D, Cattaneo E, Reed JC, Friedlander RM. Protectiverole of cop in rip2/caspase-1/caspase-4-mediated HeLa cell death. Biochim Biophys Acta2006;1762:742–754. [PubMed: 16920334]

30. Yrjanheikki J, Tikka T, Keinanen R, Goldsteins G, Chan PH, Koistinaho J. A tetracycline derivative,minocycline, reduces inflammation and protects against focal cerebral ischemia with a widetherapeutic window. Proc Natl Acad Sci U S A 1999;96:13496–13500. [PubMed: 10557349]

31. Pei Z, Pang SF, Cheung RT. Administration of melatonin after onset of ischemia reduces the volumeof cerebral infarction in a rat middle cerebral artery occlusion stroke model. Stroke 2003;34:770–775. [PubMed: 12624306]

32. Reiter RJ, Sainz RM, Lopez-Burillo S, Mayo JC, Manchester LC, Tan DX. Melatonin amelioratesneurologic damage and neurophysiologic deficits in experimental models of stroke. Ann N Y AcadSci 2003;993:35–47. [PubMed: 12853293]discussion 48-53

33. Nicholls DG. Mitochondrial function and dysfunction in the cell: Its relevance to aging and aging-related disease. Int J Biochem Cell Biol 2002;34:1372–1381. [PubMed: 12200032]

34. Kim GW, Sugawara T, Chan PH. Involvement of oxidative stress and caspase-3 in cortical infarctionafter photothrombotic ischemia in mice. J Cereb Blood Flow Metab 2000;20:1690–1701. [PubMed:11129785]

Wang et al. Page 10

Stroke. Author manuscript; available in PMC 2009 May 1.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 11: Ischemic Injury Author Manuscript NIH Public Access C ... · (Sigma) at 37°C for 20 minutes. After staining, each slice was scanned with an HP scanjet 4200C. The stained and unstained

Figure 1.Methazolamide and melatonin inhibit cell death of primary cerebrocortical neurons. Cell deathof PCNs was induced by 3-hour exposure to OGD with or without a series of concentrationsof methazolamide (A and B) and melatonin (C and D). Cell death was evaluated by the lactatedehydrogenase assay (A and C). Data from 3 independent experiments are graphed, andstatistically significant differences are indicated with * if P<0.05 and ** if P<0.001. The extentof cell death (as measured by the release of lactate dehydrogenase activity) is alwaysnormalized relative to what is measured in the absence of both death stimulus and test drugs(white bar). The black bar corresponds to the extent of cell death in response to the respectivestress without the test drug. Gray bars correspond to the extent of cell death with stress and theindicated concentration of the test drug. The relative cell death results are displayed graphically.The resulting curves (plotted semilogarithmically) define the IC50 and maximum protectionof methazolamide (B) and melatonin (D).

Wang et al. Page 11

Stroke. Author manuscript; available in PMC 2009 May 1.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 12: Ischemic Injury Author Manuscript NIH Public Access C ... · (Sigma) at 37°C for 20 minutes. After staining, each slice was scanned with an HP scanjet 4200C. The stained and unstained

Figure 2.Methazolamide and melatonin inhibit cell death of primary cerebrocortical neurons underexposure to H2O2 or NMDA. Cell death of PCNs was induced by 18-hour exposure to 1 mmol/L H2O2 (A through C and G) or 500 μmol/L NMDA (D through F and G). A through F, Celldeath was evaluated by the lactate dehydrogenase assay. Data from 3 independent experimentsare graphed, and statistically significant differences are indicated with * if P<0.05 and ** ifP<0.001. White, black, and gray bars are labeled as indicated in Figure 1. G, Phase-contrastlight micrographs of control versus H2O2-treated PCNs with or without incubation withmethazolamide (10 μmol/L) or melatonin (10 μmol/L) are shown as indicated (middle panel).Nuclei are stained with Hoechst 33342 (upper panel). Arrows point to apoptotic cells with

Wang et al. Page 12

Stroke. Author manuscript; available in PMC 2009 May 1.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 13: Ischemic Injury Author Manuscript NIH Public Access C ... · (Sigma) at 37°C for 20 minutes. After staining, each slice was scanned with an HP scanjet 4200C. The stained and unstained

condensed or fragmented chromatin. Bar=5 μm. H, TUNEL staining in cells treated withH2O2 reveals a large increase of positive nuclei with condensed chromatin and DNAfragmentation as compared to control cells. Incubation with methazolamide (10 μmol/L) ormelatonin (10 μmol/L) significantly reduces the numbers of TUNEL-positive cells. ScaleBar=5 μm.

Wang et al. Page 13

Stroke. Author manuscript; available in PMC 2009 May 1.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 14: Ischemic Injury Author Manuscript NIH Public Access C ... · (Sigma) at 37°C for 20 minutes. After staining, each slice was scanned with an HP scanjet 4200C. The stained and unstained

Figure 3.Methazolamide and melatonin inhibit the release of mitochondrial apoptogenic factors andforestall caspase-3 activation. Cell death was induced in PCNs by subjecting cells to OGD for3 hours with or without 10 μmol/L methazolamide (A and C) or 10 μmol/L melatonin (B andC). Subsequently, cells were extracted, and either cytosolic components (A and B) or wholecell lysates (C), or supernatants (D and E) were obtained. The samples, each of which contained50 μg of protein, were analyzed by Western blot using antibodies to cytochrome c, AIF(cytosolic components; A and B), or caspase-3 (whole cell lysates; C). Beta-actin was used asa loading control. This blot is representative of 3 independent experiments.

Wang et al. Page 14

Stroke. Author manuscript; available in PMC 2009 May 1.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 15: Ischemic Injury Author Manuscript NIH Public Access C ... · (Sigma) at 37°C for 20 minutes. After staining, each slice was scanned with an HP scanjet 4200C. The stained and unstained

Figure 4.Methazolamide and melatonin slow the dissipation of ΔΨm, but melatonin does not inhibitmPT. A, PCNs were subjected to OGD for 3 hours with or without methazolamide (10 μmol/L) and melatonin (10 μmol/L). The living cells were then stained with 2 μmol/L Rh 123 todetermine the electrostatic charge of the mitochondria. B, Melatonin was investigated in thein vitro system of purified liver mitochondria that had been stimulated with Ca2+ ions.Melatonin did not prevent the induction of the mPT as judged by an unchanging degree ofmitochondrial swelling. The lack of such an effect on addition of melatonin to a solution ofstimulated mitochondria is illustrated in B. The same is true of mitochondria stimulated inseveral ways, ie, with Ca2+ ions; Ca2+ and tBH, PhAsO, or Ca2+ and diamide. The dose—response curves to these stimuli hardly change when melatonin is present at 0.01 μmol/L to 2mmol/L. In all cases, mitochondrial swelling is monitored by a standard spectroscopic assayusing light of 540 nm. C, Liver mitochondria (0.25 mg/mL) were energized with 5 mmol/Lglutamate/malate and incubated with 1, 5, 10, and 20 μmol/L of melatonin in buffer containing250 mmol/L sucrose, 10 mmol/L HEPES, 2 mmol/L KH2PO4. Mitochondria were challengedwith bolus additions of 5 μmol/L Ca2+ every 2 minutes until release of sequestered Ca2+

occurred. Alamethicin (100 μg) was added in the end of each sample. Upper left panel isΔΨm, upper right is in the buffer, lower left is NADH level, and lower right is swelling. SeeMethods for additional information.

Wang et al. Page 15

Stroke. Author manuscript; available in PMC 2009 May 1.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 16: Ischemic Injury Author Manuscript NIH Public Access C ... · (Sigma) at 37°C for 20 minutes. After staining, each slice was scanned with an HP scanjet 4200C. The stained and unstained

Figure 5.Methazolamide and melatonin inhibit the release of IL-1β and caspase-1 activation. Cell deathwas induced in PCNs by subjecting cells to OGD for 3 hours with or without 10 μmol/Lmethazolamide (A, C, and D) and 10 μmol/L melatonin (B, C, and D). Subsequently,conditioned media was collected and assayed for mature IL-1β release after the completion ofOGD (A and B), or whole cells were extracted (C and D), and analyzed by Western blot (eachof which contained 50 μg of protein) using antibodies to caspase–1 (C). Beta-actin was usedas a loading control. This blot is representative of 3 independent experiments. Caspase-1activity was quantified using a fluorogenic assay in lysed cells. Results are the average of atleast 3 independent experiments. *P< 0.05, **P<0.001.

Wang et al. Page 16

Stroke. Author manuscript; available in PMC 2009 May 1.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 17: Ischemic Injury Author Manuscript NIH Public Access C ... · (Sigma) at 37°C for 20 minutes. After staining, each slice was scanned with an HP scanjet 4200C. The stained and unstained

Figure 6.Methazolamide and melatonin diminish damage from cerebral ischemia. Lesion size (A andB) and neuro-scores (C) were determined for mice injected with saline (controls), ormethazolamide (20 mg/kg body weight), or melatonin (10 mg/kg body weight). Drugs wereadministered either 1 hour before or 30 minutes after MCAO. Each test and control groupconsisted of 7 to 14 mice (n=7 to 14). In all graphs, Data are presented as mean±SE, statisticallysignificant effects are marked with * if P<0.05, and with ** if P<0.001. Brains were quicklyremoved, cut into coronal sections, and stained with 2,3,5-triphenyltetrazolium chloride (A).In addition, lysates of brain tissue were resolved into cytosolic fractions for analysis by Westernblotting (D and E). The protein samples were resolved on SDS/PAGE gels, transferred tonitrocellulose, probed with antibodies to cytochrome c or caspase-3, and reprobed with anti—β-actin. Western blots revealed changes in the cellular localization and degree of maturationof the respective target proteins (D and E). Densitometric scans of these gels allowed the signalsfrom cytoplasmic cytochrome c and activated caspase-3 to be compared to that from β-actin.Each test group consisted of 3 to 5 mice. Again, * indicates that P<0.05 and ** that P<0.001.White bars correspond to brain samples from animals that neither underwent MCAO norreceived any test drug. Black bars correspond to samples from saline-injected animals that didundergo MCAO. Grey and light grey bars correspond to samples from test animals, ie, thosethat were both treated with an experimental drug and underwent MCAO.

Wang et al. Page 17

Stroke. Author manuscript; available in PMC 2009 May 1.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript