Altered P2X7-receptor level and function in ... - The FASEB Journal

0 downloads 0 Views 2MB Size Report
Madrid, Spain; and §Departament de Biologia Cellular i Anatomia Patològica, Facultat de Medicina, ... animal models in response to small drugs that inhibit.
The FASEB Journal • Research Communication

Altered P2X7-receptor level and function in mouse models of Huntington’s disease and therapeutic efficacy of antagonist administration Miguel Díaz-Herna´ndez,*,†,‡,1 María Díez-Zaera,‡,1 Jesu´s Sa´nchez-Nogueiro,*,‡ Rosa Go´mez-Villafuertes,‡ Josep M. Canals,†,§ Jordi Alberch†,§ María Teresa Miras-Portugal,‡ and Jose´ J. Lucas*,†,2 *Centro de Biología Molecular “Severo Ochoa”, Consejo Superior de Investigaciònes Cientificas/ Universidad Autonóma de Madrid, Madrid, Spain; †Centro de Investigacio´n Biome´dica en Red sobre Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain; ‡Departamento de Bioquímica y Biología Molecular IV, Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain; and §Departament de Biologia Cellular i Anatomia Patològica, Facultat de Medicina, Institut d’Investigacions Biomèdiques August Pi i Sunyer, Universitat de Barcelona, Barcelona, Spain The precise mechanism by which mutant huntingtin elicits its toxicity remains unknown. However, synaptic alterations and increased susceptibility to neuronal death are known contributors to Huntington’s disease (HD) symptomatology. While decreased metabolism has long been associated with HD, recent findings have surprisingly demonstrated reduced neuronal apoptosis in Caenorhabditis elegans and Drosophila models of HD by drugs that diminish ATP production. Interestingly, extracellular ATP has been recently reported to elicit neuronal death through stimulation of P2X7 receptors. These are ATP-gated cation channels known to modulate neurotransmitter release from neuronal presynaptic terminals and to regulate cytokine production and release from microglia. We hypothesized that alteration in P2X7-mediated calcium permeability may contribute to HD synaptic dysfunction and increased neuronal apoptosis. Using mouse and cellular models of HD, we demonstrate increased P2X7-receptor level and altered P2X7-mediated calcium permeability in somata and terminals of HD neurons. Furthermore, cultured neurons expressing mutant huntingtin showed increased susceptibility to apoptosis triggered by P2X7receptor stimulation. Finally, in vivo administration of the P2X7-antagonist Brilliant Blue-G (BBG) to HD mice prevented neuronal apoptosis and attenuated body weight loss and motor-coordination deficits. These in vivo data strongly suggest that altered P2X7-receptor level and function contribute to HD pathogenesis and highlight the therapeutic potential of P2X7 receptor antagonists.—Díaz-Herna´ndez, M., Díez-Zaera, M., Sa´nchez-Nogueiro, J., Go´mez-Villafuertes, R., Canals, J. M., Alberch, J., Miras-Portugal, M. T., Lucas, J. J. Altered P2X7-receptor level and function in mouse models of Huntington’s disease and therapeutic efficacy of antagonist administration. FASEB J. 23, 1893–1906 (2009)

ABSTRACT

Key Words: ATP 䡠 calcium 䡠 apoptosis 䡠 polyglutamine 䡠 BBG 0892-6638/09/0023-1893 © FASEB

Huntington’s disease (HD) is an autosomal dominant neurodegenerative disorder caused by a CAG triplet-repeat expansion coding for a polyglutamine (polyQ) sequence in the N-terminal region of the huntingtin (htt) protein (1). Patients suffer from motor dysfunction, cognitive decline, and psychological disturbances over 10 to 15 yr until death. Neuropathology is characterized by brain atrophy and neuronal death predominantly in cortex and striatum and by the presence of neuropil and nuclear neuronal inclusion bodies also in cortex and striatum, as well as in less affected brain regions (2). The precise molecular interactions by which mutanthtt exerts its toxicity remain unknown, although several pathogenic mechanisms have been proposed (3-5), the relative contributions of which may vary with disease progression. Among these, synaptic alterations (6) and increased susceptibility to neuronal death (7) are known contributors to HD symptomatology, in which there is an initial phase without neuronal loss. The initial phase is characterized by neuronal dysfunction associated with decreases in neuronal volume, arborization, and fiber density in the cortex and striatum of HD patients (8, 9). Furthermore, these alterations are mirrored in mouse models of HD (10, 11). While neuronal loss is not initially observed in symptomatic mice, later stages of disease progression exhibit apoptotic (12, 13) and nonapoptotic neuronal loss (9, 14, 15), contributing to brain atrophy. Accordingly, therapeutic strategies aimed at preventing neuronal loss have been proposed for Huntington’s disease (7, 16). While decreased metabolism has long been associated with HD (17), recent findings have surprisingly 1

These authors contributed equally to this work. Correspondence: Centro de Biología Molecular “Severo Ochoa”, CSIC/UAM, Campus UAM de Cantoblanco, 28049 Madrid. Spain. E-mail: [email protected] doi: 10.1096/fj.08-122275 2

1893

demonstrated decreased neuronal apoptosis in HD animal models in response to small drugs that inhibit metabolism (18). Specifically, in vivo administration of drugs that impair ATP production such as rotenone or oligomycin decreased neuronal death in Caenorhabditis elegans and Drosophila models of HD (18). These effects may represent an attenuation of toxic effects elicited by extracellular ATP. In support of this hypothesis, a recent study reported that extracellular ATP induces both apoptotic and nonapoptotic death of SN4741 dopaminergic neurons through stimulation of the P2X7 receptor (19). The P2X7 receptor is an ATP-gated cation channel initially cloned from rat brain (20) and highly expressed in cells of hematopoietic lineage and in multiple cell types of the immune system (20). Within the brain, it was first reported in microglia (21) and later in other types of glia and in neurons, with prominent expression in synaptic terminals (22). A systematic analysis of P2X7 receptors expression throughout the entire rat brain has recently been reported (23). This detailed in situ hybridization analysis of P2X7 mRNA expression combined with double labeling with cell type-specific markers confirmed robust expression in neurons throughout the brain, as well as in scattered glial cells such as microglia, oligodendrocytes, and, to a lesser extent, astrocytes (23). In neurons, the P2X7 receptor has been shown to regulate axonal growth and branching (24) and to modulate neurotransmitter release from presynaptic terminals (22, 25, 26). Stimulation of P2X7 in the immune system regulates cytokine production and release (27). P2X7 receptors have also been shown to mediate apoptosis in response to elevations in intracellular calcium in hematopoietic and immune system cells (28-30) and in neurons (19). In view of the reported role of the P2X7 receptor in mediating ATP induced neuronal death, and the antiapoptotic effect of ATP-synthesis inhibitors in C. elegans and Drosophila models of HD, we hypothesize a potential role of P2X7 receptors in HD pathogenesis. Accordingly, we have explored the status of P2X7 receptors in cellular and mouse models of HD, as well as the therapeutic potential of P2X7 antagonist administration.

MATERIALS AND METHODS Animals Tet/HD94, R6/1, and P2X7-KO mice were generated previously (31-33). Mice were bred and maintained at the Animal facility of Centro de Biología Molecular “Severo Ochoa” (Madrid, Spain). Four to five mice were housed per cage with food and water available ad libitum. Mice were maintained in a temperature-controlled environment on a 12:12-h lightdark cycle with light onset at 7:00 AM. All animal studies were performed with authorization of the Consejo Superior de Investigaciònes Cientificas (CSIC) bioethics committee and 1894

Vol. 23

June 2009

were performed following institutional guidelines for animal handling and research. Synaptosomal preparation Tet/HD94 and R6/1 mice were cervically dislocated and decapitated, and the forebrain was removed. The synaptosomal isolation was made through a Percoll gradient, as described previously (34). Neuronal primary culture and transfection Primary cultures of striatal and cortical neurons were prepared according to modifications of established procedures (35, 36). Cortex and striatum were dissected and dissociated individually from each pup, at postnatal day 1, using the Papain Dissociation System (PDS; Worthington Biochemical Corp., Lakewood, NJ, USA). To identify pups of the Tet/ HD94 genotype, the remainder of brain tissue was maintained in X-Gal solution, whereas the pups of R6/1 genotype were identified by PCR, extracting DNA from their tails. The P2X7-KO pups did not require genotyping since they are produced from a homozygous colony. Neurons were plated at a density of 100,000 cells/cm2 and maintained in Neurobasal medium (NB; Life Technologies, Inc., Gaithersburg, MD, USA) supplemented with 1% B-27, 5% fetal calf serum (FCS), 0.5 mM glutamine, 100 U/ml penicillin, and 100 mg/ml streptomycin, and grown on 3 ␮g/ml laminin (Sigma, St. Louis, MO, USA) and 10 ␮g/ml poly-l-lysine-coated cell culture Chamber Slides (Nunc) or 12 mm coverslip. The cells were incubated in 95% air/5% CO2 in a humidified incubator at 37°C. For calcium-imaging studies, neurons were transfected at 3-4 d in vitro with expression plasmids encoding an N-terminal fragment of Htt fused to GFP (pGW1-Httex1-[Q17 or Q72]-GFP) kindly provided by Drs. Finkbeiner and Arrasate (Gladstone Institute, San Francisco, CA, USA) (37). Neuronal transfection was carried using Lipofectamine 2000 (9 ␮l; Invitrogen, Carlsbad, CA, USA) and 3 ␮g of pGW1Httex1-[Q17 or Q72]-GFP vectors. The transfection mix was removed after 3 h, and the neurons were washed and maintained in culture for 3-4 d until functional assay. Quantitative PCR experiments Total RNA from mouse cortex and striatum was isolated using the RNeasy plus mini kit (Qiagen, Hilden, Germany), following the manufacturer’s instructions. RNA isolation and RT reactions were performed, as described previously (38). Quantitative real-time PCR was performed using gene-specific primers and TaqMan MGB probes for mouse P2X7 (exon boundary 5-6) and ␤-actin (Applied Biosystems, Nutley, NJ, USA). Fast thermal cycling was performed using a StepOnePlus Real-Time PCR System (Applied Biosystems) as follows: denaturation, one cycle of 95°C for 20 s, followed by 40 cycles each of 95°C for 1 s and 60°C for 20 s. The results were normalized as indicated by parallel amplification of the endogenous control ␤-actin. Semiquantitative PCR experiments were also performed using primers that specifically amplify C-terminal region of full-length P2X7 transcript (sense primer, 5⬘-GGT GCC AGT GTG GAA ATT G-3⬘; antisense primer, 5⬘-TAG GGA TAC TTG AAG CCA CT-3⬘; 358-bp PCR product). The PCR reaction mixes contained AmpliTaq Gold PCR Master Mix (Applied Biosystems), and 0.6 ␮M of sense and antisense primers in a total volume of 25 ␮l. For P2X7, the reaction was conducted for 30 cycles with their respective primers and for 20 cycles with ␤-actin primers as internal standard (sense primer, 5⬘-GTA CCA CAG GCA TTG TGA TGG ACT-3⬘;

The FASEB Journal

DI´AZ-HERNA´NDEZ ET AL.

antisense primer, 5⬘-TGC CAC AGG ATT CCA TAC CCA AGA-3⬘; 381-bp PCR product). The number of cycles was predetermined to fall within the linear range of amplification of each PCR product. PCR involved an initial denaturation at 95°C for 10 min; denaturation at 95°C for 45 s; 57°C for 40 s annealing step; and extension at 72°C for 50 s. Antibodies The following antibodies were used: polyclonal antibodies against P2X7 receptor from GE Health Care-Pharmacia (Little Chalfont, UK), from Chemicon (Temecula, CA, USA), and from Alomone (Jerusalem, Israel); polyclonal antiDARPP-32 and monoclonal anti-neuronal nuclei (NeuN) were obtained from Chemicon (Temecula, CA, USA); monoclonal anti-␣-tubulin, monoclonal anti-synaptophysin, and monoclonal anti-glial fibrillar acidic protein (GFAP) were obtained from Sigma (St. Louis, MI, USA); monoclonal anti-␤-III tubulin was provided by Promega (Madison, WI, USA); polyclonal anti-cleaved caspase-3 was obtained from Cell Signaling (Beverly, MA, USA); and polyclonal anti-Iba-1 was obtained from Wako (Osaka, Japan). Western blot analysis Extracts for Western blot analysis were prepared by homogenizing fresh dissected mouse brain regions in ice-cold extraction buffer consisting of 20 mM HEPES, pH 7.4, 100 mM NaCl, 10 mM NaF, 1% Triton X-100, 1 mM sodium orthovanadate, 10 mM EDTA, and protease inhibitors (2 mM PMSF, 10 ␮g/ml aprotinin, 10 ␮g/ml leupeptin, and 10 ␮g/ml pepstatin). The samples were homogenized at 4°C and protein content determined by Bradford. Total protein (20 ␮g) was electrophoresed on 10% SDS-PAGE gel and transferred to a nitrocellulose membrane (Schleicher and Schuell, Keene, NH, USA). The experiments were performed using the following primary antibodies: polyclonal anti-P2X7 (1:1000), monoclonal anti-␣-tubulin (1:10,000), monoclonal anti-synaptophysin (1:2000). The filters were incubated with the antibody at 4°C overnight in 5% nonfat dried milk. A secondary goat anti-mouse (monoclonal) or goat anti-rabbit (polyclonal) antibody (both 1:1000) from DakoCytomation (Glostrup, Denmark) was used followed by ECL detection (Amersham, Piscataway, NJ, USA). Quantification was performed by using a GS-710 Calibrated Imaging densitometer scanner controlled by Quantity One PC software (Bio-Rad, Hercules, CA, USA). In all cases, the average intensity value of the pixels in a background-selected region was calculated and was subtracted from each pixel in the samples. The densitometry values obtained in the linear range of detection with these antibodies were normalized with respect to the values obtained with an anti-␣-tubulin antibody in order to correct for any deviation in loaded amounts of protein. Statistical analysis was performed using one-way ANOVA followed by Bonferroni test.

superfusion chamber in a Nikon Eclipse TE-2000 microscope (Nikon, Tokyo, Japan). This medium was the same as mentioned above, but the MgCl2 was substituted by the accurate glucose concentration to preserve osmolarity. Neurons were continuously superfused at 1.2 ml/min with Mg2⫹-free perfusion medium during functional assays. Initially, pulses of Bz-ATP 10 ␮M for 30 s were applied to synaptosomes in the absence of any other drug. Sequential 50-s pulses of BzATP ranging from 100 nM to 1 mM were assayed on neurons. To check the effect of specific P2X7 receptor antagonists, neurons were incubated for 5 min with Brilliant Blue G (BBG) (from 100 nM to 1 ␮M) or o-ATP at 100 ␮M before 100 ␮M BzATP stimulation. Finally, a pulse of 60 mM K⫹ was applied at the end of each experiment to confirm the viability of neurons and the functionality of synaptosomes under study. All compounds were dissolved in Mg2⫹-free HBM medium. Neurons were visualized using a Nikon microscope with an ⫻40 S Fluor 0.5–1.3 oil lens. The wavelength of the incoming light was filtered to 340 and 380 nm with the aid of a monochromator (10-nm bandwidth, Optoscan monochromator; Cairin, Faversham, UK). The 12-bit images were acquired with an ORCA-ER C 47 42–98 charged-couple device camera from Hamamatsu (Hamamatsu City, Japan) controlled by Metafluor 6.3r6 PC software (Universal Imaging Corp., Cambridge, UK). The exposure time was 250 ms for each wavelength and the changing time ⬍5 ms. The images were acquired continuously and buffered in a fast SCSI disk. The time course data represent the average light intensity in a small elliptical region inside each cell. The background and autofluorescence components were subtracted at each wavelength. Immunohistochemistry Brains were fixed in 4% PFA and cryoprotected in 30% sucrose solution. Thirty-micrometer sagital sections were cut on a freezing microtome (Leica Microsystems, Wetzlar, Germany) and collected in 0.1% azide-PBS solution. Next, brain sections were pretreated for 1 h with 1% BSA, 5% FBS, and 0.2% Triton X-100 and then incubated with primary antibodies at the following dilutions: polyclonal anti-P2X7 (1:200), polyclonal anti-cleaved caspase-3 (1:50), polyclonal anti-Iba-1 (1:200), and monoclonal anti-GFAP (1:200). Finally, brain sections were incubated in avidin-biotin complex using the Elite Vectastain kit (Vector Laboratories). Chromogen reactions were performed with diaminobenzidine (Sigma) and 0.003% H2O2 for 10 min. Sections were coverslipped with Fluorosave. Estimation of the number of caspase-3-, Iba-1-, or GFAP-positive cells in cortex of PBS or BBG administered mice was performed by counting the number of positive cells in the motor, somatosensory, and visual cortex areas encompassed in five equidistant 30-␮m sagital sections ranging from 1.32 to 2.04 mm lateral coordinates, according to Paxinos mouse brain atlas (39). Data are presented as the mean ⫾ se number of cells extrapolated to the whole mentioned cortical areas between the 1.32 and 2.04 mm lateral coordinates.

Calcium studies: microfluorimetric analysis Immunofluorescence studies Synaptosome preparation pellets containing 0.5 mg/ml protein were placed on polylysine-coated coverslips. Primary neurons were analyzed after at least 3 d in culture. Synaptosomes or primary cultures were then washed with HBM buffer (140 mM NaCl, 5 mM KCl, 1.2 mM NaH2PO4, 1.2 mM NaHCO3, 1 mM MgCl2, 10 mM glucose, and 10 mM HEPES, pH 7.4), and they were loaded with FURA-2AM solution (5 ␮M) for 30 min at 37°C to allow for the intracellular hydrolysis of FURA-2AM dye. The coverslips were washed again with HBM Mg2⫹-free medium and mounted in a ALTERED P2X7 RECEPTOR IN HUNTINGTON⬘S DISEASE

Sagital mouse brain sections were pretreated with Sudan Black B, 1% BSA, and 1% TX 100 in PBS buffer and incubated with primary antibodies at the following dilutions: polyclonal anti-P2X7 (1:100), polyclonal anti-DARPP-32 (1: 500), monoclonal anti-GFAP (1:200), polyclonal anti-Iba-1 (1:100), polyclonal anti-cleaved caspase-3 (1:50), monoclonal anti-NeuN (1:100), and monoclonal anti-synaptophysin (1: 200). Subsequently, the brain sections were washed with PBS buffer and incubated with secondary antibodies at the follow1895

ing dilutions: goat anti-rabbit IgG labeled with Texas Red (Molecular Probes, Eugene, OR, USA) (1:400) and goat anti-mouse IgG labeled with Oregon Green 488 from Molecular Probes (1:200). Finally, the brain sections were washed with PBS and mounted following the standard procedures. Controls were performed by following the same procedure but substituting the primary antibodies by PBS in presence of 1% BSA. Similar procedures were followed with synaptosomal preparation, except the Sudan Black B pretreatment. Colocalization of two markers was analyzed by taking successive Oregon Green 488 and Texas Red fluorescent images using an Axioskop 2⫹ microscope (Zeiss, Oberkochen, Germany) and a CCD camera (Coolsnap FX color; Photometrics, Tucson, AZ, USA). Positive signal was considered for the different antibodies if the mean intensity values were ⬎140 on a 0-255 scale with 0 ⫽ white and 255 ⫽ black. The cutoff value of 140 was determined from visual analysis of immunolabeling and by comparison with control (maximal level obtained with preabsorbed antibodies). Cultured neuron survival assays: propidium iodide– calcein staining Twenty-four hours after plating, cortical and striatal neurons cultured from Tet/HD94 and R6/1 mice were shifted to medium lacking serum and B27. Cultured neurons were then treated as previously described (24). Briefly, neurons were stimulated with Bz-ATP at concentrations from 1 ␮M to 1 mM or equal volume of PBS solution, and 48 h later, cell viability was assessed by propidium iodide– calcein staining (Molecular Probes). To eliminate the possible effect of adenosine generated by extracellular hydrolysis of Bz-ATP, 0.1 U adenosine deaminase was added to neurons. In some cases, cortical neurons were pretreated with BBG at concentrations from 3 nM to 1 ␮M for 10 min before 10 or 100 ␮M Bz-ATP was assayed. After several brief rinses, cells were visualized by fluorescence microscopy using an Axiovert 135 microscope (Zeiss). Nine fields (selected at random) were analyzed per well (⬃400 cells/field) in two independent experiments. Drug treatment BBG and A-438079 were diluted at 3 mg/ml in vehicle solutions. The vehicle solutions were calcium- and magnesium-free PBS plus 0.2% DMSO for BBG and saline solution for A-438079. BBG (45.5 mg/kg) and A-438079 (34.2 mg/ kg⫽100 ␮mol/kg) in their corresponding vehicles were then administered i.p. to mice, BBG every 48 h, and A-438079 every 24 h. Control littermates were treated with the same relation (volume/body weight) with vehicle solutions. The mice were weighed once a week during drug administration.

Spectrophotometry analysis of BBG levels in plasma and brain BBG was added at certain concentrations either to PBS, plasma, or cerebral homogenate, and the optimal “signature wavelength” was scanned between 200 and 800 nm using a spectrophotometer (Ultrospec III; Pharmacia). In all cases, the maximal absorbance was obtained at 576 nm. Levels of BBG in plasma and in brain homogenates from BBG-treated mice (wild type and R6/1) were determined according to calibration curves generated with a range of BBG dilutions made either in plasma or in brain homogenate. In mice treated during 1 mo, BBG levels reached 7.08 ⫾ 0.86 ␮M in plasma and 152.6 ⫾ 2.54 nM in brain, while those treated for 4 mo reached 20.04 ⫾ 7.44 ␮M in plasma and 226.12 ⫾ 6.8 nM in brain. 1896

Vol. 23

June 2009

Rota-Rod behavioral testing R6/1 mice were tested for motor coordination at different ages (from 5 to 8 mo old) on an accelerating Rota-Rod apparatus (Ugo Basile, Comerio, Italy). Initially, each mouse was pretrained at a fixed speed (8 rpm) by being repeatedly placed on the Rota-Rod as many times as necessary until it was able to remain on the apparatus for 60 s. After this pretraining, Rota-Rod was set to accelerate from 4 to 40 rpm over 5 min, and mice were tested twice at 3-h intervals on two consecutive days (for a total of 4 trials). During accelerating trials, the latency of each mouse to fall from the Rota-Rod was measured. Data are represented as means ⫾ se of latency to fall from the Rota-Rod in the four accelerating trials per genotype and treatment condition. Statistics All experiments were repeated at least three times, and the results are presented as the mean ⫾ se. Where indicated, we performed analyses of significance using a one-way ANOVA test followed by Bonferroni test correction or a Student’s t test followed to Mann-Whitney U-test (Origin 7.0 software; OriginLab Corporation, Northampton, MA, USA).

RESULTS P2X7 receptor levels are increased in the brain of HD mouse models To explore the status and possible pathogenic role of P2X7 receptors in HD, we first analyzed by Western blot the level of this receptor in the most affected brain regions, striatum and cortex, of two different mouse models expressing N-terminal (exon-1 encoded) mutant htt. As shown in Fig. 1 and in Supplemental Fig. 1, only the 75-kDa band corresponding to P2X7 receptor was detected by Western blot analysis in both striatal and cortical homogenates. Interestingly, P2X7 levels were increased (P⫽0.024) in the striatum of 10-mo-old R6/1 mice that ubiquitously express N-terminal mutant htt with a 115 residue PolyQ stretch (32), as compared to their nontransgenic littermates (Fig. 1A). A nonsignificant trend toward increased levels was observed at the age of 5 mo (Fig. 1A), coinciding with the onset of motor impairment (40). Similarly, Tet/HD94 mice that conditionally express exon-1 mutant htt with a shorter PolyQ repeat selectively in forebrain neurons and display a milder and later motor phenotype (13, 31), displayed increased striatal levels of P2X7 receptor beginning at the age of 12 mo (P⫽0.0025). This occurred right after onset of motor phenotype and persisted in all older Tet/HD94 mice tested (Fig. 1B). It is worth noting that P2X7 receptor levels progressively decline with age in the striatum of wild-type mice (Fig. 1A, B), while such decline does not take place in R6/1 mice and is much slower in Tet/HD94 mice. Therefore, it seems that mutant huntingtin expression rather than increasing the total striatal content of P2X7 receptor, attenuates the age-dependent decrease in P2X7 striatal level. Cortical homogenates showed higher levels of P2X7 receptor respect to striatum (2.1 fold, P⫽0.001),

The FASEB Journal

DI´AZ-HERNA´NDEZ ET AL.

Figure 1. Increased level of P2X7 receptor in striatum of two different HD mouse models. A–D) Western blot analysis of the level of P2X7 receptor protein on brain striatal (A, B) and cortical (C, D) extracts from two different HD mouse models (solid bars): R6/1 mice (A, C) and Tet/HD94 mice (B, D) in comparison to their corresponding control littermates (open bars) at different ages (n⫽5 per age and genotype). Membranes were probed with anti-tubulin antibody to correct for any possible deviation on protein loading. E, F) Analysis of P2X7 mRNA level by quantitative RT-PCR in striatum (St) and cortex (Cx) of 10-mo-old R6/1 mice (n⫽3) (E), and at 16-mo-old Tet/HD94 mice (n⫽3) (F) and in their corresponding control littermates of the same age (n⫽8). Results were normalized using the values obtained for ␤-actin mRNA and expressed as the mean ⫾ se of 4 different experiments.

but no differences were found in cortical samples from either HD mouse models as compared to corresponding control littermates (Fig. 1C, D). Using quantitative RT-PCR, we then analyzed the level of P2X7 receptor mRNA in both mouse models with primers that recognize the exon 5-6 boundary of P2X7 mRNA. A higher level of P2X7 mRNA was observed in the cortex than in the striatum (Fig. 1E, F). This finding is in good agreement with the Western blot results and with recently reported mapping of P2X7 mRNA expression in rat brain by in situ hybridization and by RT-PCR (23). Interestingly, increased mRNA levels were observed in the cortex of both mouse models as compared to their wild-type littermates by real-time quantitative PCR (Fig. 1E, F) and also by semiquantitative RT-PCR with previously reALTERED P2X7 RECEPTOR IN HUNTINGTON⬘S DISEASE

ported primers (38) that amplify sequences on P2X7 exon 13 from wild type but not from P2X7 knockout samples (data not shown). In contrast, no alterations in mRNA were found in striatal samples (Fig. 1E, F). In view of these results, the previously reported axonal localization of P2X7 receptor (22, 24, 25) and the fact that cortex is the major source of axonal afference into the striatum, we reasoned that the striatal increase in P2X7 receptor level most likely occurs in axons of cortical neurons projecting to the striatum. Immunofluorescence studies were performed on brain sections from wild-type, R6/1, and Tet/HD94 mice to analyze brain cell subpopulations expressing the P2X7 receptor and to explore its subcellular localization in cortex and striatum. The pattern of P2X7 immunostaining was similar in wild-type and HD mice. P2X71897

immunopositive cell bodies were found in the cortex (Fig. 2A–C) but rarely in the striatum (Fig. 2E–H). In good agreement with P2X7 mRNA expression in rat brain (23), double labeling with the neuronal marker NeuN revealed that the vast majority of cortical cells showing P2X7 immunostaining in their somata were neurons (Fig. 2A). Also supporting findings by Yu et al. (23), a smaller fraction of cells with P2X7-immunopositive somata corresponded to microglia, as evidenced by double immunofluorescence with the Iba-1 marker (Fig. 2B), and virtually no P2X7 labeling could be detected in astrocytes as evidenced by double labeling with the astrocyte marker GFAP (data not shown). In both cortex and striatum, with the exception of the limited P2X7 immunostaining in cell bodies, the bulk of P2X7 immunostaining is located in the neuropil, where it can be found in projections (see asterisks in Fig. 2A–E) and in buttons (see arrowheads in 2A–E and double labeling with synaptophysin in 2F–H). Double labeling with the neuronal cytoskeleton marker ␤IIItubulin confirmed that those projections were axonal processes (Fig. 2C). Such processes were also found traversing the corpus callosum (Fig. 2D) and in the striatum (Fig. 2E). In the latter, however, the punctate pattern is predominant, and double immunofluorescence with markers of striatal neurons (DARPP-32, Fig. 2E) and synaptic terminals (synaptophysin Fig. 2F–H) revealed that these buttons are axon terminals, many of which can be found lining the somata of striatal neurons. In summary, the analysis of P2X7 receptor level and distribution in the HD mouse models demonstrated that P2X7 receptor mRNA levels are increased in the cortex of symptomatic R6/1 and Tet/HD94 mice, while protein levels are found increased only in the striatum. Because cortical neurons project to the striatum and

P2X7 receptor is located in axonal projections, the cortical increase in P2X7 mRNA results in increased P2X7 receptor protein in the striatum where it can be found at the level of synaptic terminals. Altered P2X7 receptor function in synaptic terminals from Tet/HD94 mice We next investigated whether the increased level of P2X7 receptor in the striatum of HD mouse models would result in altered calcium permeability in synaptic terminals in response to stimulation of P2X7 receptors. To this end, we prepared synaptosomes from striatum of 10-mo-old R6/1 mice and of 16-mo-old Tet/HD94 mice. In agreement with the Western blot and immunofluorescence data, the percentage of synaptosomes immunolabeled for P2X7 receptor was higher in the synaptosomal preparations of HD mice as compared to control littermates. As shown in Fig. 3A–C, P2X7/ synaptophysin double immunofluorescence revealed that 27.1 ⫾ 3.5% of Tet/HD94 synaptosomes were immunoreactive for P2X7 receptor vs. 18.3 ⫾ 3.2% of wild-type synaptosomes (P⫽0.032). Furthermore, Western blot of synaptosomal preparation samples revealed an increase in total P2X7 receptor content in the Tet/HD94 samples (Fig. 3D) comparable to that observed in whole tissue homogenates. Similar results were found in the R6/1 mouse samples (data not shown). Together with the double immunofluorescence data on whole tissue, these data confirm that increased levels of P2X7 in synaptic terminals account for the demonstrated increase in whole striatal homogenates. We next analyzed calcium permeability in individual synaptosomes in response to a 30-s exposure to 10

Figure 2. Immunofluorescence analysis of P2X7 receptor localization in brain of HD mice. Immunofluorescence analysis of cortical (A–D) and striatal (D–H) tissue from 10-mo-old R6/1 mice stained with an antibody against P2X7 receptor (green) with double-labeling (red) using different marker antibodies: anti-NeuN (A), anti-Iba-1 (B), anti-␤III-tubulin (C), anti-DARPP-32 (E), and synaptophysin (F–H). Panel C also shows DAPI nuclear staining (blue). White arrow in panel A points to one of the many double-labeled neuronal somata. Empty arrow in panel B indicates a double-labeled microglial cell. Asterisks in panels A–E show the trajectory of P2X7-positive projections. Small arrowheads in panels A, B, E indicate examples of punctate staining of synaptic terminals. Panels F, G show double-labeling immunofluorescence with P2X7 receptor (green) and synaptophysin (red) antibodies, confirming that the punctate pattern corresponds to synaptic terminals. Cx, cortex; CC, corpus callosum; St, striatum. Scale bars ⫽ 30 ␮m. 1898

Vol. 23

June 2009

The FASEB Journal

DI´AZ-HERNA´NDEZ ET AL.

Figure 3. Increased level of P2X7 receptor and altered P2X7-mediated calcium permeability in synaptic terminals from HD mice. Striatal synaptosomal preparations were performed from 10-mo-old R6/1 mice and from 16-mo-old Tet/HD94 mice and their control littermates. A–C) Double immunofluorescence labeling of nerve endings with anti-synaptophysin (green) and anti-P2X7 receptor (red) antibodies from wild-type (A) and Tet/ HD94 (B) mice; quantification is shown as mean ⫾ se number of P2X7 double-labeled synaptic terminals from wild type (open bar) and Tet/HD94 (solid bar) analyzed in 10 fields (selected at random) per genotype (⬃700 synaptosomes/field) in three independent experiments (C). D) Western blot analysis of synaptosomal preparations with P2X7 and synaptophysin antibodies with histogram showing densitometric quantification in Tet/HD94 vs. wild-type samples. E) Quantification of the number of synaptic terminals that respond to 10 ␮M Bz-ATP stimulation in wild-type (open bar) and Tet/HD94 (solid bar) preparations. Data are presented as the mean ⫾ se number of synaptic terminals that respond to Bz-ATP analyzed in 9 fields (selected at random, ⬃300 synaptosomes/field) per genotype in 4 independent experiments. F) Representative profiles of Fura-2 fluorescence induced by 10 ␮M Bz-ATP stimulation on brain synaptic terminals. Solid bars indicate stimulation periods with 10 ␮M Bz-ATP. Percentages of synaptic terminals showing each type of kinetic profile in response to Bz-ATP are also indicated. All analyzed terminals were also verified to respond to 30 mM K⫹ stimulation. Scale bars ⫽ 20 ␮m.

␮M concentration of the P2X7 agonist 2⬘,3⬘-O-(4benzoyl-4-benzoyl)-ATP (BzATP). In good agreement with the higher percentage of P2X7/synaptophysin double-labeled synaptosomes shown in Fig. 3C, the percentage of synaptosomes that responded to the agonist was significantly higher in the synaptosomal preparation from HD mice (27.3⫾2.0 vs. 21.6⫾1.8%, P⫽0.01; Fig. 3E). Interestingly, the calcium imaging recordings in response to BzATP exposure rendered two types of kinetics (Fig. 3F). In one case, individual synaptosomes displayed an initial peak that remained elevated throughout the exposure to BzATP and progressively declined during BzATP washout (we termed this kinetic as declining profile). The second kinetic profile observed was characterized by multiple upscaling peaks of fluorescence that persisted for at least 25 s after BzATP removal (what we termed upscaling profile). In synaptosomal preparations from wild-type mice, both profiles were similarly represented (55.6⫾3.2% declining, 44.4⫾4.1% upscaling). However, in HD mouse synaptosomal preparations, the percentage shifted in favor of the latter (24.5⫾3.1% declining, 76.5⫾6.1% upscaling; Fig. 3E). These data suggest that, in addition to increased P2X7 receptor levels in the striatum of HD mice, an alteration occurs in the ALTERED P2X7 RECEPTOR IN HUNTINGTON⬘S DISEASE

functional state of the receptor in the synaptic terminal. Altered P2X7 receptor function in terminals and somata of primary neurons expressing mutant htt The alteration in calcium homeostasis in synaptic terminals from HD mouse models described above fits well with the proposed role of synaptic alterations in mediating HD symptomatology (6). However, as mentioned above, altered calcium permeability through P2X7 receptors might also contribute to neuronal apoptosis (19, 30, 41). To this end, we investigated alterations in P2X7-mediated calcium permeability in response to expression of mutant-htt in both terminals and somata of primary cultured neurons. To compare mutant-htt-expressing and –nonexpressing neurons in the same culture, we transfected wild-type mouse primary neurons with plasmids driving expression of GFP-fused exon 1 htt with either 17Q or 72 Q repeats. FURA-2 imaging in response to BzATP was then performed both in somata and terminals of neurons expressing either version of htt (identified by their expression of GFP) and of control nontransfected neurons from the same plate (Fig. 4). In good agreement with the data obtained in the synaptosomal preparations, the same 1899

Figure 4. Altered P2X7-mediated calcium permeability in terminals and somata of primary neurons expressing mutant-htt. Primary neuronal cultures were prepared from cortex of wild-type mice and transfected with plasmids driving expression of GFPfused exon 1 htt with either 17Q or 72Q repeats. Then, calcium imaging in response to 10 ␮M BzATP was performed both in somata and terminals of neurons expressing either version of htt (identified by their expression of GFP) and of control nontransfected neurons from the same plate. A) GFP fluorescence of a 72Q-transfected neuron. Empty arrow shows the soma of the transfected neuron. B) Fura-2 dye image of the same field that also contains other nontransfected neurons. Normal arrows indicate nerve terminals of the transfected neuron. Circular-head arrows indicate nerve terminals of the nontransfected neurons. C) Representative Fura-2 fluorescence time course changes induced by 10 ␮M Bz-ATP stimulation on nerve ending of nontransfected (CONT) and transfected (17Q or 72Q) neurons. The solid bars indicate the periods of 10 ␮M Bz-ATP stimulation. Percentages of synaptic terminals showing each type of kinetic profile on response to Bz-ATP are also indicated. D) Representative Fura-2 fluorescence changes induced in somata of 17Q- or 72Q-transfected neurons by 1 to 100 ␮M Bz-ATP stimulations. E) Dose-response curves of Bz-ATP-induced calcium influx in somata of 17Q-transfected (open symbols) or 72Q-transfected (solid symbols) primary neurons either from wild-type or P2X7-KO mice. F) Specificity of the intracellular calcium increase induced by Bz-ATP stimulation is demonstrated by its blockade by preincubation with the P2X7 antagonist BBG (1 ␮M) for 2 min. Besides, all analyzed neurons were routinely verified to respond to 30 mM K⫹ stimulation. Solid bars indicate stimulation periods with different compounds.

two types of kinetics (declining and upscaling) were observed in response to BzATP in terminals of cultured neurons (Fig. 4C). Moreover, in accordance with the findings in synaptosomal preparations from wild-type mice, approximately half of the terminals of control (nontransfected) and of 17 Q (transfected with exon 1 17Q-GFP) neurons displayed one or the other kinetic (52.3⫾3.1% declining and 47.7⫾4.5% upscaling in control terminals; 50.2⫾1.9% declining and 49.8⫾3.5% upscaling in 17Q terminals). As expected, the percentage of terminals of neurons transfected with exon 1 72Q-GFP displaying the upscaling profile was much higher (78.2⫾9.5 vs. 21.8⫾7.5% of terminals showing a declining profile; P⫽0.0012), recapitulating the observation in synaptosomes from HD transgenic mice. Regarding the effect of BzATP in neuronal somata, where endoplasmic reticulum also contributes to the net concentration of intracellular calcium, only one type of calcium imaging profile was observed. This was very similar to the declining kinetic that was observed in terminals but with an additional and 1900

Vol. 23

June 2009

short lasting initial peak (Fig. 4D, F). Interestingly, when we performed a dose response curve of the effect of BzATP in somata of 17Q- and 72Q-transfected neurons, we observed a sensitization induced by mutant Htt. More precisely, the EC50 of the effect of BzATP on calcium influx was 37.9 ⫾ 1.21 ␮M in somata of 17Q-transfected neurons and 4.91 ⫾ 1.86 ␮M in somata of 72Q-transfected neurons (Fig. 4D, E). To confirm that the increased in calcium influx was due to sensitization of the P2X7 receptor, we performed similar experiments in primary neurons from P2X7-knockout (P2⫻7-KO) mice (33). In good agreement with previous reports (42), the calcium influx in response of BzATP was much smaller in primary neurons from P2X7-KO mice and, more important, the sensitization induced by 72Q-Htt in wild-type neurons did not take place in P2X7-KO neurons (Fig. 4E). More precisely, the EC50 of the effect of BzATP on calcium influx was 29.82.9 ⫾ 1.14 ␮M in somata of 17Q-transfected P2X7-KO neurons and 21.03 ⫾ 1.32 ␮M in somata of 72Q-transfected P2X7-KO neurons (Fig. 4D, E). In summary, the

The FASEB Journal

DI´AZ-HERNA´NDEZ ET AL.

increased sensitivity to P2X7 agonist-induced calcium influx found in 72Q-transfected wild-type neurons demonstrates that P2X7 receptor function is also altered in neuronal somata as a consequence of N-terminal mutant-htt expression. HD mouse primary neurons show increased vulnerability to P2X7-mediated apoptosis To test whether increased P2X7 receptor function would sensitize mutant-htt-expressing neurons to apoptosis triggered by extracellular ATP, we performed survival assays on primary cortical and striatal neuronal cultures from Tet/HD94 mice using the propidium iodine-calcein staining method (Fig. 5). As previously reported (10), viability of untreated primary neurons from Tet/HD94 mice was indistinguishable from that of wild-type mice (Fig. 5C, D). In the case of Tet/HD94 striatal neurons, a trend toward decreased viability was observed in response to exposure to 10 ␮M BzATP, though this did not reach statistical significance (Fig. 5D). However, in cortical neurons, such exposure to the ATP analog that did not affect survival of wild-type neurons dramatically reduced viability of Tet/HD94 neurons by 46.1 ⫾ 7.7% (P⫽0.0017, Fig. 5A–C). To further confirm that this apoptotic effect was due to stimulation of P2X7 receptors, cells were preincubated with different doses (0.3 nM to 1 ␮M) of the P2X7 antagonist BBG starting 10 min before the exposure to

Bz-ATP. As shown in Fig. 5C, E, 1 ␮M BBG fully prevented the deleterious effect of Bz-ATP on neuron viability. The IC50 of this inhibitory effect of BBG was 170.07 ⫾ 1.38 nM (Fig. 5E), thus fitting with the reported efficacy of BBG at antagonizing the P2X7 receptor (43). We then performed primary neuronal cultures from cortex of R6/1 mice and of their wildtype littermates and analyzed the effect of Bz-ATP in neuronal survival. In good agreement with the results obtained from Tet/HD94 neurons, Bz-ATP-induced death of R6/1 neurons at doses that did not affect survival of wild-type neurons (Fig. 5F) Taken together, these findings indicate that expression of mutant-htt renders neurons more vulnerable to P2X7 mediated apoptosis. Decreased incidence of neuronal apoptosis and phenotypic improvement by in vivo administration of P2X7 antagonist to HD mice In view of the neuroprotective effect of BBG on P2X7mediated neuronal death in culture, we investigated the potential therapeutic efficacy of this antagonist in vivo in a mouse model of HD. Because a significant increase in P2X7 level in the striatum of R6/1 mice is first observed at the age of 8 mo (data not shown) BBG (45.5 mg/kg, i.p.) or vehicle (PBS) were administered every 48 h during 4-wk to 8-mo-old R6/1 mice and to their corresponding wild-type littermates. This dosage

Figure 5. Increased susceptibility to P2X7-mediated apoptosis in HD primary neurons. Primary cortical and striatal neuronal cultures were prepared from Tet/HD94 mice and from control littermates. The cultures were incubated with 10 ␮M Bz-ATP for 2 h, and 48 h later, they were analyzed by the propidium iodide/calcein method. A, B) Fluorescence images of wild-type (A) and Tet/HD94 (B) cortical cultures showing live (green) calcein- or dead (red) propidium iodide-labeled neurons. Scale bar ⫽ 100 ␮m. C, D) Histograms showing quantification of live (calcein-positive) neurons from wild-type (open bars) or Tet/HD94 (solid bars) mice after Bz-ATP treatment in presence or absence of 1 ␮M BBG. Data represent means ⫾ se of cells analyzed in 9 fields (selected at random) per treatment and genotype (⬃350 cells/field) in 2 independent experiments. E) Dose response of the protective effect of BBG on BzATP-induced death of Tet/HD94 primary-cultured neurons. F) Histograms showing quantification of live (calcein-positive) neurons from wild-type (open bars) or R6/1 (solid bars) mice after treatment with 50 or 100 ␮M Bz-ATP. ALTERED P2X7 RECEPTOR IN HUNTINGTON⬘S DISEASE

1901

paradigm was chosen because it results in a 152.6 ⫾ 2.56 nM concentration in the brain of treated mice (see Materials and Methods), thus in the range of the IC50 of BBG to antagonize P2X7 (10-200 nM) (43) and one order of magnitude below the DL50 of Brilliant BlueCFC (44). As shown in Fig. 6A, the progressive decline in body weight that normally occurs in untreated R6/1 mice also occurred in vehicle (PBS)-administered R6/1 mice. However, BBG administration fully prevented this body weight loss in R6/1 mice (Fig. 6A) with no change evident in the body weight in wild-type mice (data not shown). More important, we also monitored the effect of BBG on motor coordination in the RotaRod apparatus. As shown in Fig. 6B, C, vehicle-treatedR6/1 mice displayed a deterioration in motor performance over the 4-wk testing period (P⫽0.009). However, BBG-treated mice displayed no significant deterioration over the 4 wk and by the end of the testing period performed significantly better than vehicle-treated mice (P⫽0.02; Fig. 6B, C). In contrast, BBG treatment did not affect the motor performance of wild-type mice since both PBS- and BBG-treated wildtype mice showed the typical progressive improvement in motor performance (P⫽0.01 and P⫽0.03) with repeated testing over the 4-wk period (Fig. 6C). In view of the behavioral improvement of R6/1 mice with the BBG antagonist, we decided to test also the recently developed P2X7 antagonist A-438079 that has been reported to modulate pathological nociception after acute administration to rats (45, 46). Since 76 ␮mol/kg

(i.p.) is the reported ED50 of A-438079 in acute reversal of allodynia in the rat model (45), we decided to administer 100 ␮mol/kg i.p. once daily to 8-mo-old R6/1 mice during 4 wk. As shown in Fig. 6A, A-438079 administration resulted in a marked tendency to prevent the decline in body weight that takes place in vehicle-administered mice (P⫽0.072). However, in the Rota-Rod test, A-438079-administered mice were indistinguishable from vehicle-administered mice except for the last time point (after 28 d of administration) at which a moderate tendency toward improvement was detected (Fig. 6B). These minor effects of A-438079 compared to those of BBG probably reflect the reported very short half-life (1.02 h) and limited bioavailability (19%) of this compound (45). We then explored whether the efficacy of BBG in preventing additional weight loss and deterioration in motor coordination also correlated with a reduction in the rate of neuronal death observed in R6/1 mice. As shown in Fig. 7A–F, neuronal apoptosis can be detected in the cortex of 8-mo-old R6/1 mice by immunohistochemistry or immunofluorescence against cleaved caspase-3. Interestingly, and in good agreement with the protective effect of BBG on HD primary cultured neurons, the increased number of cleaved caspase-3immunoreactive neurons seen in vehicle-administered R6/1 mice was fully prevented in BBG-administered R6/1 mice (Fig. 7A–C). Double immunofluorescence labeling experiments with NeuN and P2X7 antibodies confirmed the neuronal identity of the caspase-3positive cells and the fact that they express P2X7

Figure 6. Administration of BBG to R6/1 mice prevents loss of body weight and attenuates their motor-coordination deficit. Eight-mo-old wild-type and R6/1 mice were administered BBG (45.5 mg/kg, i.p.) every 48 h, or A-438079 (100 ␮mol/kg i.p.) every 24 h, or vehicle during 28 d. A) Evolution of body weight in R6/1 mice along 28 d of treatment with vehicle (solid circles), or BBG (open circles), or A-438079 (open squares). Mice were tested every week. B, C) Analysis of motor coordination in the accelerating Rota-Rod test. B) BBG administration to R6/1 improves their Rota-Rod performance as compared to vehicle (PBS)-administered R6/1 mice. C) Effect of BBG or PBS administration on progression of the latency to fall from Rota-Rod of wild-type and R6/1 mice. Mice were tested every week, and mean ⫾ se time spent on rod during the 4 accelerating trials was calculated per each genotype, age, and treatment condition. Data were collected from 3 independent experiments with a total of 62 mice; n ⫽ 9-11/group. 1902

Vol. 23

June 2009

The FASEB Journal

DI´AZ-HERNA´NDEZ ET AL.

Figure 7. Administration of BBG to R6/1 mice prevents neuronal apoptosis without affecting microgliosis or astrocytosis. Eight-mo-old wild-type and R6/1 mice were administered BBG (45.5 mg/kg, i.p.) or vehicle (PBS) every 48 h during 4 wk. A–F) Neuronal apoptosis was detected by immunohistochemistry (A–C) or immunofluorescence (D–F) with cleaved caspase-3 antibody. A) Detection of cleaved caspase-3-positive cells in cortex of a vehicle-administered R6/1 mouse. Insets i–iii correspond to an ⫻4.0 view of the apoptotic cells identified by boxes in A. B) Example of a BBG-administered R6/1 mouse with dramatically reduced cleaved caspase-3 labeling. cc, corpus callosum. C) Histogram showing the quantification of cleaved caspase-3 positive cells in wild-type (open bars) or R6/1 (solid bars) mice treated with vehicle or BBG. Data are presented as mean ⫾ se immunopositive cells per 7 mm2 in a 30-mm section (n⫽10 per genotype and treatment, P⬍0.05). D, E) Cleaved caspase-3/NeuN double immunofluorescence labeling in cortex of a vehicle-administered R6/1 mouse. F) Cleaved caspase-3/P2X7 double-immunofluorescence labeling in cortex of a vehicle-administered R6/1 mouse. G–I) Immunohistochemical analysis of microglial marker Iba-1 in cortex R6/1 mice treated with vehicle (G) or with BBG (H); histogram showing quantification of Iba-1-positive cells (I). J–L) Immunohistochemical analysis of the astrocytic marker GFAP in cortex of R6/1 mice treated with vehicle (K) or with BBG (J); histogram showing quantification of GFAP-positive cells (L). Scale bars ⫽ 200 ␮m (A, B); 50 ␮m (D, F, J); 100 ␮m (G).

receptor (Fig. 7D–F). In summary, these results demonstrate a beneficial effect of the P2X7 antagonist in mutant-htt-induced neuronal apoptosis in vivo, that recapitulates the protective effect observed on primary neuronal cultures and which parallels the in vivo positive effect of the antagonist in the body weight and motor coordination phenotypes. In view of these in vivo results and of those obtained in primary HD cultured neurons, the protective effect of P2X7 antagonist administration seems to be clearly exerted in neurons. However, since the P2X7 receptor is also found in glial cells, we analyzed whether BBG administration also affected the gliosis that takes place in HD mice. Immunohistochemical detection of the microglial marker Iba-1 revealed a tendency for an increased number of reactive microglial cells in the cortex (Fig. 7G–I) and striatum (not shown) of R6/1 mice with respect to their wild-type littermates that did not reach statistical significance and, more important, that was not affected by the BBG treatment (Fig G–I). Similarly, although analysis of GFAP immunohistochemistry did reveal a clear reactive astrocytosis in the cortex (Fig. 7J–L) and striatum (not shown) of R6/1 mice, this was also unaffected by the BBG administration paradigm (Fig. 7J–L) that prevented neuronal apoptosis. In view of the protective effect of BBG administration ALTERED P2X7 RECEPTOR IN HUNTINGTON⬘S DISEASE

to 8-mo-old R6/1 mice, we decided to explore potential beneficial effects of BBG in younger mice. We performed a similar experiment with 5-mo-old mice that do not yet display significant increases in P2X7 receptor level (Fig. 1A) nor increased incidence of cleaved caspase-3 immunoreactivity (data not shown). In these younger R6/1 mice, body weight and Rota-Rod performance were not affected by the 4-wk administration paradigm that was found beneficial in 8-mo-old mice. For this reason, mice were maintained on the BBG or vehicle regimen and were tested every 4 wk until the age of 8 mo. However, despite the longer administration paradigm, antagonist administration did not affect body weight nor Rota-Rod performance in these younger mice, suggesting that P2X7 antagonist therapy may be beneficial only in more advanced stages of disease progression when both increase in P2X7 level and neuronal death occur.

DISCUSSION By analyzing level and function of the P2X7 purinergic receptor in mouse and cellular models of HD, we report increased levels of this receptor and altered P2X7-mediated calcium homeostasis both in somata and terminals of neurons expressing mutant-htt. Fur1903

thermore, we found that primary cultured neurons expressing mutant-htt show increased susceptibility to apoptosis induced by P2X7 receptor stimulation. Finally, in vivo administration of a P2X7 receptor antagonist to HD mice attenuated neuronal apoptosis, as well as the body weight loss and decline in motor coordination observed in advanced stages of phenotype progression. Levels of P2X7 receptor are found increased in the striatum of two different mouse models. We also found a concomitant increase in P2X7 receptor transcription in the cortex and a prominent subcellular localization of the protein in cortical axonal projections and in striatal axon terminals. It thus appears that increased levels in the striatum are due to increased levels in corticostriatal projections, which, in turn, are secondary to increased transcription in cortical neurons. Transcriptional deregulation triggered by the expression of mutant-htt has been extensively postulated as a key mediator in HD pathogenesis (47, 48). We now add P2X7 to the list of genes whose expression is altered in HD, and it would be possible that its increased transcription underlies the multiple P2X7-related alterations that we report here. While many P2X7 gene polymorphisms have been reported, including several in the promoter region (49), none of these alter receptor abundance or activity under the tested conditions. With the exception of the above-mentioned study and to the best of our knowledge, no other detailed studies of the promoter region have been reported. Thus, further investigation is required to elucidate the mechanism by which mutant htt expression elicits increased P2X7 transcription. It would be of interest to explore whether any of the above-mentioned P2X7 gene polymorphisms correlate with those currently being investigated as potential mediators of the variability in severity and in age of onset in HD patients with the same CAG repeat mutation length (50). In synaptosomes and axon terminals of mutant-htt expressing neurons, we have found a prevalence of the so-called upscaling calcium imaging profile in response to a P2X7 agonist. This might simply be a consequence of higher levels of the receptor in the terminals as indicated by the Western blot data and the quantification of the P2X7 immunofluorescent synaptosomes. An alternative explanation would be the existence of two different functional states of the P2X7 receptor, depending on mutant-htt-induced post-translational changes. Interestingly, recordings of P2X7-operated single-channel currents have demonstrated that this receptor can mediate two different types of currents characterized by shortlived and long-lived openings, respectively (51). Because this was observed in single channel recordings, a difference in receptor density cannot account for the two different kinetics, and the authors suggest posttranslational modifications such as phosphorylation. In this regard, changes in kinase-dependent signaling cascades have been previously reported in mutant-httexpressing cells (52). Regarding the cell type in which P2X7 function is 1904

Vol. 23

June 2009

altered and at which BBG exerts its beneficial effect, it is obvious that neurons are affected and that they are involved in the protective effect of the antagonist. This is based on the alteration in P2X7-mediated calcium permeability observed in HD-cultured neurons and in synaptosomes from HD mice, on the fact that in vivo administration of BBG prevents the neuronal apoptosis seen in untreated HD mice, and on the fact that BBG also prevented the increased vulnerability to P2X7mediated apoptosis observed in HD neuronal cultures devoid of glial cells. However, the possibility of the beneficial action of BBG being in part mediated also by glial cells can not be excluded. In this regard, P2X7 antagonists have been shown to exert anti-inflammatory effect in microglia (27, 30) and anti-inflammatory drugs, such as minocycline have also been postulated for treatment of HD (53), though concerns remain regarding its efficacy in animal models (54, 55) and its safety in humans (56). Therefore, apart from the mentioned neuronal effects, in vivo administration of BBG might also have produced an anti-inflammatory effect. However, this does not seem to be the case based on the fact that we did not observe decreased microgliosis or astrocytosis in BBG-administered HD mice. Finally, the herein-reported therapeutic efficacy of the P2X7 antagonist BBG in HD mice strongly suggest potential of P2X7 antagonists for treatment of HD patients. BBG is slightly less potent at antagonizing human P2X7 receptor than at rodent receptors (43). However, other P2X7 antagonists such as KN62 are more potent at human P2X7 receptors (57). Regarding the more recent selective antagonist A-438079, its pharmacodynamics and bioavailability do not seem appropriate for chronic i.p. administration protocols. However, given the current research on its applicability for the treatment of other conditions such as chronic pain (58), it is conceivable that other routes of administration or close derivatives will be explored soon for chronic administration. Therefore, in view of our data and of the current state of research on new P2X7 antagonist, BBG probably has the best potential for translating into clinical studies. Notably, BBG might have the advantage of predicted low toxicity based on the safety of its analog Brilliant Blue-CFC that has been used as a food additive dye for decades (44). Preclinical studies are currently being conducted with BBG itself for ophthalmologic applications (59). In summary, the biochemical, histological, calciumimaging, and in vivo data presented here support the hypothesis that altered level and function of P2X7 receptor may contribute to HD pathogenesis and strongly suggest a therapeutic potential of P2X7 receptor antagonists for HD. The authors thank Drs. M. R. Ferna´ndez and C. Toma´s for helpful discussion and comments; Dr. O. Howard for comments and style correction; and Drs. S. Finkbeiner and M. Arrasate (Gladstone Institute, San Francisco, CA, USA) for kindly providing plasmids expressing exon 1 htt fused to GFP with 17 or 72 Q repeats. The authors are also grateful to Javier Palacín, Desiree´ Ruiz, and Alicia Tomico for technical assistance. This work was

The FASEB Journal

DI´AZ-HERNA´NDEZ ET AL.

supported by Centro de Investigacio´n Biome´dica en Red de Enfermedades Neurodegenerativas (CiberNed) and by grants from Ministerio de Educacio´n y Ciencia, Comunidad Auto´noma de Madrid, Fundacio´n “La Caixa,” Fundacio´n Marcelino Botín, and by an institutional grant from Fundacio´n Ramo´n Areces. M.D.H. was a recipient of a “Juan de la Cierva” contract from Ministerio de Educacio´n y Ciencia and is currently a recipient of a “Ramo´n y Cajal” contract.

20. 21.

22.

REFERENCES 1.

2. 3. 4. 5. 6. 7. 8.

9. 10.

11.

12.

13.

14.

15.

16. 17. 18.

19.

Huntington’s Disease Collaborative Research Group (1993) A novel gene containing a trinucleotide repeat that is expanded and unstable on Huntington’s disease chromosomes. Cell 72, 971–983 Vonsattel, J. P., and DiFiglia, M. (1998) Huntington disease. J. Neuropathol. Exp. Neurol. 57, 369 –384 Ross, C. A. (2002) Polyglutamine pathogenesis: emergence of unifying mechanisms for Huntington’s disease and related disorders. Neuron 35, 819 – 822 Ortega, Z., Diaz-Hernandez, M., and Lucas, J. J. (2007) Is the ubiquitin-proteasome system impaired in Huntington’s disease? Cell. Mol. Life Sci. 64, 2245–2257 Landles, C., and Bates, G. P. (2004) Huntingtin and the molecular pathogenesis of Huntington’s disease. Fourth in molecular medicine review series. EMBO Rep. 5, 958 –963 Li, J. Y., Plomann, M., and Brundin, P. (2003) Huntington’s disease: a synaptopathy? Trends Mol. Med. 9, 414 – 420 Vila, M., and Przedborski, S. (2003) Targeting programmed cell death in neurodegenerative diseases. Nat. Rev. 4, 365–375 DiProspero, N. A., Chen, E. Y., Charles, V., Plomann, M., Kordower, J. H., and Tagle, D. A. (2004) Early changes in Huntington’s disease patient brains involve alterations in cytoskeletal and synaptic elements. J. Neurocytol. 33, 517–533 Vonsattel, J. P. (2008) Huntington disease models and human neuropathology: similarities and differences. Acta Neuropathol. (Berl.) 115, 55-69 Martin-Aparicio, E., Yamamoto, A., Hernandez, F., Hen, R., Avila, J., and Lucas, J. J. (2001) Proteasomal-dependent aggregate reversal and absence of cell death in a conditional mouse model of Huntington’s disease. J. Neurosci. 21, 8772– 8781 Guidetti, P., Charles, V., Chen, E. Y., Reddy, P. H., Kordower, J. H., Whetsell, W. O., Jr., Schwarcz, R., and Tagle, D. A. (2001) Early degenerative changes in transgenic mice expressing mutant huntingtin involve dendritic abnormalities but no impairment of mitochondrial energy production. Exp. Neurol. 169, 340 –350 Portera-Cailliau, C., Hedreen, J. C., Price, D. L., and Koliatsos, V. E. (1995) Evidence for apoptotic cell death in Huntington disease and excitotoxic animal models. J. Neurosci. 15, 3775– 3787 Diaz-Hernandez, M., Torres-Peraza, J., Salvatori-Abarca, A., Moran, M. A., Gomez-Ramos, P., Alberch, J., and Lucas, J. J. (2005) Full motor recovery despite striatal neuron loss and formation of irreversible amyloid-like inclusions in a conditional mouse model of Huntington’s disease. J. Neurosci. 25, 9773–9781 Turmaine, M., Raza, A., Mahal, A., Mangiarini, L., Bates, G. P., and Davies, S. W. (2000) Nonapoptotic neurodegeneration in a transgenic mouse model of Huntington’s disease. Proc. Natl. Acad. Sci. U. S. A. 97, 8093– 8097 Diaz-Hernandez, M., Hernandez, F., Martin-Aparicio, E., GomezRamos, P., Moran, M. A., Castano, J. G., Ferrer, I., Avila, J., and Lucas, J. J. (2003) Neuronal induction of the immunoproteasome in Huntington’s disease. J. Neurosci. 23, 11653–11661 Friedlander, R. M. (2003) Apoptosis and caspases in neurodegenerative diseases. N. Engl. J. Med. 348, 1365–1375 Beal, M. F. (2000) Energetics in the pathogenesis of neurodegenerative diseases. Trends Neurosci. 23, 298 –304 Varma, H., Cheng, R., Voisine, C., Hart, A. C., and Stockwell, B. R. (2007) Inhibitors of metabolism rescue cell death in Huntington’s disease models. Proc. Natl. Acad. Sci. U. S. A. 104, 14,525–14,530 Jun, D. J., Kim, J., Jung, S. Y., Song, R., Noh, J. H., Park, Y. S., Ryu, S. H., Kim, J. H., Kong, Y. Y., Chung, J. M., and Kim, K. T.

ALTERED P2X7 RECEPTOR IN HUNTINGTON⬘S DISEASE

23.

24.

25.

26. 27. 28. 29.

30. 31. 32.

33.

34. 35. 36. 37.

38.

39. 40.

(2007) Extracellular ATP mediates necrotic cell swelling in SN4741 dopaminergic neurons through p2x7 receptors. J. Biol. Chem. 282, 37,350-37,358 Surprenant, A., Rassendren, F., Kawashima, E., North, R. A., and Buell, G. (1996) The cytolytic P2Z receptor for extracellular ATP identified as a P2X receptor (P2X7). Science 272, 735–738 Ferrari, D., Villalba, M., Chiozzi, P., Falzoni, S., RicciardiCastagnoli, P., and Di Virgilio, F. (1996) Mouse microglial cells express a plasma membrane pore gated by extracellular ATP. J. Immunol. 156, 1531–1539 Deuchars, S. A., Atkinson, L., Brooke, R. E., Musa, H., Milligan, C. J., Batten, T. F., Buckley, N. J., Parson, S. H., and Deuchars, J. (2001) Neuronal P2X7 receptors are targeted to presynaptic terminals in the central and peripheral nervous systems. J. Neurosci. 21, 7143–7152 Yu, Y., Ugawa, S., Ueda, T., Ishida, Y., Inoue, K., Kyaw Nyunt, A., Umemura, A., Mase, M., Yamada, K., and Shimada, S. (2008) Cellular localization of P2X7 receptor mRNA in the rat brain. Brain Res. 1194, 45–55 Diaz-Hernandez, M., Diaz-Hernandez, J. I., Diez-Zaera, M., del Puerto, A., Lucas, J. J., Garrido, J. J., and Miras-Portugal, M. T. (2008) Inhibition of the ATP-gated P2X7 receptor promotes axonal growth and branching in cultured hippocampal neurons. J. Cell Sci. 121, 3717–3728 Miras-Portugal, M. T., Diaz-Hernandez, M., Giraldez, L., Hervas, C., Gomez-Villafuertes, R., Sen, R. P., Gualix, J., and Pintor, J. (2003) P2X7 receptors in rat brain: presence in synaptic terminals and granule cells. Neurochem. Res. 28, 1597–1605 North, R. A. (2002) Molecular physiology of P2X receptors. Physiol. Rev. 82, 1013–1067 Inoue, K. (2002) Microglial activation by purines and pyrimidines. Glia 40, 156 –163 Di Virgilio, F., Chiozzi, P., Falzoni, S., Ferrari, D., Sanz, J. M., Venketaraman, V., and Baricordi, O. R. (1998) Cytolytic P2X purinoceptors. Cell Death Differ. 5, 191–199 Tsukimoto, M., Maehata, M., Harada, H., Ikari, A., Takagi, K., and Degawa, M. (2006) P2X7 receptor-dependent cell death is modulated during murine T cell maturation and mediated by dual signaling pathways. J. Immunol. 177, 2842–2850 Le Feuvre, R., Brough, D., and Rothwell, N. (2002) Extracellular ATP and P2X7 receptors in neurodegeneration. Eur. J. Pharmacol. 447, 261–269 Yamamoto, A., Lucas, J. J., and Hen, R. (2000) Reversal of neuropathology and motor dysfunction in a conditional model of Huntington’s disease. Cell 101, 57– 66 Mangiarini, L., Sathasivam, K., Seller, M., Cozens, B., Harper, A., Hetherington, C., Lawton, M., Trottier, Y., Lehrach, H., Davies, S. W., and Bates, G. P. (1996) Exon 1 of the HD gene with an expanded CAG repeat is sufficient to cause a progressive neurological phenotype in transgenic mice. Cell 87, 493–506 Solle, M., Labasi, J., Perregaux, D. G., Stam, E., Petrushova, N., Koller, B. H., Griffiths, R. J., and Gabel, C. A. (2001) Altered cytokine production in mice lacking P2X(7) receptors. J. Biol. Chem. 276, 125–132 Dunkley, P. R., Jarvie, P. E., Heath, J. W., Kidd, G. J., and Rostas, J. A. (1986) A rapid method for isolation of synaptosomes on Percoll gradients. Brain Res. 372, 115–129 Huettner, J. E., and Baughman, R. W. (1986) Primary culture of identified neurons from the visual cortex of postnatal rats. J. Neurosci. 6, 3044 –3060 Dubinsky, J. M. (1989) Development of inhibitory synapses among striatal neurons in vitro. J. Neurosci. 9, 3955–3965 Arrasate, M., Mitra, S., Schweitzer, E. S., Segal, M. R., and Finkbeiner, S. (2004) Inclusion body formation reduces levels of mutant huntingtin and the risk of neuronal death. Nature 431, 805– 810 Marin-Garcia, P., Sanchez-Nogueiro, J., Gomez-Villafuertes, R., Leon, D., and Miras-Portugal, M. T. (2008) Synaptic terminals from mice midbrain exhibit functional P2X7 receptor. Neuroscience 151, 361–373 Paxinos, G., and Franklin, K. B. J. (2001) The Mouse Brain in Stereotaxic Coordinates, Academic Press, New York Canals, J. M., Pineda, J. R., Torres-Peraza, J. F., Bosch, M., Martin-Ibanez, R., Munoz, M. T., Mengod, G., Ernfors, P., and Alberch, J. (2004) Brain-derived neurotrophic factor regulates the onset and severity of motor dysfunction associated with

1905

41.

42.

43. 44.

45.

46.

47. 48. 49.

1906

enkephalinergic neuronal degeneration in Huntington’s disease. J. Neurosci. 24, 7727–7739 Wang, X., Arcuino, G., Takano, T., Lin, J., Peng, W. G., Wan, P., Li, P., Xu, Q., Liu, Q. S., Goldman, S. A., and Nedergaard, M. (2004) P2X7 receptor inhibition improves recovery after spinal cord injury. Nat. Med. 10, 821– 827 Sanchez-Nogueiro, J., Marin-Garcia, P., and Miras-Portugal, M. T. (2005) Characterization of a functional P2X(7)-like receptor in cerebellar granule neurons from P2X(7) knockout mice. FEBS Lett. 579, 3783–3788 Jiang, L. H., Mackenzie, A. B., North, R. A., and Surprenant, A. (2000) Brilliant blue G selectively blocks ATP-gated rat P2X(7) receptors. Mol. Pharmacol. 58, 82– 88 Gross, E. (1961) [On induction of sarcomas with specially purified triphenylmethane dyes, Light Green, S. F., and Patent Blue AE, following repeated subcutaneous injection in rats.]. Z. Krebsforsch. 64, 287-304 Nelson, D. W., Gregg, R. J., Kort, M. E., Perez-Medrano, A., Voight, E. A., Wang, Y., Grayson, G., Namovic, M. T., DonnellyRoberts, D. L., Niforatos, W., Honore, P., Jarvis, M. F., Faltynek, C. R., and Carroll, W. A. (2006) Structure-activity relationship studies on a series of novel, substituted 1-benzyl-5-phenyltetrazole P2X7 antagonists. J. Med. Chem. 49, 3659 –3666 McGaraughty, S., Chu, K. L., Namovic, M. T., Donnelly-Roberts, D. L., Harris, R. R., Zhang, X. F., Shieh, C. C., Wismer, C. T., Zhu, C. Z., Gauvin, D. M., Fabiyi, A. C., Honore, P., Gregg, R. J., Kort, M. E., Nelson, D. W., Carroll, W. A., Marsh, K., Faltynek, C. R., and Jarvis, M. F. (2007) P2X7-related modulation of pathological nociception in rats. Neuroscience 146, 1817–1828 Sugars, K. L., and Rubinsztein, D. C. (2003) Transcriptional abnormalities in Huntington disease. Trends Genet. 19, 233–238 Riley, B. E., and Orr, H. T. (2006) Polyglutamine neurodegenerative diseases and regulation of transcription: assembling the puzzle. Genes Dev. 20, 2183–2192 Li, C. M., Campbell, S. J., Kumararatne, D. S., Hill, A. V., and Lammas, D. A. (2002) Response heterogeneity of human macrophages to ATP is associated with P2X7 receptor expression but not to polymorphisms in the P2RX7 promoter. FEBS Lett. 531, 127–131

Vol. 23

June 2009

50. 51. 52. 53. 54.

55.

56. 57.

58.

59.

Gusella, J. F., and Macdonald, M. E. (2006) Huntington’s disease: seeing the pathogenic process through a genetic lens. Trends Biochem. Sci. 31, 533–540 Riedel, T., Lozinsky, I., Schmalzing, G., and Markwardt, F. (2007) Kinetics of P2X7 receptor-operated single channels currents. Biophys. J. 92, 2377–2391 Borrell-Pages, M., Zala, D., Humbert, S., and Saudou, F. (2006) Huntington’s disease: from huntingtin function and dysfunction to therapeutic strategies. Cell. Mol. Life. Sci. 63, 2642–2660 Yong, V. W., Wells, J., Giuliani, F., Casha, S., Power, C., and Metz, L. M. (2004) The promise of minocycline in neurology. Lancet Neurol. 3, 744 –751 Hersch, S., Fink, K., Vonsattel, J. P., and Friedlander, R. M. (2003) Minocycline is protective in a mouse model of Huntington’s disease. Ann. Neurol. 54, 841; author reply 842– 843 Smith, D. L., Woodman, B., Mahal, A., Sathasivam, K., GhaziNoori, S., Lowden, P. A., Bates, G. P., and Hockly, E. (2003) Minocycline and doxycycline are not beneficial in a model of Huntington’s disease. Ann. Neurol. 54, 186 –196 Reynolds, N. (2007) Revisiting safety of minocycline as neuroprotection in Huntington’s disease. Mov. Disord. 22, 292 Humphreys, B. D., Virginio, C., Surprenant, A., Rice, J., and Dubyak, G. R. (1998) Isoquinolines as antagonists of the P2X7 nucleotide receptor: high selectivity for the human versus rat receptor homologues. Mol. Pharmacol. 54, 22–32 Donnelly-Roberts, D. L., and Jarvis, M. F. (2007) Discovery of P2X7 receptor-selective antagonists offers new insights into P2X7 receptor function and indicates a role in chronic pain states. Br. J. Pharmacol. 151, 571–579 Hisatomi, T., Enaida, H., Matsumoto, H., Kagimoto, T., Ueno, A., Hata, Y., Kubota, T., Goto, Y., and Ishibashi, T. (2006) Staining ability and biocompatibility of brilliant blue G: preclinical study of brilliant blue G as an adjunct for capsular staining. Arch. Ophthalmol. 124, 514 –519

The FASEB Journal

Received for publication September 18, 2008. Accepted for publication January 8, 2009.

DI´AZ-HERNA´NDEZ ET AL.