28 results on '"Rachit, Bakshi"'
Search Results
2. Does Serum Urate Change as Parkinson’s Disease Progresses?
- Author
-
Xiqun Chen, Rachit Bakshi, Eric A. Macklin, Yasemin G Hasimoglu, and Michael A. Schwarzschild
- Subjects
Male ,0301 basic medicine ,medicine.medical_specialty ,Parkinson's disease ,Short Communication ,Disease ,Severity of Illness Index ,Gastroenterology ,Part iii ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,disease progression ,0302 clinical medicine ,Internal medicine ,medicine ,Humans ,Longitudinal Studies ,Stage (cooking) ,Aged ,Dopamine Plasma Membrane Transport Proteins ,business.industry ,Disease progression ,longitudinal study ,Parkinson Disease ,Middle Aged ,medicine.disease ,Corpus Striatum ,Uric Acid ,Serum urate ,030104 developmental biology ,Parkinson’s disease ,urate ,Biomarker (medicine) ,Female ,Neurology (clinical) ,business ,Biomarkers ,030217 neurology & neurosurgery - Abstract
Higher serum urate concentration is associated with decreased risk of Parkinson’s disease (PD) as well as slower disease progression, but its relationship with severity of PD remains unclear. This study investigated whether changes in serum urate concentration over 5 years were associated with disease progression assessed by MDS-UPDRS Part III score, Hoehn and Yahr stage, or DaTscan imaging. Average serum urate concentration was stable over time and change in serum urate concentration did not correlate with worsening of measures of PD progression. These results suggest that serum urate concentration is not a monitoring biomarker of PD progression in early stages.
- Published
- 2020
- Full Text
- View/download PDF
3. Association of caffeine and related analytes with resistance to Parkinson disease among LRRK2 mutation carriers
- Author
-
Manuel Montalban, Xiqun Chen, Jamal I. Alkabsh, Sarah Huntwork-Rodriguez, Michael A. Schwarzschild, Giuseppe Astarita, Junhua Wang, Eric A. Macklin, Rachit Bakshi, Alberto Ascherio, Romeo Maciuca, Sonnet S. Davis, and Grace F. Crotty
- Subjects
0301 basic medicine ,Mutation ,Analyte ,business.industry ,Disease ,Pharmacology ,medicine.disease_cause ,LRRK2 ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,Metabolomics ,chemistry ,Trigonelline ,Cohort ,Medicine ,Neurology (clinical) ,Caffeine ,business ,030217 neurology & neurosurgery - Abstract
ObjectiveTo identify markers of resistance to developing Parkinson disease (PD) among LRRK2 mutation carriers (LRRK2+), we carried out metabolomic profiling in individuals with PD and unaffected controls (UC), with and without the LRRK2 mutation.MethodsPlasma from 368 patients with PD and UC in the LRRK2 Cohort Consortium (LCC), comprising 118 LRRK2+/PD+, 115 LRRK2+/UC, 70 LRRK2−/PD+, and 65 LRRK2−/UC, and CSF available from 68 of them, were analyzed by liquid chromatography with mass spectrometry. For 282 analytes quantified in plasma and CSF, we assessed differences among the 4 groups and interactions between LRRK2 and PD status, using analysis of covariance models adjusted by age, study site cohort, and sex, with p value corrections for multiple comparisons.ResultsPlasma caffeine concentration was lower in patients with PD vs UC (p < 0.001), more so among LRRK2+ carriers (by 76%) than among LRRK2− participants (by 31%), with significant interaction between LRRK2 and PD status (p = 0.005). Similar results were found for caffeine metabolites (paraxanthine, theophylline, 1-methylxanthine) and a nonxanthine marker of coffee consumption (trigonelline) in plasma, and in the subset of corresponding CSF samples. Dietary caffeine was also lower in LRRK2+/PD+ compared to LRRK2+/UC with significant interaction effect with the LRRK2+ mutation (p < 0.001).ConclusionsMetabolomic analyses of the LCC samples identified caffeine, its demethylation metabolites, and trigonelline as prominent markers of resistance to PD linked to pathogenic LRRK2 mutations, more so than to idiopathic PD. Because these analytes are known both as correlates of coffee consumption and as neuroprotectants in animal PD models, the findings may reflect their avoidance by those predisposed to develop PD or their protective effects among LRRK2 mutation carriers.
- Published
- 2020
- Full Text
- View/download PDF
4. Associations of Lower Caffeine Intake and Plasma Urate Levels with Idiopathic Parkinson’s Disease in the Harvard Biomarkers Study
- Author
-
Clemens R. Scherzer, Anne-Marie Wills, Bradley T. Hyman, Michael A. Schwarzschild, Stephen N. Gomperts, Eric A. Macklin, Rachit Bakshi, Michael T. Hayes, Alberto Ascherio, Albert Y. Hung, and John H. Growdon
- Subjects
Male ,0301 basic medicine ,medicine.medical_specialty ,Longitudinal study ,Parkinson's disease ,Gastroenterology ,Article ,Eating ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,Sex Factors ,0302 clinical medicine ,Caffeine ,Internal medicine ,Humans ,Medicine ,Aged ,business.industry ,Parkinson Disease ,Middle Aged ,medicine.disease ,Uric Acid ,Cross-Sectional Studies ,030104 developmental biology ,chemistry ,Case-Control Studies ,Cohort ,Uric acid ,Biomarker (medicine) ,Female ,Neurology (clinical) ,Caffeine intake ,business ,Body mass index ,Biomarkers ,030217 neurology & neurosurgery - Abstract
Two purines, caffeine and urate, have been associated with a reduced risk of idiopathic Parkinson's disease (PD) in multiple cohorts and populations. The Harvard Biomarkers Study (HBS) is a longitudinal study designed to accelerate the discovery and validation of molecular diagnostic and progression markers of early-stage PD. To investigate whether these 'reduced risk' factors are associated with PD within this cohort, we conducted a cross-sectional, case-control study in 566 subjects consisting of idiopathic PD patients and healthy controls. Caffeine intake as assessed by a validated questionnaire was significantly lower in idiopathic PD patients compared to healthy controls in males (mean difference -125 mg/day, p < 0.001) but not in females (mean difference -30 mg/day, p = 0.29). A strong inverse association was also observed with plasma urate levels both in males (mean difference -0.46 mg/dL, p = 0.017) and females (mean difference -0.45 mg/dL, p = 0.001). Both analyses stratified for sex and adjusted for age, body mass index, and either urate level or caffeine consumption, respectively. These results highlight the robustness of caffeine intake and urate as factors inversely associated with idiopathic PD.
- Published
- 2020
- Full Text
- View/download PDF
5. Sex differences by design and outcome in the Safety of Urate Elevation in PD (SURE-PD) trial
- Author
-
Albert Y. Hung, David S. Russell, Michael A. Schwarzschild, Eric A. Macklin, Marie Saint-Hilaire, Gary C. Curhan, Grace Bwala, Joshua M. Hare, Alice Rudolph, Christopher G. Goetz, Shamik Battacharyya, Rachit Bakshi, Robert Logan, John L. Goudreau, Sotirios A. Parashos, Alberto J. Espay, and Alberto Ascherio
- Subjects
Male ,medicine.medical_specialty ,Placebo ,Gastroenterology ,Article ,law.invention ,Randomized controlled trial ,law ,Internal medicine ,Humans ,Medicine ,Inosine ,Sex Characteristics ,business.industry ,Parkinsonism ,Follow up studies ,Parkinson Disease ,Mental Status and Dementia Tests ,medicine.disease ,Uric Acid ,Serum urate ,Antioxidant capacity ,Treatment Outcome ,Disease Progression ,Female ,Neurology (clinical) ,business ,Biomarkers ,Follow-Up Studies ,Sex characteristics ,medicine.drug - Abstract
ObjectiveTo investigate whether women and men with Parkinson disease (PD) differ in their biochemical and clinical responses to long-term treatment with inosine.MethodsThe Safety of Urate Elevation in Parkinson’s Disease (SURE-PD) trial enrolled 75 people with early PD and baseline serum urate below 6 mg/dL and randomized them to 3 double-blinded treatment arms: oral placebo or inosine titrated to produce mild (6.1–7.0 mg/dL) or moderate (7.1–8.0 mg/dL) serum urate elevation for up to 2 years. Parkinsonism, serum urate, and plasma antioxidant capacity were measured at baseline and repeatedly on treatment; CSF urate was assessed once, at 3 months. Here in secondary analyses results are stratified by sex.ResultsInosine produced an absolute increase in average serum urate from baseline that was 50% greater in women (3.0 mg/dL) than in men (2.0 mg/dL), consistent with expected lower baseline levels in women. Similarly, only among women was CSF urate significantly greater on mild or moderate inosine (+87% [p < 0.001] and +98% [p < 0.001], respectively) than on placebo (in contrast to men: +10% [p = 0.6] and +14% [p = 0.4], respectively). Women in the higher inosine dosing group showed a 7.0 Unified Parkinson’s Disease Rating Scale (UPDRS) points/year lower rate of decline vs placebo (p = 0.01). In women, slower rates of UPDRS change were associated with greater increases in serum urate (r = −0.52; p = 0.001), and with greater increases in plasma antioxidant capacity (r = −0.44; p = 0.006). No significant associations were observed in men.ConclusionsInosine produced greater increases in serum and CSF urate in women compared to men in the SURE-PD trial, consistent with the study's design and with preliminary evidence for slower clinical decline in early PD among women treated with urate-elevating doses of inosine.Clinicaltrials.gov identifierNCT00833690.Classification of evidenceThis study provides Class II evidence that inosine produced greater urate elevation in women than men and may slow PD progression in women.
- Published
- 2019
- Full Text
- View/download PDF
6. Higher urate in LRRK2 mutation carriers resistant to Parkinson disease
- Author
-
Eric A. Macklin, Michael A. Schwarzschild, Rachit Bakshi, Grace F. Crotty, Ning Xia, Robert Logan, Alberto Ascherio, Xiqun Chen, Ellen Zhang, and Musab M. Zorlu
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Plasma samples ,business.industry ,Disease ,Disease pathogenesis ,Age and sex ,Gastroenterology ,Penetrance ,LRRK2 ,Mean difference ,nervous system diseases ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Neurology ,Internal medicine ,Cohort ,medicine ,Neurology (clinical) ,business ,030217 neurology & neurosurgery - Abstract
OBJECTIVE LRRK2 mutations, the most common genetic cause of Parkinson disease (PD), display incomplete penetrance, indicating the importance of other genetic and environmental influences on disease pathogenesis in LRRK2 mutation carriers. The present study investigates whether urate, an antioxidant, Nrf2 activator, and inverse risk factor for idiopathic PD, is one such candidate biomarker of PD risk modulation in pathogenic LRRK2 mutation carriers. METHODS Banked plasma samples or urate levels were obtained for 3 cohorts of age- and sex-matched subjects with and without a known LRRK2 mutation in PD and unaffected controls to conduct a pilot study of 192 subjects from the LRRK2 Cohort Consortium (LCC) and 2 validation studies of 380 additional subjects from the LCC and 922 subjects from the Parkinson's Progression Markers Initiative. Urate levels were compared by multiple regression between subjects with and without a PD diagnosis conditional on LRRK2 status, controlling for age and sex. RESULTS Nonmanifesting LRRK2 mutation carriers had significantly higher levels of urate than those who developed PD in each of the 3 independent cohorts. A meta-analysis demonstrated an adjusted mean difference of 0.62 mg/dL (p
- Published
- 2019
- Full Text
- View/download PDF
7. Association of caffeine and related analytes with resistance to Parkinson disease among
- Author
-
Grace F, Crotty, Romeo, Maciuca, Eric A, Macklin, Junhua, Wang, Manuel, Montalban, Sonnet S, Davis, Jamal I, Alkabsh, Rachit, Bakshi, Xiqun, Chen, Alberto, Ascherio, Giuseppe, Astarita, Sarah, Huntwork-Rodriguez, and Michael A, Schwarzschild
- Subjects
Male ,Heterozygote ,Parkinson Disease ,Middle Aged ,Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 ,Mass Spectrometry ,Article ,nervous system diseases ,Cohort Studies ,Alkaloids ,Neuroprotective Agents ,Theophylline ,Caffeine ,Xanthines ,Humans ,Metabolomics ,Female ,Aged ,Chromatography, Liquid - Abstract
Objective To identify markers of resistance to developing Parkinson disease (PD) among LRRK2 mutation carriers (LRRK2+), we carried out metabolomic profiling in individuals with PD and unaffected controls (UC), with and without the LRRK2 mutation. Methods Plasma from 368 patients with PD and UC in the LRRK2 Cohort Consortium (LCC), comprising 118 LRRK2+/PD+, 115 LRRK2+/UC, 70 LRRK2−/PD+, and 65 LRRK2−/UC, and CSF available from 68 of them, were analyzed by liquid chromatography with mass spectrometry. For 282 analytes quantified in plasma and CSF, we assessed differences among the 4 groups and interactions between LRRK2 and PD status, using analysis of covariance models adjusted by age, study site cohort, and sex, with p value corrections for multiple comparisons. Results Plasma caffeine concentration was lower in patients with PD vs UC (p < 0.001), more so among LRRK2+ carriers (by 76%) than among LRRK2− participants (by 31%), with significant interaction between LRRK2 and PD status (p = 0.005). Similar results were found for caffeine metabolites (paraxanthine, theophylline, 1-methylxanthine) and a nonxanthine marker of coffee consumption (trigonelline) in plasma, and in the subset of corresponding CSF samples. Dietary caffeine was also lower in LRRK2+/PD+ compared to LRRK2+/UC with significant interaction effect with the LRRK2+ mutation (p < 0.001). Conclusions Metabolomic analyses of the LCC samples identified caffeine, its demethylation metabolites, and trigonelline as prominent markers of resistance to PD linked to pathogenic LRRK2 mutations, more so than to idiopathic PD. Because these analytes are known both as correlates of coffee consumption and as neuroprotectants in animal PD models, the findings may reflect their avoidance by those predisposed to develop PD or their protective effects among LRRK2 mutation carriers.
- Published
- 2020
8. Pilot trial of inosine to elevate urate levels in amyotrophic lateral sclerosis
- Author
-
Eva-Maria Ratai, Mark Levine-Weinberg, Sabrina Paganoni, Daniela L. Grasso, Eric A. Macklin, Samad Jahandideh, Ovidiu C. Andronesi, Merit Cudkowicz, Christopher T. Breen, Anne-Marie Wills, Rachit Bakshi, Albert A. Taylor, Michael A. Schwarzschild, Katharine Nicholson, James Chan, Danielle Beaulieu, and David L. Ennist
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Neuroprotection ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Amyotrophic lateral sclerosis ,Adverse effect ,Inosine ,Feeding tube ,Research Articles ,business.industry ,General Neuroscience ,16. Peace & justice ,medicine.disease ,3. Good health ,Gout ,Clinical trial ,030104 developmental biology ,Tolerability ,Neurology (clinical) ,business ,030217 neurology & neurosurgery ,Research Article ,medicine.drug - Abstract
Objective To test the safety, tolerability, and urate‐elevating capability of the urate precursor inosine taken orally or by feeding tube in people with amyotrophic lateral sclerosis (ALS). Methods This was a pilot, open‐label trial in 25 participants with ALS. Treatment duration was 12 weeks. The dose of inosine was titrated at pre‐specified time points to elevate serum urate levels to 7–8 mg/dL. Primary outcomes were safety (as assessed by the occurrence of adverse events [AEs]) and tolerability (defined as the ability to complete the 12‐week study on study drug). Secondary outcomes included biomarkers of oxidative stress and damage. As an exploratory analysis, observed outcomes were compared with a virtual control arm built using prediction algorithms to estimate ALSFRS‐R scores. Results Twenty‐four out of 25 participants (96%) completed 12 weeks of study drug treatment. One participant was unable to comply with study visits and was lost to follow‐up. Serum urate rose to target levels in 6 weeks. No serious AEs attributed to study drug and no AEs of special concern, such as urolithiasis and gout, occurred. Selected biomarkers of oxidative stress and damage had significant changes during the study period. Observed changes in ALSFRS‐R did not differ from baseline predictions. Interpretation Inosine appeared safe, well tolerated, and effective in raising serum urate levels in people with ALS. These findings, together with epidemiological observations and preclinical data supporting a neuroprotective role of urate in ALS models, provide the rationale for larger clinical trials testing inosine as a potential disease‐modifying therapy for ALS.
- Published
- 2018
- Full Text
- View/download PDF
9. Urate mitigates oxidative stress and motor neuron toxicity of astrocytes derived from ALS-linked SOD1 mutant mice
- Author
-
Kaly A. Mueller, Michael A. Schwarzschild, Rachit Bakshi, Eric J. Granucci, Yuehang Xu, Ghazaleh Sadri-Vakili, Sabrina Paganoni, and Xiqun Chen
- Subjects
0301 basic medicine ,Antioxidant ,Superoxide ,medicine.medical_treatment ,SOD1 ,Cell Biology ,Biology ,Motor neuron ,medicine.disease ,medicine.disease_cause ,Neuroprotection ,Cell biology ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,chemistry ,medicine ,Amyotrophic lateral sclerosis ,Molecular Biology ,030217 neurology & neurosurgery ,Oxidative stress ,Astrocyte - Abstract
Dominant mutations in an antioxidant enzyme superoxide dismutase-1 (SOD1) cause amyotrophic lateral sclerosis (ALS), an adult-onset neurodegenerative disease characterized by loss of motor neurons. Oxidative stress has also been linked to many of the neurodegenerative diseases and is likely a central mechanism of motor neuron death in ALS. Astrocytes derived from mutant SOD1G93A mouse models or patients play a significant role in the degeneration of spinal motor neurons in ALS through a non-cell-autonomous process. Here we characterize the neuroprotective effects and mechanisms of urate (a.k.a. uric acid), a major endogenous antioxidant and a biomarker of favorable ALS progression rates, in a cellular model of ALS. Our results demonstrate a significant protective effect of urate against motor neuron injury evoked by mutant astrocytes derived from SOD1G93A mice or hydrogen peroxide induced oxidative stress. Overall, these results implicate astrocyte dependent protective effect of urate in a cellular model of ALS. These findings together with our biomarker data may advance novel targets for treating motor neuron disease.
- Published
- 2018
- Full Text
- View/download PDF
10. Distinguishing between biochemical and cellular function: Are there peptide signatures for cellular function of proteins?
- Author
-
Ankita Narang, Rachit Bakshi, K K Bhattacharyya, Shruti Jain, and Vani Brahmachari
- Subjects
0301 basic medicine ,Genetics ,biology ,Kinase ,Computational biology ,Biochemical Activity ,Biochemistry ,Homology (biology) ,Receptor tyrosine kinase ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Structural Biology ,biology.protein ,Human genome ,Receptor ,Homeotic gene ,Molecular Biology ,Gene ,030217 neurology & neurosurgery - Abstract
The genome annotation and identification of gene function depends on conserved biochemical activity. However, in the cell, proteins with the same biochemical function can participate in different cellular pathways and cannot complement one another. Similarly, two proteins of very different biochemical functions are put in the same class of cellular function; for example, the classification of a gene as an oncogene or a tumour suppressor gene is not related to its biochemical function, but is related to its cellular function. We have taken an approach to identify peptide signatures for cellular function in proteins with known biochemical function. ATPases as a test case, we classified ATPases (2360 proteins) and kinases (517 proteins) from the human genome into different cellular function categories such as transcriptional, replicative, and chromatin remodelling proteins. Using publicly available tool, MEME, we identify peptide signatures shared among the members of a given category but not between cellular functional categories; for example, no motif sharing is seen between chromatin remodelling and transporter ATPases, similarly between receptor Serine/Threonine Kinase and Receptor Tyrosine Kinase. There are motifs shared within each category with significant E value and high occurrence. This concept of signature for cellular function was applied to developmental regulators, the polycomb and trithorax proteins which led to the prediction of the role of INO80, a chromatin remodelling protein, in development. This has been experimentally validated earlier for its role in homeotic gene regulation and its interaction with regulatory complexes like the Polycomb and Trithorax complex. Proteins 2017; 85:682-693. © 2016 Wiley Periodicals, Inc.
- Published
- 2017
- Full Text
- View/download PDF
11. Oral Inosine Persistently Elevates Plasma antioxidant capacity in Parkinson's disease
- Author
-
Alberto Ascherio, Robert Logan, Michael A. Schwarzschild, Shamik Bhattacharyya, Eric A. Macklin, and Rachit Bakshi
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Antioxidant ,Parkinson's disease ,medicine.medical_treatment ,Urinary system ,Urine ,medicine.disease_cause ,Placebo ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Internal medicine ,medicine ,Inosine ,business.industry ,medicine.disease ,030104 developmental biology ,Endocrinology ,Neurology ,chemistry ,Biochemistry ,Uric acid ,Neurology (clinical) ,business ,030217 neurology & neurosurgery ,Oxidative stress ,medicine.drug - Abstract
Introduction Higher serum urate predicts slower progression in PD. The aim of this work was to assess whether oral inosine alters antioxidant capacity of plasma or CSF or urinary markers of oxidative injury in early PD. Methods We assayed plasma and CSF antioxidant capacity by ferric-reducing antioxidant power and measured DNA oxidation adduct 8-hydroxydeoxyguanosine from urine in Safety of URate Elevation in PD, a randomized, placebo-controlled trial of oral inosine assessing safety of elevating serum urate from
- Published
- 2016
- Full Text
- View/download PDF
12. A Feed-Forward Circuit of EndogenousPGC-1αandEstrogen Related Receptor αRegulates the Neuronal Electron Transport Chain
- Author
-
Zhixiang Liao, Clemens R. Scherzer, Rachit Bakshi, and Shuchi Mittal
- Subjects
0301 basic medicine ,Article Subject ,Neuroscience (miscellaneous) ,Regulator ,Endogeny ,Peroxisome ,Biology ,lcsh:RC346-429 ,Cell biology ,03 medical and health sciences ,Psychiatry and Mental health ,Estrogen-related receptor ,030104 developmental biology ,Biochemistry ,Transcription (biology) ,Coactivator ,Gene expression ,Neurology (clinical) ,Receptor ,lcsh:Neurology. Diseases of the nervous system ,Research Article - Abstract
Peroxisome proliferator-activated receptor γcoactivator 1α(PGC-1α) is a central regulator of cellular and mitochondrial metabolism. Cellular bioenergetics are critically important in “energy-guzzling” neurons, but the components and wiring of the transcriptional circuit through whichPGC-1αregulates the neuronal electron transport chain have not been established. This information may be vital for restoring neuronal bioenergetics gene expression that is compromised during incipient Parkinson’s neuropathology and in aging-dependent brain diseases. Here we delineate a neuronal transcriptional circuit controlled by endogenousPGC-1α. We show that a feed-forward circuit of endogenous neuronalPGC-1αand the orphan nuclear estrogen-related receptorα(ERRα) activates the nuclear-encoded mitochondrial electron transport chain.PGC-1αnot onlytrans-activated expression ofERRα, but also coactivatedERRαtarget genes in complexes I, II, IV, and V of the neuronal electron transport chain via association with evolutionary conservedERRαpromoter binding motifs. Chemical activation of this transcriptional program induced transcription of the neuronal electron transport chain. These data highlight a neuronal transcriptional circuit regulated byPGC-1αthat can be therapeutically targeted for Parkinson’s and other neurodegenerative diseases.
- Published
- 2016
- Full Text
- View/download PDF
13. Urate mitigates oxidative stress and motor neuron toxicity of astrocytes derived from ALS-linked SOD1
- Author
-
Rachit, Bakshi, Yuehang, Xu, Kaly A, Mueller, Xiqun, Chen, Eric, Granucci, Sabrina, Paganoni, Ghazaleh, Sadri-Vakili, and Michael A, Schwarzschild
- Subjects
Motor Neurons ,Mice ,Oxidative Stress ,Superoxide Dismutase-1 ,Astrocytes ,Culture Media, Conditioned ,Amyotrophic Lateral Sclerosis ,Mutation ,Animals ,Antioxidants ,Cells, Cultured ,Cell Line ,Uric Acid - Abstract
Dominant mutations in an antioxidant enzyme superoxide dismutase-1 (SOD1) cause amyotrophic lateral sclerosis (ALS), an adult-onset neurodegenerative disease characterized by loss of motor neurons. Oxidative stress has also been linked to many of the neurodegenerative diseases and is likely a central mechanism of motor neuron death in ALS. Astrocytes derived from mutant SOD1
- Published
- 2018
14. Reply to 'Mitochondrial DNA deletions discriminate affected from unaffected LRRK 2 mutation carriers'
- Author
-
Eric A. Macklin, Michael A. Schwarzschild, and Rachit Bakshi
- Subjects
Genetics ,Neurology ,Mutation (genetic algorithm) ,Mitochondrial DNA deletions ,Neurology (clinical) ,Biology ,LRRK2 - Published
- 2019
- Full Text
- View/download PDF
15. Protection by inosine in a cellular model of Parkinson’s disease
- Author
-
Sara Cipriani, Michael A. Schwarzschild, and Rachit Bakshi
- Subjects
Free Radicals ,Cell Survival ,medicine.disease_cause ,Article ,Cell Line ,Antiparkinson Agents ,Protein Carbonylation ,Mice ,chemistry.chemical_compound ,Parkinsonian Disorders ,medicine ,Animals ,Purine metabolism ,Inosine ,Cells, Cultured ,Nitrites ,Hypoxanthine ,Chemistry ,Dopaminergic Neurons ,General Neuroscience ,Hydrogen Peroxide ,Xanthine ,Adenosine ,Coculture Techniques ,Rats ,Uric Acid ,Mice, Inbred C57BL ,Oxidative Stress ,Neuroprotective Agents ,Biochemistry ,Purines ,Astrocytes ,Uric acid ,Nucleoside ,Oxidative stress ,medicine.drug - Abstract
Inosine (hypoxanthine 9-beta-D-ribofuranoside), a purine nucleoside with multiple intracellular roles, also serves as an extracellular modulatory signal. On neurons, it can produce anti-inflammatory and trophic effects that confer protection against toxic influences in vivo and in vitro. The protective effects of inosine treatment might also be mediated by its metabolite urate. Urate in fact possesses potent antioxidant properties and has been reported to be protective in preclinical Parkinson's disease (PD) studies and to be an inverse risk factor for both the development and progression of PD. In this study we assessed whether inosine might protect rodent MES 23.5 dopaminergic cell line from oxidative stress in a cellular model of PD, and whether its effects could be attributed to urate. MES 23.5 cells cultured alone or in presence of enriched murine astroglial cultures MES 23.5-astrocytes co-cultures were pretreated with inosine (0.1-100 μM) for 24 h before addition of the oxidative stress inducer H₂O₂ (200 μM). Twenty-four hours later, cell viability was quantified by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay or immunocytochemistry in pure and MES 23.5-astrocytes co-cultures, respectively. H₂O₂-toxic effect on dopaminergic cells was reduced when they were cultured with astrocytes, but not when they were cultured alone. Moreover, in MES 23.5-astrocytes co-cultures, indicators of free radical generation and oxidative damage, evaluated by nitrite (NO₂(-)) release and protein carbonyl content, respectively, were attenuated. Conditioned medium experiments indicated that the protective effect of inosine relies on the release of a protective factor from inosine-stimulated astrocytes. Purine levels were measured in the cellular extract and conditioned medium using high-performance liquid chromatography (HPLC) method. Urate concentration was not significantly increased by inosine treatment however there was a significant increase in levels of other purine metabolites, such as adenosine, hypoxanthine and xanthine. In particular, in MES 23.5-astrocytes co-cultures, inosine medium content was reduced by 99% and hypoxanthine increased by 127-fold. Taken together these data raise the possibility that inosine might have a protective effect in PD that is independent of any effects mediated through its metabolite urate.
- Published
- 2014
- Full Text
- View/download PDF
16. Distinguishing between biochemical and cellular function: Are there peptide signatures for cellular function of proteins?
- Author
-
Shruti, Jain, Kausik, Bhattacharyya, Rachit, Bakshi, Ankita, Narang, and Vani, Brahmachari
- Subjects
Adenosine Triphosphatases ,Genome, Human ,Amino Acid Motifs ,DNA Helicases ,Gene Expression Regulation, Developmental ,Polycomb-Group Proteins ,Biological Transport ,Molecular Sequence Annotation ,Histone-Lysine N-Methyltransferase ,Chromatin Assembly and Disassembly ,Chromatin ,DNA-Binding Proteins ,Gene Ontology ,ATPases Associated with Diverse Cellular Activities ,Humans ,Protein Kinases ,Myeloid-Lymphoid Leukemia Protein - Abstract
The genome annotation and identification of gene function depends on conserved biochemical activity. However, in the cell, proteins with the same biochemical function can participate in different cellular pathways and cannot complement one another. Similarly, two proteins of very different biochemical functions are put in the same class of cellular function; for example, the classification of a gene as an oncogene or a tumour suppressor gene is not related to its biochemical function, but is related to its cellular function. We have taken an approach to identify peptide signatures for cellular function in proteins with known biochemical function. ATPases as a test case, we classified ATPases (2360 proteins) and kinases (517 proteins) from the human genome into different cellular function categories such as transcriptional, replicative, and chromatin remodelling proteins. Using publicly available tool, MEME, we identify peptide signatures shared among the members of a given category but not between cellular functional categories; for example, no motif sharing is seen between chromatin remodelling and transporter ATPases, similarly between receptor Serine/Threonine Kinase and Receptor Tyrosine Kinase. There are motifs shared within each category with significant E value and high occurrence. This concept of signature for cellular function was applied to developmental regulators, the polycomb and trithorax proteins which led to the prediction of the role of INO80, a chromatin remodelling protein, in development. This has been experimentally validated earlier for its role in homeotic gene regulation and its interaction with regulatory complexes like the Polycomb and Trithorax complex. Proteins 2017; 85:682-693. © 2016 Wiley Periodicals, Inc.
- Published
- 2016
17. Oral Inosine Persistently Elevates Plasma antioxidant capacity in Parkinson's disease
- Author
-
Shamik, Bhattacharyya, Rachit, Bakshi, Robert, Logan, Alberto, Ascherio, Eric A, Macklin, and Michael A, Schwarzschild
- Subjects
Male ,Oxidative Stress ,Treatment Outcome ,Disease Progression ,Humans ,Female ,Parkinson Disease ,Middle Aged ,Antioxidants ,Inosine ,Uric Acid - Abstract
Higher serum urate predicts slower progression in PD. The aim of this work was to assess whether oral inosine alters antioxidant capacity of plasma or CSF or urinary markers of oxidative injury in early PD.We assayed plasma and CSF antioxidant capacity by ferric-reducing antioxidant power and measured DNA oxidation adduct 8-hydroxydeoxyguanosine from urine in Safety of URate Elevation in PD, a randomized, placebo-controlled trial of oral inosine assessing safety of elevating serum urate from6 mg/dL to 6.1-7.0 or 7.1-8.0 mg/dL in patients with early PD.At 6 months, antioxidant capacity was 29% higher among mild and 43% higher among moderate group participants compared to placebo and correlated with change in serum urate (r = 0.86) and inversely with rate of clinical decline (r = -0.26). CSF antioxidant capacity and urine 8-hydroxydeoxyguanosine did not differ.The findings demonstrate a dose-dependent, persistent elevation of plasma antioxidant capacity from oral inosine of potential therapeutic relevance.
- Published
- 2015
18. Blockade of metabotropic glutamate receptor 5 protects against DNA damage in a rotenone-induced Parkinson's disease model
- Author
-
Hui Yang, Hong Zhang, Shu Ting Wang, Li Gu, Li Liu, Qian Zhang, Ning Xia, Rachit Bakshi, and Hui Min Yang
- Subjects
Male ,DNA Repair ,DNA damage ,DNA repair ,Receptor, Metabotropic Glutamate 5 ,Blotting, Western ,Excitotoxicity ,Fluorescent Antibody Technique ,Enzyme-Linked Immunosorbent Assay ,Biology ,Pharmacology ,medicine.disease_cause ,Biochemistry ,Cell Line ,Rats, Sprague-Dawley ,Parkinsonian Disorders ,Physiology (medical) ,Rotenone ,medicine ,Animals ,Membrane Potential, Mitochondrial ,Metabotropic glutamate receptor 5 ,Uncoupling Agents ,Glutamate receptor ,Neurotoxicity ,medicine.disease ,Molecular biology ,Immunohistochemistry ,Rats ,Comet assay ,Disease Models, Animal ,Oxidative Stress ,Comet Assay ,Oxidative stress ,DNA Damage - Abstract
Glutamate excitotoxicity contributes to the development of Parkinson's disease (PD) and pharmacological blockade of metabotropic glutamate receptor 5 (mGluR5) has beneficial anti-akinetic effects in animal models of PD; however, the mechanism by which these antagonists alleviate PD symptoms is largely unknown. In our study, the effects of mGluR5 inhibition on DNA damage were investigated in a rotenone-induced model of PD. We first found that the selective mGluR5 antagonist, 2-methyl-6- (phenylethynyl) pyridine, prevented rotenone-induced DNA damage in MN9D dopaminergic neurons through a mechanism involving the downregulation of intracellular calcium release which was associated with a reduction in endoplasmic reticulum stress and reactive oxygen species (ROS)-related mitochondrial dysfunction. Interestingly, the ROS-related mitochondrial dysfunction was accompanied by an increase in expression of the antioxidant protein, Trx2. Treatment of cells with the calcium chelating agent 1,2-bis-(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid or the ROS scavenger N-acetyl-L-cysteine, also reduced rotenone-induced DNA damage, while transfection of a dominant-negative form of Trx2 increased it. In addition, mGluR5 inhibition altered the expression profiles of proteins involved in DNA repair activity. Specifically, the expression of phosphorylated ERK (p-ERK) and CREB, as well as APE1 and Rad51 were elevated after rotenone stimulation and were subsequently downregulated following blockade of mGluR5. These findings were confirmed in vivo in a rotenone-induced rat model of PD. Inhibition of mGluR5 protected against neurotoxicity by mitigating oxidative stress-related DNA damage associated with 8-hydroxy-2'-deoxyguanosine production and also reduced p-ERK activity and Trx2 expression. These findings provide a novel link between mGluR5 and DNA damage in a model of PD, and reveal a potential mechanism by which mGluR5 mediates DNA damage in neurodegenerative diseases.
- Published
- 2015
19. Purines in Parkinson’s: Adenosine A2A Receptors and Urate as Targets for Neuroprotection
- Author
-
Michael A. Schwarzschild, Rachit Bakshi, and Robert Logan
- Subjects
G protein ,Chemistry ,medicine ,Adenosine A2A receptor ,Pharmacology ,Purinergic signalling ,Purine metabolism ,Adenosine receptor ,Neuroprotection ,Adenosine ,medicine.drug ,G protein-coupled receptor - Abstract
Purines are essential constituents of all living cells. The nucleoside adenosine is not only a precursor of ATP and cyclic AMP but is also released by a wide variety of cells under various physiological and pathological conditions. In mammals, adenosine acts on four subtypes of guanine nucleotide binding protein (G protein)-coupled receptor (GPCR)—A1, A2A, A2B and A3. Among these the adenosine A2A receptor has emerged as a particularly attractive target of therapeutics development for Parkinson’s disease (PD), in part because it is highly expressed in brain regions innervated by the dopaminergic neurons that degenerate in PD. Urate (also known as uric acid —2,6,8-trioxypurine) is the most abundant plasma antioxidant as well as the end product of purine metabolism in humans. Emerging clinical, epidemiological, and laboratory evidence has identified urate as a potential neuroprotectant for the treatment of PD. The primary intent of this review is to explore the neuroprotective effects of adenosine receptor antagonists and urate and their therapeutic potential in PD with particular attention to epidemiological and preclinical findings linking these purines to PD and other neurodegenerative diseases. This review also summarizes current clinical development of purines as candidate neuroprotectants.
- Published
- 2015
- Full Text
- View/download PDF
20. In silico characterization of the INO80 subfamily of SWI2/SNF2 chromatin remodeling proteins
- Author
-
Debasis Dash, Rachit Bakshi, Tulika Prakash, and Vani Brahmachari
- Subjects
Saccharomyces cerevisiae Proteins ,Subfamily ,Chromosomal Proteins, Non-Histone ,In silico ,Protein subunit ,Molecular Sequence Data ,Biophysics ,Biology ,Biochemistry ,Chromatin remodeling ,Sequence Analysis, Protein ,Animals ,Humans ,Ino80 complex ,Amino Acid Sequence ,Epigenetics ,SMARCB1 ,Databases, Protein ,Molecular Biology ,Phylogeny ,Genetics ,Sequence Homology, Amino Acid ,DNA Helicases ,Nuclear Proteins ,Cell Biology ,Chromatin Assembly and Disassembly ,DNA-Binding Proteins ,Sequence Alignment ,Function (biology) ,Transcription Factors - Abstract
Proteins belonging to SNF2 family of DNA dependent ATPases are important members of the chromatin remodeling complexes that are implicated in epigenetic control of gene expression. The yeast Ino80, the catalytic ATPase subunit of the INO80 complex, is the most recently described member of the SNF2 family. Outside the conserved ATPase domain, it has very little similarity with other well-characterized SNF2 proteins hence it is believed to represent a new subfamily. We have identified new members of this subfamily in different organisms and have detected characteristic features of this subfamily. Using various data mining tools we have identified a new, previously undetected domain in all members of this subfamily. This domain designated DBINO is characteristic of the INO80 subfamily and is predicted to have DNA-binding function. The presence of this domain in all the INO80 subfamily proteins from different organisms suggests its conserved function in evolution.
- Published
- 2004
- Full Text
- View/download PDF
21. Association of α-synuclein gene expression with Parkinson’s disease is attenuated with higher serum urate in the PPMI cohort
- Author
-
Clemens R. Scherzer, Eric A. Macklin, Rachit Bakshi, Kathryn C. Fitzgerald, Michael A. Schwarzschild, and Alberto Ascherio
- Subjects
medicine.medical_specialty ,Parkinson's disease ,business.industry ,medicine.disease ,Serum urate ,Endocrinology ,Neurology ,Internal medicine ,Gene expression ,Cohort ,medicine ,α synuclein ,Neurology (clinical) ,Geriatrics and Gerontology ,business - Published
- 2016
- Full Text
- View/download PDF
22. The human SWI/SNF complex associates with RUNX1 to control transcription of hematopoietic target genes
- Author
-
Mohammad Q. Hassan, Jitesh Pratap, Anthony N. Imbalzano, Jane B. Lian, Gary S. Stein, Andre J. Van Wijnen, Martin Montecino, Rachit Bakshi, and Janet L. Stein
- Subjects
Physiology ,Chromosomal Proteins, Non-Histone ,Clinical Biochemistry ,Biology ,Chromatin remodeling ,Article ,Histones ,Jurkat Cells ,hemic and lymphatic diseases ,Humans ,Epigenetics ,Chromatin structure remodeling (RSC) complex ,SMARCB1 ,Promoter Regions, Genetic ,Transcription factor ,DNA Helicases ,Granulocyte-Macrophage Colony-Stimulating Factor ,Nuclear Proteins ,Cell Biology ,SMARCB1 Protein ,Molecular biology ,SWI/SNF ,Chromatin ,Hematopoiesis ,DNA-Binding Proteins ,Gene Expression Regulation ,Core Binding Factor Alpha 2 Subunit ,SMARCA4 ,biology.protein ,Interleukin-3 ,Transcription Factors - Abstract
The acute myeloid leukemia 1 (AML1, RUNX1) transcription factor is a key regulator of hematopoietic differentiation that forms multi-protein complexes with co-regulatory proteins. These complexes are assembled at target gene promoters in nuclear microenvironments to mediate phenotypic gene expression and chromatin related epigenetic modifications. Here, immunofluorescence microscopy and biochemical assays are used to show that RUNX1 associates with the human ATP-dependent SWI/SNF chromatin remodeling complex. The SWI/SNF subunits BRG1 and INI1 bind in vivo to RUNX1 target gene promoters (e.g., GMCSF, IL3, MCSF-R, MIP and p21). These interactions correlate with histone modifications characteristic of active chromatin, including acetylated H4 and dimethylated H3 Lysine 4. Down-regulation of RUNX1 by RNA interference diminishes the binding of BRG1 and INI1 at selected target genes. Taken together, our findings indicate that RUNX1 interacts with the human SWI/SNF complex to control hematopoietic-specific gene expression.
- Published
- 2010
23. Subnuclear Localization and Intranuclear Trafficking of Transcription Factors
- Author
-
Sayyed K. Zaidi, Ricardo F. Medina, Andre J. van Wijnen, Krishna P. Kota, Amjad Javed, Gary S. Stein, Daniel W. Young, Jane B. Lian, Jeffrey A. Nickerson, Martin Montecino, Syed A. Ali, Rachit Bakshi, Shirwin M. Pockwinse, and Janet L. Stein
- Subjects
Microscopy ,Cell Culture Techniques ,Intermediate Filaments ,Intracellular Space ,Computational Biology ,Fluorescence recovery after photobleaching ,Intranuclear Space ,Biology ,Compartmentalization (psychology) ,Nuclear matrix ,Article ,Transport protein ,Cell biology ,Protein Transport ,Gene expression ,Cell Adhesion ,Animals ,Nuclear Matrix ,Cell adhesion ,Transcription factor ,Metaphase ,Fluorescence Recovery After Photobleaching ,Transcription Factors - Abstract
Nuclear microenvironments are architecturally organized subnuclear sites where the regulatory machinery for gene expression, replication, and repair resides. This compartmentalization is necessary to attain required stoichiometry for organization and assembly of regulatory complexes for combinatorial control. Combined and methodical application of molecular, cellular, biochemical, and in vivo genetic approaches is required to fully understand complexities of biological control. Here we provide methodologies to characterize nuclear organization of regulatory machinery by in situ immunofluorescence microscopy.
- Published
- 2010
- Full Text
- View/download PDF
24. The leukemogenic t(8;21) fusion protein AML1-ETO controls rRNA genes and associates with nucleolar-organizing regions at mitotic chromosomes
- Author
-
Sayyed K. Zaidi, Mohammad Q. Hassan, Gary S. Stein, Sandhya Pande, Janet L. Stein, Andre J. Van Wijnen, Daniel W. Young, Rachit Bakshi, Jane B. Lian, and Martin Montecino
- Subjects
Oncogene Proteins, Fusion ,Nucleolus ,Chromosomes, Human, Pair 21 ,Fluorescent Antibody Technique ,Mitosis ,Biology ,Translocation, Genetic ,Article ,chemistry.chemical_compound ,RUNX1 Translocation Partner 1 Protein ,Transcription (biology) ,hemic and lymphatic diseases ,Cell Line, Tumor ,RNA polymerase I ,Nucleolus Organizer Region ,Humans ,Transcription factor ,neoplasms ,RNA ,Genes, rRNA ,Cell Biology ,Molecular biology ,Leukemia, Myeloid, Acute ,RUNX1 ,chemistry ,DNA methylation ,Core Binding Factor Alpha 2 Subunit ,Cell Nucleolus ,Chromosomes, Human, Pair 8 - Abstract
RUNX1/AML1 is required for definitive hematopoiesis and is frequently targeted by chromosomal translocations in acute myeloid leukemia (AML). The t(8;21)-related AML1-ETO fusion protein blocks differentiation of myeloid progenitors. Here, we show by immunofluorescence microscopy that during interphase, endogenous AML1-ETO localizes to nuclear microenvironments distinct from those containing native RUNX1/AML1 protein. At mitosis, we clearly detect binding of AML1-ETO to nucleolar-organizing regions in AML-derived Kasumi-1 cells and binding of RUNX1/AML1 to the same regions in Jurkat cells. Both RUNX1/AML1 and AML1-ETO occupy ribosomal DNA repeats during interphase, as well as interact with the endogenous RNA Pol I transcription factor UBF1. Promoter cytosine methylation analysis indicates that RUNX1/AML1 binds to rDNA repeats that are more highly CpG methylated than those bound by AML1-ETO. Downregulation by RNA interference reveals that RUNX1/AML1 negatively regulates rDNA transcription, whereas AML1-ETO is a positive regulator in Kasumi-1 cells. Taken together, our findings identify a novel role for the leukemia-related AML1-ETO protein in epigenetic control of cell growth through upregulation of ribosomal gene transcription mediated by RNA Pol I, consistent with the hyper-proliferative phenotype of myeloid cells in AML patients.
- Published
- 2008
25. In situ nuclear organization of regulatory machinery
- Author
-
Shirwin M, Pockwinse, Sayyed K, Zaidi, Ricardo F, Medina, Rachit, Bakshi, Krishna P, Kota, Syed A, Ali, Daniel W, Young, Jeffery A, Nickerson, Amjad, Javed, Martin, Montecino, Andre J, van Wijnen, Jane B, Lian, Janet L, Stein, and Gary S, Stein
- Subjects
Cell Nucleus ,Gene Expression Regulation ,Microscopy, Fluorescence ,Cell Cycle ,Cell Culture Techniques ,Intermediate Filaments ,Animals ,Humans ,Cells, Cultured ,Chromosomes ,Fluorescence Recovery After Photobleaching - Abstract
Regulatory machinery for gene expression, replication, and repair are architecturally organized in nuclear microenvironments. This compartmentalization provides threshold concentrations of macromolecules for the organization and assembly of regulatory complexes for combinatorial control. A mechanistic under standing of biological control requires the combined application of molecular, cellular, biochemical, and in vivo genetic approaches. This chapter provides methodologies to characterize nuclear organization of regulatory machinery by in situ immunofluorescence microscopy.
- Published
- 2008
26. In Situ Nuclear Organization of Regulatory Machinery
- Author
-
Jane B. Lian, Jeffrey A. Nickerson, Shirwin M. Pockwinse, Gary S. Stein, Amjad Javed, Andre J. van Wijnen, Martin Montecino, Ricardo F. Medina, Syed A. Ali, Sayyed K. Zaidi, Janet L. Stein, Krishna P. Kota, Daniel W. Young, and Rachit Bakshi
- Subjects
In situ ,Gene expression ,Nuclear organization ,Immunofluorescence Microscopy ,Biology ,Compartmentalization (psychology) ,Nuclear matrix ,Cell biology - Abstract
Regulatory machinery for gene expression, replication, and repair are architecturally organized in nuclear microenvironments. This compartmentalization provides threshold concentrations of macromolecules for the organization and assembly of regulatory complexes for combinatorial control. A mechanistic under standing of biological control requires the combined application of molecular, cellular, biochemical, and in vivo genetic approaches. This chapter provides methodologies to characterize nuclear organization of regulatory machinery by in situ immunofluorescence microscopy.
- Published
- 2008
- Full Text
- View/download PDF
27. Characterization of a human SWI2/SNF2 like protein hINO80: demonstration of catalytic and DNA binding activity
- Author
-
Santosh Pasha, Vani Brahmachari, Ritu Sharma, Souvik Maiti, Rachit Bakshi, and Abhishek Kumar Mehta
- Subjects
Therapeutic gene modulation ,HMG-box ,Biophysics ,Biology ,Biochemistry ,Catalysis ,Cell Line ,Mice ,Coactivator ,Animals ,Humans ,Protein–DNA interaction ,Cloning, Molecular ,Molecular Biology ,Adenosine Triphosphatases ,Cell Nucleus ,DNA Helicases ,Cell Biology ,DNA-binding domain ,Molecular biology ,Cell biology ,Chromatin ,Protein Structure, Tertiary ,DNA binding site ,DNA-Binding Proteins ,Gene Expression Regulation ,Organ Specificity ,Binding domain - Abstract
The proteins belonging to SWI2/SNF2 family of DNA dependent ATPases are important members of the chromatin remodeling complexes that are implicated in epigenetic control of gene expression. We have identified a human gene with a putative DNA binding domain, which belongs to the INO80 subfamily of SWI2/SNF2 proteins. Here we report the cloning, expression, and functional activity of the domains from hINO80 gene both in terms of the DNA dependent ATPase as well as DNA binding activity. A differential expression of the various domains within this gene is detected in human tissues while a ubiquitous expression is detected in mice. The intranuclear localization is demonstrated using antibodies directed against the DBINO domain of hINO80.
- Published
- 2005
28. P3.042 A GATA-2-switch model of a-synuclein overexpression in sporadic Parkinson's disease
- Author
-
E. Bresnick, M. Schlossmacher, Z. Liao, Clemens R. Scherzer, B. Zheng, and Rachit Bakshi
- Subjects
Parkinson's disease ,Neurology ,business.industry ,Cancer research ,Synuclein ,Medicine ,Neurology (clinical) ,Geriatrics and Gerontology ,business ,medicine.disease - Published
- 2009
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.