Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Shankar J. Chinta is active.

Publication


Featured researches published by Shankar J. Chinta.


Biochimica et Biophysica Acta | 2008

Redox imbalance in Parkinson's disease.

Shankar J. Chinta; Julie K. Andersen

Parkinsons disease (PD) is an adult-onset neurodegenerative disorder characterized by preferential loss of dopaminergic neurons in an area of the midbrain called the substantia nigra (SN) along with occurrence of intraneuronal inclusions called Lewy bodies. The majority of cases of PD are sporadic in nature with late onset (95% of patients); however a few PD cases (5%) are seen in familial clusters with generally earlier onset. Although PD has been heavily researched, so far the exact cause of the rather selective cell death is unknown. Multiple lines of evidence suggest an important role for oxidative stress. Dopaminergic neurons (DA) are particularly prone to oxidative stress due to DA metabolism and auto-oxidation combined with increased iron, decreased total glutathione levels and mitochondrial complex I inhibition-induced ROS production in the SN which can lead to cell death by exceeding the oxidative capacity of DA-containing cells in the region. Enhancing antioxidant capabilities and chelating labile iron pools in this region therefore constitutes a rational approach to prevent or slow ongoing damage of DA neurons. In this review, we summarize the various sources of reactive oxygen species that may cause redox imbalance in PD as well as potential therapeutic targets for attenuation of oxidative stress associated with PD.


Neuroscience Letters | 2010

Mitochondrial alpha-synuclein accumulation impairs complex I function in dopaminergic neurons and results in increased mitophagy in vivo

Shankar J. Chinta; Jyothi K. Mallajosyula; Anand Rane; Julie K. Andersen

Alpha-synuclein is the major protein component of Lewy bodies, a cardinal pathological feature of the degenerating Parkinsonian brain. Alpha-synuclein has been reported to be able to intercalate into membranes via formation of an alpha-helical structure at its N-terminal end. Recent in vitro studies from various laboratories have demonstrated that α-synuclein can physically associate with mitochondria and interfere with mitochondrial function. α-Syn predominantly associates with the inner mitochondrial membrane, where it can apparently interact with complex I resulting in reduced mitochondrial complex I activity and increased free radical production. However, the effect of in vivo α-synuclein accumulation within dopaminergic neurons on mitochondrial function has not been thoroughly studied. Examination of transgenic animals which overexpress the familial mutant A53T form of the protein selectively within dopaminergic neurons reveals that A53T localizes to the mitochondrial membranes as monomers and oligomers particularly under conditions of proteasomal inhibitory stress, and that this localization coincides with a selective age-related mitochondrial complex I inhibition and decreased substrate-specific respiration along with increases in mitochondrial autophagy (mitophagy).


PLOS ONE | 2008

MAO-B Elevation in Mouse Brain Astrocytes Results in Parkinson's Pathology

Jyothi K. Mallajosyula; Deepinder Kaur; Shankar J. Chinta; Subramanian Rajagopalan; Anand Rane; David G. Nicholls; Donato A. Di Monte; Heather Macarthur; Julie K. Andersen

Age-related increases in monoamine oxidase B (MAO-B) may contribute to neurodegeneration associated with Parkinsons disease (PD). The MAO-B inhibitor deprenyl, a long-standing antiparkinsonian therapy, is currently used clinically in concert with the dopamine precursor L-DOPA. Clinical studies suggesting that deprenyl treatment alone is not protective against PD associated mortality were targeted to symptomatic patients. However, dopamine loss is at least 60% by the time PD is symptomatically detectable, therefore lack of effect of MAO-B inhibition in these patients does not negate a role for MAO-B in pre-symptomatic dopaminergic loss. In order to directly evaluate the role of age-related elevations in astroglial MAO-B in the early initiation or progression of PD, we created genetically engineered transgenic mice in which MAO-B levels could be specifically induced within astroglia in adult animals. Elevated astrocytic MAO-B mimicking age related increase resulted in specific, selective and progressive loss of dopaminergic neurons in the substantia nigra (SN), the same subset of neurons primarily impacted in the human condition. This was accompanied by other PD-related alterations including selective decreases in mitochondrial complex I activity and increased mitochondrial oxidative stress. Along with a global astrogliosis, we observed local microglial activation within the SN. These pathologies correlated with decreased locomotor activity. Importantly, these events occurred even in the absence of the PD-inducing neurotoxin MPTP. Our data demonstrates that elevation of murine astrocytic MAO-B by itself can induce several phenotypes of PD, signifying that MAO-B could be directly involved in multiple aspects of disease neuropathology. Mechanistically this may involve increases in membrane permeant H2O2 which can oxidize dopamine within dopaminergic neurons to dopaminochrome which, via interaction with mitochondrial complex I, can result in increased mitochondrial superoxide. Our inducible astrocytic MAO-B transgenic provides a novel model for exploring pathways involved in initiation and progression of several key features associated with PD pathology and for therapeutic drug testing.


The Journal of Neuroscience | 2007

Inducible Alterations of Glutathione Levels in Adult Dopaminergic Midbrain Neurons Result in Nigrostriatal Degeneration

Shankar J. Chinta; M. J. Kumar; Michael Hsu; Subramanian Rajagopalan; Deepinder Kaur; Anand Rane; David G. Nicholls; Jinah Choi; Julie K. Andersen

Parkinsons disease is a neurodegenerative disorder characterized by the preferential loss of midbrain dopaminergic neurons in the substantia nigra (SN). One of the earliest detectable biochemical alterations that occurs in the Parkinsonian brain is a marked reduction in SN levels of total glutathione (glutathione plus glutathione disulfide), occurring before losses in mitochondrial complex I (CI) activity, striatal dopamine levels, or midbrain dopaminergic neurodegeneration associated with the disease. Previous in vitro data from our laboratory has suggested that prolonged depletion of dopaminergic glutathione results in selective impairment of mitochondrial complex I activity through a reversible thiol oxidation event. To address the effects of depletion in dopaminergic glutathione levels in vivo on the nigrostriatal system, we created genetically engineered transgenic mouse lines in which expression of γ-glutamyl cysteine ligase, the rate-limiting enzyme in de novo glutathione synthesis, can be inducibly downregulated in catecholaminergic neurons, including those of the SN. A novel method for isolation of purified dopaminergic striatal synaptosomes was used to study the impact of dopaminergic glutathione depletion on mitochondrial events demonstrated previously to occur in vitro as a consequence of this alteration. Dopaminergic glutathione depletion was found to result in a selective reversible thiol-oxidation-dependent mitochondrial complex I inhibition, followed by an age-related nigrostriatal neurodegeneration. This suggests that depletion in glutathione within dopaminergic SN neurons has a direct impact on mitochondrial complex I activity via increased nitric oxide-related thiol oxidation and age-related dopaminergic SN cell loss.


Neurobiology of Aging | 2007

Increased murine neonatal iron intake results in Parkinson-like neurodegeneration with age

Deepinder Kaur; Jun Peng; Shankar J. Chinta; Subramanian Rajagopalan; Donato Di Monte; Robert A. Cherny; Julie K. Andersen

Iron elevation is well-documented in the Parkinsonian midbrain but its cause and contribution to subsequent neurodegeneration remain unknown. Mice administered iron at doses equivalent to those found in iron-fortified human infant formula during a developmental period equivalent to the first human year of life display progressive midbrain neurodegeneration and enhanced vulnerability to toxic injury. This may have major implications for the impact of neonatal iron intake as a potential risk factor for later development of Parkinsons disease (PD).


Free Radical Biology and Medicine | 2012

Selective binding of nuclear alpha-synuclein to the PGC1alpha promoter under conditions of oxidative stress may contribute to losses in mitochondrial function: implications for Parkinson’s disease

Almas Siddiqui; Shankar J. Chinta; Jyothi K. Mallajosyula; Subramanian Rajagopolan; Ingrid M. Hanson; Anand Rane; Simon Melov; Julie K. Andersen

Alpha-synuclein has been reported to be present in the nucleus and levels enhanced by oxidative stress. Herein, we sought to investigate the mechanistic role of nuclear alpha-synuclein. We found that alpha-synuclein nuclear localization coincided with enhanced chromatin binding both in an in vitro and a corresponding in vivo brain oxidative stress model previously characterized by our laboratory as well as in PD brain tissues. Genome-wide chromatin immunoprecipitation (ChIP)-on-chip analysis of alpha-synuclein:promoter binding in response to oxidative stress in vitro revealed that binding occurs at several promoters belonging to a range of functional categories including transcriptional regulation. Interestingly, given the important role of mitochondrial dysfunction in PD, this included binding to the promoter for the master mitochondrial transcription activator, PGC1alpha in vitro, in vivo, and in human brain tissue with age and PD. To test the possible mechanistic impact of alpha-synuclein PGC1alpha promotor binding, we assessed PGC1alpha promoter activity, mRNA, and protein levels and expression of candidate PGC1alpha target genes in our in vitro model. All were found to be reduced in conjunction with increased levels of aberrant mitochondrial morphology and impaired mitochondrial function. Exogenous PGC1alpha expression was found to attenuate alpha-synuclein-mediated mitochondrial dysfunction and subsequent neurotoxicity in vitro. Our data suggest that nuclear alpha-synuclein localization under conditions of oxidative stress may impact on mitochondrial function in part via the proteins capacity to act as a transcriptional modulator of PGC1alpha. This represents a novel role for alpha-synuclein as it relates to mitochondrial dysfunction in PD.


Free Radical Research | 2011

Nitrosylation and nitration of mitochondrial complex I in Parkinson's disease

Shankar J. Chinta; Julie K. Andersen

Abstract Impairment of the mitochondrial electron transport chain has been suggested to be a critical factor in the neuropathogenesis of Parkinson’s disease (PD), as inhibition of mitochondrial complex I (CI) activity is consistently detected in PD patients as well as in mitochondrial toxin models of the disorder. Increased levels of various reactive oxygen and nitrogen species appear to contribute to CI inhibition and mitochondrial dysfunction in PD. Reactive nitrogen species (RNS) such as nitric oxide (NO) and its metabolite peroxynitrite (PN) may inhibit CI activity via several different mechanisms including S-nitrosylation, nitration, and protein thiol formation. Studies using various cell and animal PD models have demonstrated that selective mitochondrial CI inhibition in dopaminergic cells may be due to both NO-mediated S-nitrosylation and nitration of CI sub-units. Strategies to modulate mitochondrial NO levels will therefore likely be a promising approach to enhance mitochondrial function and protect dopaminergic neurons against oxidative or nitrosative insult.


Journal of Internal Medicine | 2013

Environmental stress, ageing and glial cell senescence: a novel mechanistic link to Parkinson’s disease?

Shankar J. Chinta; Christopher A. Lieu; Marco Demaria; Remi-Martin Laberge; Judith Campisi; Julie K. Andersen

Exposure to environmental toxins is associated with a variety of age‐related diseases including cancer and neurodegeneration. For example, in Parkinsons disease (PD), chronic environmental exposure to certain toxins has been linked to the age‐related development of neuropathology. Neuronal damage is believed to involve the induction of neuroinflammatory events as a consequence of glial cell activation. Cellular senescence is a potent anti‐cancer mechanism that occurs in a number of proliferative cell types and causes the arrest of proliferation of cells at risk of malignant transformation following exposure to potentially oncogenic stimuli. With age, senescent cells accumulate and express a senescence‐associated secretory phenotype (SASP; that is the robust secretion of many inflammatory cytokines, growth factors and proteases). Whereas cell senescence in peripheral tissues has been causally linked to a number of age‐related pathologies, little is known about the induction of cellular senescence and the SASP in the brain. On the basis of recently reported findings, we propose that environmental stressors associated with PD may act in part by eliciting senescence and the SASP within non neuronal glial cells in the ageing brain, thus contributing to the characteristic decline in neuronal integrity that occurs in this disorder.


Molecular Brain Research | 2002

Constitutive expression and localization of the major drug metabolizing enzyme, cytochrome P4502D in human brain

Shankar J. Chinta; Harish V. Pai; Sudarshan C. Upadhya; Michael R. Boyd; Vijayalakshmi Ravindranath

Cytochrome P4502D6, an important isoform of cytochrome P450, mediates the metabolism of several psychoactive drugs in liver. Quantitatively, liver is the major drug metabolizing organ, however metabolism of drugs in brain could modulate pharmacological and pharmacodynamic effects of psychoactive drugs at their site of action and explain some of the variation typically seen in patient population. We have measured cytochrome P450 content and examined constitutive expression of CYP2D mRNA and protein in human brain regions by reverse transcription polymerase chain reaction, Northern and immunoblotting and localized it by in situ hybridization and immunohistochemistry. CYP2D mRNA was expressed constitutively in neurons of cerebral cortex, Purkinje and granule cell layers of cerebellum, reticular neurons of midbrain and pyramidal neurons of CA1, CA2 and CA3 subfields of hippocampus. Immunoblot studies demonstrated the presence of cytochrome P4502D protein in cortex, cerebellum, midbrain, striatum and thalamus of human brain. Immunohistochemical localization showed the predominant presence of cytochrome P4502D not only in neuronal soma but also in dendrites of Purkinje and cortical neurons. These studies demonstrate constitutive expression of cytochrome P4502D in neuronal cell population in human brain, indicating its possible role in metabolism of psychoactive drugs directly at or near their site of action, in neurons, in human brain.


The Journal of Neuroscience | 2015

Mitochondrial Quality Control via the PGC1α-TFEB Signaling Pathway Is Compromised by Parkin Q311X Mutation But Independently Restored by Rapamycin

Almas Siddiqui; Dipa Bhaumik; Shankar J. Chinta; Anand Rane; Subramanian Rajagopalan; Christopher A. Lieu; Gordon J. Lithgow; Julie K. Andersen

Following its activation by PINK1, parkin is recruited to depolarized mitochondria where it ubiquitinates outer mitochondrial membrane proteins, initiating lysosomal-mediated degradation of these organelles. Mutations in the gene encoding parkin, PARK2, result in both familial and sporadic forms of Parkinsons disease (PD) in conjunction with reductions in removal of damaged mitochondria. In contrast to what has been reported for other PARK2 mutations, expression of the Q311X mutation in vivo in mice appears to involve a downstream step in the autophagic pathway at the level of lysosomal function. This coincides with increased PARIS expression and reduced expression of a reciprocal signaling pathway involving the master mitochondrial regulator peroxisome proliferator-activated receptor-gamma coactivator (PGC1α) and the lysosomal regulator transcription factor EB (TFEB). Treatment with rapamycin was found to independently restore PGC1α-TFEB signaling in a manner not requiring parkin activity and to abrogate impairment of mitochondrial quality control and neurodegenerative features associated with this in vivo model. Losses in PGC1α-TFEB signaling in cultured rat DAergic cells expressing the Q311X mutation associated with reduced mitochondrial function and cell viability were found to be PARIS-dependent and to be independently restored by rapamycin in a manner requiring TFEB. Studies in human iPSC-derived neurons demonstrate that TFEB induction can restore mitochondrial function and cell viability in a mitochondrially compromised human cell model. Based on these data, we propose that the parkin Q311X mutation impacts on mitochondrial quality control via PARIS-mediated regulation of PGC1α-TFEB signaling and that this can be independently restored via upregulation of TFEB function. SIGNIFICANCE STATEMENT Mutations in PARK2 are generally associated with loss in ability to interact with PINK1, impacting on autophagic initiation. Our data suggest that, in the case of at least one parkin mutation, Q311X, detrimental effects are due to inhibition at the level of downstream lysosomal function. Mechanistically, this involves elevations in PARIS protein levels and subsequent effects on PGC1α-TFEB signaling that normally regulates mitochondrial quality control. Treatment with rapamycin independently restores PGC1α-TFEB signaling in a manner not requiring parkin activity and abrogates subsequent mitochondrial impairment and neuronal cell loss. Taken in total, our data suggest that the parkin Q311X mutation impacts on mitochondrial quality control via PARIS-mediated regulation of PGC1α-TFEB signaling and that this can be independently restored via rapamycin.

Collaboration


Dive into the Shankar J. Chinta's collaboration.

Top Co-Authors

Avatar

Julie K. Andersen

Buck Institute for Research on Aging

View shared research outputs
Top Co-Authors

Avatar

Anand Rane

Buck Institute for Research on Aging

View shared research outputs
Top Co-Authors

Avatar

Subramanian Rajagopalan

Buck Institute for Research on Aging

View shared research outputs
Top Co-Authors

Avatar

Deepinder Kaur

Buck Institute for Research on Aging

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Almas Siddiqui

Buck Institute for Research on Aging

View shared research outputs
Top Co-Authors

Avatar

Christopher A. Lieu

Buck Institute for Research on Aging

View shared research outputs
Top Co-Authors

Avatar

David G. Nicholls

Buck Institute for Research on Aging

View shared research outputs
Top Co-Authors

Avatar

Jyothi K. Mallajosyula

Buck Institute for Research on Aging

View shared research outputs
Top Co-Authors

Avatar

Harish V. Pai

National Institute of Mental Health and Neurosciences

View shared research outputs
Researchain Logo
Decentralizing Knowledge