Network


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

Hotspot


Dive into the research topics where Ishita Parikh is active.

Publication


Featured researches published by Ishita Parikh.


The Journal of Neuroscience | 2013

CD33 Alzheimer's Risk-Altering Polymorphism, CD33 Expression, and Exon 2 Splicing

Manasi Malik; James Simpson; Ishita Parikh; Bernard R. Wilfred; David W. Fardo; Peter T. Nelson; Steven Estus

Genome-wide association studies are identifying novel Alzheimers disease (AD) risk factors. Elucidating the mechanism underlying these polymorphisms is critical to the validation process and, by identifying rate-limiting steps in AD risk, may yield novel therapeutic targets. Here, we elucidate the mechanism of action of the AD-associated polymorphism rs3865444 in the promoter of CD33, a member of the sialic acid-binding Ig-superfamily of lectins (SIGLECs). Immunostaining established that CD33 is expressed in microglia in human brain. Consistent with this finding, CD33 mRNA expression correlated well with expression of the microglial genes CD11b and AIF-1 and was modestly increased with AD status and the rs3865444C AD-risk allele. Analysis of CD33 isoforms identified a common isoform lacking exon 2 (D2-CD33). The proportion of CD33 expressed as D2-CD33 correlated robustly with rs3865444 genotype. Because rs3865444 is in the CD33 promoter region, we sought the functional polymorphism by sequencing CD33 from the promoter through exon 4. We identified a single polymorphism that is coinherited with rs3865444, i.e., rs12459419 in exon 2. Minigene RNA splicing studies in BV2 microglial cells established that rs12459419 is a functional single nucleotide polymorphism (SNP) that modulates exon 2 splicing efficiency. Thus, our primary findings are that CD33 is a microglial mRNA and that rs3865444 is a proxy SNP for rs12459419 that modulates CD33 exon 2 splicing. Exon 2 encodes the CD33 IgV domain that typically mediates sialic acid binding in SIGLEC family members. In summary, these results suggest a novel model wherein SNP-modulated RNA splicing modulates CD33 function and, thereby, AD risk.


PLOS ONE | 2014

Genetics of PICALM Expression and Alzheimer's Disease

Ishita Parikh; David W. Fardo; Steven Estus

Novel Alzheimers disease (AD) risk factors have been identified by genome-wide association studies. Elucidating the mechanism underlying these factors is critical to the validation process and, by identifying rate-limiting steps in AD risk, may yield novel therapeutic targets. Here, we evaluated the association between the AD-associated polymorphism rs3851179 near PICALM, which encodes a clathrin-coated pit accessory protein. Immunostaining established that PICALM is expressed predominately in microvessels in human brain. Consistent with this finding, PICALM mRNA expression correlated with expression of the endothelial genes vWF and CD31. Additionally, we found that PICALM expression was modestly increased with the rs3851179A AD-protective allele. Analysis of PICALM isoforms found several isoforms lacking exons encoding elements previously identified as critical to PICALM function. Increased expression of the common isoform lacking exon 13 was also associated with the rs3851179A protective allele; this association was not apparent when this isoform was compared with total PICALM expression, indicating that the SNP is associated with total PICALM expression and not this isoform per se. Interestingly, PICALM lacking exons 2–4 was not associated with rs3851179 but was associated with rs592297, which is located in exon 5. Thus, our primary findings are that multiple PICALM isoforms are expressed in the human brain, that PICALM is robustly expressed in microvessels, and that expression of total PICALM is modestly correlated with the AD-associated SNP rs3851179. We interpret these results as suggesting that increased PICALM expression in the microvasculature may reduce AD risk.


Aging (Albany NY) | 2016

Caloric restriction preserves memory and reduces anxiety of aging mice with early enhancement of neurovascular functions

Ishita Parikh; Janet Guo; Kai-Hsiang Chuang; Yu Zhong; Ralf G. Rempe; Jared D. Hoffman; Rachel Armstrong; Björn Bauer; Anika M.S. Hartz; Ai Ling Lin

Neurovascular integrity plays an important role in protecting cognitive and mental health in aging. Lifestyle interventions that sustain neurovascular integrity may thus be critical on preserving brain functions in aging and reducing the risk for age-related neurodegenerative disorders. Here we show that caloric restriction (CR) had an early effect on neurovascular enhancements, and played a critical role in preserving vascular, cognitive and mental health in aging. In particular, we found that CR significantly enhanced cerebral blood flow (CBF) and blood-brain barrier function in young mice at 5-6 months of age. The neurovascular enhancements were associated with reduced mammalian target of rapamycin expression, elevated endothelial nitric oxide synthase signaling, and increased ketone bodies utilization. With age, CR decelerated the rate of decline in CBF. The preserved CBF in hippocampus and frontal cortex were highly correlated with preserved memory and learning, and reduced anxiety, of the aging mice treated with CR (18-20 months of age). Our results suggest that dietary intervention started in the early stage (e.g., young adults) may benefit cognitive and mental reserve in aging. Understanding nutritional effects on neurovascular functions may have profound implications in human brain aging and age-related neurodegenerative disorders.


Molecular Neurodegeneration | 2014

An intronic PICALM polymorphism, rs588076, is associated with allelic expression of a PICALM isoform

Ishita Parikh; Christopher Medway; Steven G. Younkin; David W. Fardo; Steven Estus

BackgroundAlthough genome wide studies have associated single nucleotide polymorphisms (SNP)s near PICALM with Alzheimer’s disease (AD), the mechanism underlying this association is unclear. PICALM is involved in clathrin-mediated endocytosis and modulates Aß clearance in vitro. Comparing allelic expression provides the means to detect cis-acting regulatory polymorphisms. Thus, we evaluated whether PICALM showed allele expression imbalance (AEI) and whether this imbalance was associated with the AD-associated polymorphism, rs3851179.ResultsWe measured PICALM allelic expression in 42 human brain samples by using next-generation sequencing. Overall, PICALM demonstrated equal allelic expression with no detectable influence by rs3851179. A single sample demonstrated robust global PICALM allelic expression imbalance (AEI), i.e., each of the measured isoforms showed AEI. Moreover, the PICALM isoform lacking exons 18 and 19 (D18-19 PICALM) showed significant AEI in a subset of individuals. Sequencing these individuals and subsequent genotyping revealed that rs588076, located in PICALM intron 17, was robustly associated with this imbalance in D18-19 PICALM allelic expression (p = 9.54 x 10-5). This polymorphism has been associated previously with systolic blood pressure response to calcium channel blocking agents. To evaluate whether this polymorphism was associated with AD, we genotyped 3269 individuals and found that rs588076 was modestly associated with AD. However, when both the primary AD SNP rs3851179 was added to the logistic regression model, only rs3851179 was significantly associated with AD.ConclusionsPICALM expression shows no evidence of AEI associated with rs3851179. Robust global AEI was detected in one sample, suggesting the existence of a rare SNP that strongly modulates PICALM expression. AEI was detected for the D18-19 PICALM isoform, and rs588076 was associated with this AEI pattern. Conditional on rs3851179, rs588076 was not associated with AD risk, suggesting that D18-19 PICALM is not critical in AD. In summary, this analysis of PICALM allelic expression provides novel insights into the genetics of PICALM expression and AD risk.


Frontiers in Aging Neuroscience | 2017

Age drives distortion of brain metabolic, vascular and cognitive functions, and the gut microbiome

Jared D. Hoffman; Ishita Parikh; Stefan J. Green; George Chlipala; Robert P. Mohney; Mignon Keaton; Bjoern Bauer; Anika M.S. Hartz; Ai Ling Lin

Advancing age is the top risk factor for the development of neurodegenerative disorders, including Alzheimer’s disease (AD). However, the contribution of aging processes to AD etiology remains unclear. Emerging evidence shows that reduced brain metabolic and vascular functions occur decades before the onset of cognitive impairments, and these reductions are highly associated with low-grade, chronic inflammation developed in the brain over time. Interestingly, recent findings suggest that the gut microbiota may also play a critical role in modulating immune responses in the brain via the brain-gut axis. In this study, our goal was to identify associations between deleterious changes in brain metabolism, cerebral blood flow (CBF), gut microbiome and cognition in aging, and potential implications for AD development. We conducted our study with a group of young mice (5–6 months of age) and compared those to old mice (18–20 months of age) by utilizing metabolic profiling, neuroimaging, gut microbiome analysis, behavioral assessments and biochemical assays. We found that compared to young mice, old mice had significantly increased levels of numerous amino acids and fatty acids that are highly associated with inflammation and AD biomarkers. In the gut microbiome analyses, we found that old mice had increased Firmicutes/Bacteroidetes ratio and alpha diversity. We also found impaired blood-brain barrier (BBB) function and reduced CBF as well as compromised learning and memory and increased anxiety, clinical symptoms often seen in AD patients, in old mice. Our study suggests that the aging process involves deleterious changes in brain metabolic, vascular and cognitive functions, and gut microbiome structure and diversity, all which may lead to inflammation and thus increase the risk for AD. Future studies conducting comprehensive and integrative characterization of brain aging, including crosstalk with peripheral systems and factors, will be necessary to define the mechanisms underlying the shift from normal aging to pathological processes in the etiology of AD.


Scientific Reports | 2018

Ketogenic diet enhances neurovascular function with altered gut microbiome in young healthy mice.

David Ma; Amy Wang; Ishita Parikh; Stefan J. Green; Jared D. Hoffman; George Chlipala; M. Paul Murphy; Brent S. Sokola; Björn Bauer; Anika M.S. Hartz; Ai Ling Lin

Neurovascular integrity, including cerebral blood flow (CBF) and blood-brain barrier (BBB) function, plays a major role in determining cognitive capability. Recent studies suggest that neurovascular integrity could be regulated by the gut microbiome. The purpose of the study was to identify if ketogenic diet (KD) intervention would alter gut microbiome and enhance neurovascular functions, and thus reduce risk for neurodegeneration in young healthy mice (12–14 weeks old). Here we show that with 16 weeks of KD, mice had significant increases in CBF and P-glycoprotein transports on BBB to facilitate clearance of amyloid-beta, a hallmark of Alzheimer’s disease (AD). These neurovascular enhancements were associated with reduced mechanistic target of rapamycin (mTOR) and increased endothelial nitric oxide synthase (eNOS) protein expressions. KD also increased the relative abundance of putatively beneficial gut microbiota (Akkermansia muciniphila and Lactobacillus), and reduced that of putatively pro-inflammatory taxa (Desulfovibrio and Turicibacter). We also observed that KD reduced blood glucose levels and body weight, and increased blood ketone levels, which might be associated with gut microbiome alteration. Our findings suggest that KD intervention started in the early stage may enhance brain vascular function, increase beneficial gut microbiota, improve metabolic profile, and reduce risk for AD.


Current Nutrition Reports | 2017

Neuroimaging Biomarkers of Caloric Restriction on Brain Metabolic and Vascular Functions

Ai Ling Lin; Ishita Parikh; Jared D. Hoffman; David Ma

Purpose of ReviewNon-invasive neuroimaging methods have been developed as powerful tools for identifying in vivo brain functions for studies in humans and animals. Here, we review the imaging biomarkers that are being used to determine the changes within brain metabolic and vascular functions induced by caloric restriction (CR) and their potential usefulness for future studies with dietary interventions in humans.Recent FindingsCR causes an early shift in brain metabolism of glucose to ketone bodies and enhances ATP production, neuronal activity, and cerebral blood flow (CBF). With age, CR preserves mitochondrial activity, neurotransmission, CBF, and spatial memory. CR also reduces anxiety in aging mice. Neuroimaging studies in humans show that CR restores abnormal brain activity in the amygdala of women with obesity and enhances brain connectivity in old adults.SummaryNeuroimaging methods have excellent translational values and can be widely applied in future studies to identify dietary effects on brain functions in humans.


Frontiers in Aging Neuroscience | 2018

Neuroimaging Biomarkers of mTOR Inhibition on Vascular and Metabolic Functions in Aging Brain and Alzheimer’s Disease

Jennifer Lee; Lucille M. Yanckello; David Ma; Jared D. Hoffman; Ishita Parikh; Scott Thalman; Bjoern Bauer; Anika M.S. Hartz; Fahmeed Hyder; Ai Ling Lin

The mechanistic target of rapamycin (mTOR) is a nutrient sensor of eukaryotic cells. Inhibition of mechanistic mTOR signaling can increase life and health span in various species via interventions that include rapamycin and caloric restriction (CR). In the central nervous system, mTOR inhibition demonstrates neuroprotective patterns in aging and Alzheimer’s disease (AD) by preserving mitochondrial function and reducing amyloid beta retention. However, the effects of mTOR inhibition for in vivo brain physiology remain largely unknown. Here, we review recent findings of in vivo metabolic and vascular measures using non-invasive, multimodal neuroimaging methods in rodent models for brain aging and AD. Specifically, we focus on pharmacological treatment (e.g., rapamycin) for restoring brain functions in animals modeling human AD; nutritional interventions (e.g., CR and ketogenic diet) for enhancing brain vascular and metabolic functions in rodents at young age (5–6 months of age) and preserving those functions in aging (18–20 months of age). Various magnetic resonance (MR) methods [i.e., imaging (MRI), angiography (MRA), and spectroscopy (MRS)], confocal microscopic imaging, and positron emission tomography (PET) provided in vivo metabolic and vascular measures. We also discuss the translational potential of mTOR interventions. Since PET and various MR neuroimaging methods, as well as the different interventions (e.g., rapamycin, CR, and ketogenic diet) are also available for humans, these findings may have tremendous implications in future clinical trials of neurological disorders in aging populations.


Alzheimers & Dementia | 2018

RAPAMYCIN TREATMENT INCREASES CEREBRAL BLOOD FLOW AND GUT MICROBIOME DIVERSITY IN EFAD MICE

Ishita Parikh; David W.L. Ma; Jared D. Hoffman; Amy Wang; Scott Thalman; Stefan J. Green; Ai Ling Lin

negatively correlated with the AD neuropathology.Methods:To understand the mechanism by which the TTC3 alteration may be acting to contribute to LOAD risk, induced pluripotent stem cells (iPSC) lines were developed to examine cellular consequences in neuronal cells, which can otherwise only be collected postmortem. Peripheral blood mononuclear cells (PBMCs) were extracted from the whole blood of three LOAD individuals bearing the p.S1038C change, as well as three age and ethnically matched control individuals. iPSC reprogramming was performed using Sendai virus. Results: Each iPSC line generated was validated for pluripotency through immunocytochemical staining and shown to be negative for any large-scale chromosomal abnormalities via karyotyping. These iPSC lines are being differentiated into cortical neurons by viral transduction of Neurogenin 2 to promote rapid maturation. There is evidence that modulation of TTC3 affects neurite growth (Berto, et al, 2007); therefore, morphological measures of axon and synapse formation will be assessed in differentiating neuronal cultures using live cell imaging. Alzheimer-specific phenotypes such as the levels of secreted beta amyloid and intercellular whole and phosphorylated tau will also be measured. Conclusions:Utilizing patient-specific iPSC lines carrying a segregating alteration in TTC3 will enable us to determine how this genetic alteration which may contribute to AD specific cellular phenotypes and, on a broader scale, LOAD risk.


Molecular Neurodegeneration | 2015

Genetics ignite focus on microglial inflammation in Alzheimer’s disease

Manasi Malik; Ishita Parikh; Jared B. Vasquez; Conor Smith; Leon M. Tai; Guojun Bu; Mary Jo LaDu; David W. Fardo; G. William Rebeck; Steven Estus

Collaboration


Dive into the Ishita Parikh's collaboration.

Top Co-Authors

Avatar

Ai Ling Lin

University of Kentucky

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Amy Wang

University of Kentucky

View shared research outputs
Top Co-Authors

Avatar

David Ma

University of Kentucky

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Stefan J. Green

University of Illinois at Chicago

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge