Network


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

Hotspot


Dive into the research topics where Himanshu Vashistha is active.

Publication


Featured researches published by Himanshu Vashistha.


American Journal of Physiology-heart and Circulatory Physiology | 2009

Inhibition of p66ShcA redox activity in cardiac muscle cells attenuates hyperglycemia-induced oxidative stress and apoptosis.

Ashwani Malhotra; Himanshu Vashistha; Virendra S Yadav; Michael G. Dube; Satya P. Kalra; Maha Abdellatif; Leonard G. Meggs

Apoptotic myocyte cell death, diastolic dysfunction, and progressive deterioration in left ventricular pump function characterize the clinical course of diabetic cardiomyopathy. A key question concerns the mechanism(s) by which hyperglycemia (HG) transmits danger signals in cardiac muscle cells. The growth factor adapter protein p66ShcA is a genetic determinant of longevity, which controls mitochondrial metabolism and cellular responses to oxidative stress. Here we demonstrate that interventions which attenuate or prevent HG-induced phosphorylation at critical position 36 Ser residue (phospho-Ser36) inhibit the redox function of p66ShcA and promote the survival phenotype. Adult rat ventricular myocytes obtained by enzymatic dissociation were transduced with mutant-36 p66ShcA (mu-36) dominant-negative expression vector and plated in serum-free media containing 5 or 25 mM glucose. At HG, adult rat ventricular myocytes exhibit a marked increase in reactive oxygen species production, upregulation of phospho-Ser36, collapse of mitochondrial transmembrane potential, and increased formation of p66ShcA/cytochrome-c complexes. These indexes of oxidative stress were accompanied by a 40% increase in apoptosis and the upregulation of cleaved caspase-3 and the apoptosis-related proteins p53 and Bax. To test whether p66ShcA functions as a redox-sensitive molecular switch in vivo, we examined the hearts of male Akita diabetic nonobese (C57BL/6J) mice. Western blot analysis detected the upregulation of phospho-Ser36, the translocation of p66ShcA to mitochondria, and the formation of p66ShcA/cytochrome-c complexes. Conversely, the correction of HG by recombinant adeno-associated viral delivery of leptin reversed these alterations. We conclude that p66ShcA is a molecular switch whose redox function is turned on by phospho-Ser36 and turned off by interventions that prevent this modification.


Neoplasia | 2014

Therapeutic Efficacy of Aldoxorubicin in an Intracranial Xenograft Mouse Model of Human Glioblastoma

Luis Marrero; Dorota Wyczechowska; Alberto E. Musto; Anna Wilk; Himanshu Vashistha; Adriana Zapata; Chelsey Walker; Cruz Velasco-Gonzalez; Christopher Parsons; Scott Wieland; Daniel J. Levitt; Krzysztof Reiss; Om Prakash

Glioblastoma multiforme (GBM) is the most aggressive primary brain tumor with a median survival of 12 to 15 months after diagnosis. Acquired chemoresistance, high systemic toxicity, and low penetration of the blood brain barrier by many anticancer drugs contribute to the failure of anti-GBM therapies. To circumvent some of these obstacles, we tested a novel prodrug approach to evaluate anti-GBM efficacy by utilizing serum albumin-binding doxorubicin (Doxo), aldoxorubicin (Aldoxo), which is less toxic, is released from albumin in an acidic environment and accumulates in tumor tissues. A human GBM cell line that expresses a luciferase reporter (U87-luc) was stereotactically injected into the left striatum of the brain of immunodeficient mice. Following initial tumor growth for 12 days, mice were injected once a week in the tail-vein with Aldoxo [24 mg/kg or 18 mg/kg of doxorubicin equivalents—3/4 maximum tolerated dose (MTD)], Doxo [6 mg/kg (3/4 MTD)], or vehicle. Aldoxo-treated mice demonstrated significantly slower growth of the tumor when compared to vehicle-treated or Doxo-treated mice. Five out of eight Aldoxo-treated mice remained alive more than 60 days with a median survival of 62 days, while the median survival of vehicle- and Doxo-treated mice was only 26 days. Importantly, Aldoxo-treated mice exhibited high levels of Doxo within the tumor tissue, accompanied by low tumor cell proliferation (Ki67) and abundant intratumoral programmed cell death (cleaved caspase-3). Effective accumulation of Aldoxo in brain tumor tissues but not normal brain, its anti-tumor efficacy, and low toxicity, provide a strong rationale for evaluating this novel drug conjugate as a treatment for patients afflicted with GBM.


American Journal of Physiology-renal Physiology | 2012

Null mutations at the p66 and bradykinin 2 receptor loci induce divergent phenotypes in the diabetic kidney

Himanshu Vashistha; Pravin C. Singhal; Ashwani Malhotra; Mohammad Husain; Peter W. Mathieson; Moin A. Saleem; Cyril Kuriakose; Surya V. Seshan; Anna Wilk; Luis DelValle; Francesca Peruzzi; Marco Giorgio; Pier Giuseppe Pelicci; Oliver Smithies; Hyung Suk Kim; Masao Kakoki; Krzysztof Reiss; Leonard G. Meggs

Candidate genes have been identified that confer increased risk for diabetic glomerulosclerosis (DG). Mice heterozygous for the Akita (Ins2(+/C96Y)) diabetogenic mutation with a second mutation introduced at the bradykinin 2 receptor (B2R(-/-)) locus express a disease phenotype that approximates human DG. Src homology 2 domain transforming protein 1 (p66) controls mitochondrial metabolism and cellular responses to oxidative stress, aging, and apoptosis. We generated p66-null Akita mice to test whether inactivating mutations at the p66 locus will rescue kidneys of Akita mice from disease-causing mutations at the Ins2 and B2R loci. Here we show null mutations at the p66 and B2R loci interact with the Akita (Ins2(+/C96Y)) mutation, independently and in combination, inducing divergent phenotypes in the kidney. The B2R(-/-) mutation induces detrimental phenotypes, as judged by increased systemic and renal levels of oxidative stress, histology, and urine albumin excretion, whereas the p66-null mutation confers a powerful protection phenotype. To elucidate the mechanism(s) of the protection phenotype, we turned to our in vitro system. Experiments with cultured podocytes revealed previously unrecognized cross talk between p66 and the redox-sensitive transcription factor p53 that controls hyperglycemia-induced ROS metabolism, transcription of p53 target genes (angiotensinogen, angiotensin II type-1 receptor, and bax), angiotensin II generation, and apoptosis. RNA-interference targeting p66 inhibits all of the above. Finally, protein levels of p53 target genes were upregulated in kidneys of Akita mice but unchanged in p66-null Akita mice. Taken together, p66 is a potential molecular target for therapeutic intervention in DG.


Experimental and Molecular Pathology | 2013

Deficit of p66ShcA restores redox-sensitive stress response program in cisplatin-induced acute kidney injury.

Rungwasee Rattanavich; Andrei Plagov; Dileep Kumar; Partab Rai; Rivka Lederman; Divya Salhan; Himanshu Vashistha; Ashwani Malhotra; Leonard G. Meggs; Pravin C. Singhal

Overwhelming oxidative stress and compromised tubular cell antioxidant response have been incriminated for cisplatin (Cis)-induced acute kidney injury (AKI). We hypothesized that Cis-induced AKI was the outcome of the deactivated redox-sensitive stress response program (RSSRP). Wild type (WT) and heterozygous p66ShcA(p66(+/-)) mice in groups of six were administered either normal saline (WT) or Cis (12.5 mg/kg, intraperitoneal, Cis/WT). Renal biomarkers were collected and kidneys were harvested for renal histology. Cis/WT showed elevated blood urea nitrogen levels and enhanced tubular cell apoptosis, necrosis, and dilated tubules filled with casts when compared to Cis/p66(+/-). Cis/p66(+/-) developed only a clinically occult AKI (normal blood urea levels and only microscopic alterations). Immunoblots from the lysates of renal tissues of Cis/WT displayed enhanced expression of phospho-p66ShcA, and phospho-Foxo3A but attenuated expression of MnSOD and catalase; conversely, p66 deficit prevented these alterations in Cis milieu. In in vitro studies, Cis treated mouse proximal tubular cells (MPTCs) displayed enhanced phosphorylation of p66ShcA and no increase in tubular cell expression of MnSOD. In addition, renal tissues of Cis/WT and Cis-treated MPTCs displayed enhanced phosphorylation of p53 and Bax expression. However, MPTC partially silenced for p66ShcA displayed partial inhibition of Cis-induced tubular cell apoptosis as well as necrosis. These findings indicate that Cis-induced AKI is the outcome of the deactivated RSSRP (attenuated anti-oxidant response) and activation of pro-apoptotic (p53-induced Bax expression) pathway.


Canadian Journal of Physiology and Pharmacology | 2008

Overexpression of Gsalpha compensates for myocyte loss in diabetic cardiomyopathy.

Ashwani Malhotra; Barinder P. S. Kang; Himanshu Vashistha; Virender S.YadavV.S. Yadav; Leonard G. Meggs

The stimulatory G protein Gsalpha transmits signals from activated beta-adrenergic receptors via the cyclic AMP-PKA pathway, targeting the key regulatory protein phospholamban. We hypothesized that mice with intrinsic activation of cardiac Gsalpha are resistant to the development of the diabetic cardiomyopathy phenotype. Accordingly, streptozotocin (STZ)-diabetes mellitus was induced in genetically engineered mice with cardiac-specific Gsalpha overexpression and in nontransgenic (NTG) littermates. At 8 weeks, Gsalpha diabetic mice showed no impairment of LV contractility nor increase in myocyte apoptosis, whereas NTG diabetic mice showed a 30% decrease in +dP/dt and -dP/dt with sustained (3-fold) myocyte loss by apoptosis. To assess the level of myocardial reactive oxygen species, we measured malondialdehyde, a surrogate marker of oxidative stress, which was increased in the hearts of NTG and Gsalpha diabetic mice. In addition, chronic hyperglycemia also increased the activity of catalase and superoxide dismutase in the hearts of NTG and Gsalpha diabetic mice. Hearts of NTG diabetic mice, but not Gsalpha mice, showed increased expression of proapoptosis Bax, downregulation in Bcl2, and an increase in the Bax/Bcl2 ratio. Hearts of NTG diabetic mice showed 60% reduction in phosphorylation at the critical Ser16 residue of phospholamban, whereas phosphorylation at Ser16 was restored in hearts of Gsalpha-diabetic mice. We conclude that cardiac-specific overexpression of Gsalpha compensates for the loss of cardiac function in diabetes mellitus.


bioRxiv | 2018

Apolipoprotein L1 Dynamics in Human Parietal Epithelial Cell Molecular Phenotype Kinetics

Vinod Kumar; Himanshu Vashistha; Xiqian Lan; Nirupama Chandel; Kamesh Ayasolla; Rukhsana Aslam; Seyedeh Shadafarin Marshi Shoshtari; Nitpriya Paliwal; Frank Abbruscato; Joanna Mikulak; Waldemar Popik; Ashwani Malhotra; Catherine Meyer-Scwesinger; Karl Skorecki; Pravin C. Singhal

Human Parietal Epithelial cells (PECs) are considered as a source of progenitor cells to sustain podocyte (PD) homeostasis. We hypothesized that the absence of apolipoprotein (APO) L1 favors the PEC phenotype and that induction of APOL1 transitions to PD renewal. During PECs’ transition, APOL1 expression coincided with the expression of PD markers (PEC transition) along with down regulation of miR193a. The induction of APOL1 down regulated miR193a and induced PD markers in PECs/HEKs; whereas, the APOL1-silencing in transited (Tr)-PECs/HepG2s up regulated miR193a expression suggesting a reciprocally linked feedback loop relationship between APOL1 and miR193a. HIV, IFN-y, and vitamin D receptor agonist (VDA) induced APOL1 expression and PEC transition markers but down regulated miR193a in PECs/HEKs. Glomeruli in HIV patients and HIV: APOL1 transgenic mice displayed foci of PECs expressing synaptopodin, a PEC transition marker. Since APOL1 silencing in PECs partially attenuated HIV-, VDA-, and IFN-y-induced PECs transition, this would suggest that APOL1 is an important functional constituent of APOL1-miR193a axis.


American Journal of Physiology-renal Physiology | 2018

Aging phenotype(s) in kidneys of diabetic mice are p66ShcA dependent.

Himanshu Vashistha; Luis Marrero; Krzysztof Reiss; Ari J. Cohen; Ashwani Malhotra; Tariq Javed; Allyson E Bradley; Frank Abbruscato; Sixto Giusti; Antonio Jimenez; Smriti Mehra; Deepak Kaushal; Marco Giorgio; Pier Giuseppe Pelicci; Masao Kakoki; Pravin C. Singhal; Bruce A. Bunnell; Leonard G. Meggs

The p66ShcA protein controls cellular responses to oxidative stress, senescence, and apoptosis. Here, we test the hypothesis that aging phenotype(s) commonly associated with the broad category of chronic kidney disease are accelerated in diabetic kidneys and linked to the p66ShcA locus. At the organ level, tissue stem cells antagonize senescent phenotypes by replacing old dysfunctional cells. Using established methods, we isolated a highly purified population of stem cell antigen-1-positive mesenchymal stem cells (Sca-1+ MSCs) from kidneys of wild-type (WT) and p66 knockout (p66 KO) mice. Cells were plated in culture medium containing normal glucose (NG) or high glucose (HG). Reactive oxygen species (ROS) metabolism was substantially increased in WT MSCs in HG medium in association with increased cell death by apoptosis and acquisition of the senescent phenotype. DNA microarray analysis detected striking differences in the expression profiles of WT and p66 KO-MSCs in HG medium. Unexpectedly, the analysis for p66 KO-MSCs revealed upregulation of Wnt genes implicated in self-renewal and differentiation. To test the in vivo consequences of constitutive p66 expression in diabetic kidneys, we crossed the Akita diabetic mouse with the p66KO mouse. Homozygous mutation at the p66 locus delays or prevents aging phenotype(s) in the kidney that may be precursors to diabetic nephropathy.


Cancer Research | 2012

Abstract 1801: Lyn and Fyn inhibition as a potential novel treatment for glioblastoma multiforme

Om Prakash; Anna Wilk; Himanshu Vashistha; Shih-Chao Lin; Rita Aregbezola; Daniel J. Levitt; Krzysztof Reiss

Proceedings: AACR 103rd Annual Meeting 2012‐‐ Mar 31‐Apr 4, 2012; Chicago, IL Background: Glioblastoma multiforme (GBM) is the most malignant and most aggressive of the adult brain tumors. The current therapies including surgical resection, radiation therapy and chemotherapy remain palliative with median survival of 14 months. Two of the Src family kinases, Lyn and Fyn, have been reported to be involved in the pathogenesis and aggressiveness of GBM in clinical and laboratory research studies. Specific Aims: We evaluated the effect of Lyn and Fyn kinase inhibition on GBM cell motility and survival by utilizing a clinically relevant Lyn and Fyn kinase inhibitor, bafetinib or formerly INNO-406 (CytRx Corporation, Los Angeles, CA). Bafetinib, a dual Bcr-Abl and Lyn tyrosine kinase inhibitor has been shown to inhibit 4 of the 79 tyrosine kinases, namely Abl, Abl-related gene (Arg), and two of the Src family kinases Lyn and Fyn but not Src. Methodology: We used Lyn- expressing T98G and Fyn-expressing U87MG human GBM cell lines. The effect of bafetinib on the motile activity was evaluated by the wound healing, single cell trajectory, and the trans-well assays. The cell survival and mitochondrial membrane potential were evaluated by Guava Easycyte *HT flowcytometer (Millipore) in cells treated with bafetinib for 24 h. Further, we examined the effect of bafetinib on cell survival in combination with temozolomide (TMZ), a widely used chemotherapy agent for the management of GBM, in U87MG cells using microplate MTT assay. Results: At 16 h post-wounding, bafetinib at 5 µM concentration inhibited the motility of T98G and U87MG cells by 33% (P < 0.01) and 24% (P < 0.01), respectively. This significant inhibition in the wound healing was accompanied by the inhibition of total length of cell trajectory, total length of the final cell displacement and average speed of cell locomotion. Bafetinib induced 62% mitochondrial membrane depolarization in U87MG cells and 20% depolarization in T98G cells. Higher depolarization levels in U87MG cells correlated with higher apoptosis (49.4%). Treatment of U87MG cells for 4 days with a combination of bafetinib and TMZ reduced the IC50 values as much as 60% and 80%, respectively, compared to the values obtained with each of the drugs, suggesting that bafetinib has the potential to improve the therapeutic effect of TMZ in GBM cells. Conclusion: Our preliminary findings suggest that targeting Lyn and Fyn kinase pathways with bafetinib may have a therapeutic potential for GBM. Our in vivo studies in mouse models with bafetinib will further establish the feasibility of targeting Lyn and Fyn kinases as possible therapeutic targets in GBM. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 1801. doi:1538-7445.AM2012-1801


Archive | 2011

Cardiorenal Protection in Diabetes Mellitus

Himanshu Vashistha; Leonard G. Meggs; Ashwani Malhotra

Diabetic cardiomyopathy is a major complication of diabetes that is independent of high blood pressure or atherosclerosis. In addition to diastolic dysfunction, the diabetic heart is more susceptible to oxidative stress. Hyperglycemia (HG) dominates the pathophysiology and clinical course of type 1 and type 2 Diabetes. An important question concerns the signals used by high concentrations of extracellular glucose to alter the biochemical and mechanical properties of cardiac muscle cells. Recruitment of the Protein Kinase C (PKC) family of serine–threonine kinases is an integral component of the signaling events that direct the cardiac phenotype expressed during postnatal cardiac development and in response to pathological stimuli. We have described that genetically engineered mice with cardiac-specific expression of an isozyme-specific PKC-e translocation activator exhibit protection from hyperglycemia-induced apoptosis and LV dysfunction. The ψe-RACK peptide facilitated the intracellular trafficking of PKC-e, and thereby prevented hyperglycemia-mediated decreases in immunoreactivity in both membrane and mitochondrial compartments. A unifying hypothesis has been proposed for the development of diabetic complications, based on the overproduction of Reactive Oxygen Species (ROS). The adapter protein p66Shc A is a part of a signal transduction pathway and may be a key component of the cell signal response to oxidative stress contributing to the lifespan in mammals. p66ShcA functions as a potentially harmful regulatory gene, which is required for the generation of HG-induced oxidative stress and apoptosis. At high ambient glucose (HG), p66ShcA-deficient cells exhibit resistance to HG-induced ROS generation and attenuation in the amplitude of the kinetic curves for intracellular ROS metabolism, indicative of the pivotal role of WTp66ShcA in the generation of HG oxidant stress. Inhibition of WTp66ShcA function shuts down HG-induced ROS production in cytosolic and mitochondrial compartments.


Circulation Research | 2009

Downregulation of MiR-199a Derepresses Hypoxia-Inducible Factor-1α and Sirtuin 1 and Recapitulates Hypoxia Preconditioning in Cardiac Myocytes

Shweta Rane; Minzhen He; Danish Sayed; Himanshu Vashistha; Ashwani Malhotra; Junichi Sadoshima; Dorothy E. Vatner; Stephen F. Vatner; Maha Abdellatif

Collaboration


Dive into the Himanshu Vashistha's collaboration.

Top Co-Authors

Avatar

Ashwani Malhotra

The Feinstein Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Pravin C. Singhal

The Feinstein Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Maha Abdellatif

University of Medicine and Dentistry of New Jersey

View shared research outputs
Top Co-Authors

Avatar

Kamesh Ayasolla

The Feinstein Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Masao Kakoki

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Nitpriya Paliwal

The Feinstein Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Rukhsana Aslam

The Feinstein Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Vinod Kumar

The Feinstein Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge