Network


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

Hotspot


Dive into the research topics where Apurva Sarin is active.

Publication


Featured researches published by Apurva Sarin.


Journal of Biological Chemistry | 2004

The Anti-apoptotic Effect of Notch-1 Requires p56lck-dependent, Akt/PKB-mediated Signaling in T Cells

Hadassah Sade; Sudhir Krishna; Apurva Sarin

The Notch family of transmembrane receptors have been implicated in a variety of cellular decisions in different cell types. Here we investigate the mechanism underlying Notch-1-mediated anti-apoptotic function in T cells using model cell lines as the experimental system. Ectopic expression of the intracellular domain of Notch-1/activated Notch (AcN1) increases expression of anti-apoptotic proteins of the inhibitors of apoptosis (IAP) family, the Bcl-2 family, and the FLICE-like inhibitor protein (FLIP) and inhibits death triggered by multiple stimuli that activate intrinsic or extrinsic pathways of apoptosis in human and murine T cell lines. Numb inhibited the AcN1-dependent induction of anti-apoptotic proteins and anti-apoptotic function. Using pharmacological inhibitors and dominant-negative approaches, we describe a functional role for phosphatidylinositol 3-kinase (PI3K)-dependent activation of the serine-threonine kinase Akt/PKB in the regulation of AcN1-mediated anti-apoptotic function and the expression of FLIP and IAP family proteins. Using a cell line deficient for the T cell-specific, Src family protein, the tyrosine kinase p56lck and by reconstitution approaches we demonstrate that p56lck is required for the Notch-1-mediated activation of Akt/PKB function. Furthermore, the Src tyrosine kinase inhibitor, PP2, abrogated ectopically expressed AcN1-mediated anti-apoptotic function and phosphorylation of p56lck. We present evidence that endogenous Notch-1 associates with p56lck and PI3K but that Akt/PKB does not co-immunoprecipitate with the Notch1·p56lck·PI3K complex. Finally, we demonstrate that the Notch1·p56lck·PI3K complex is present in primary T cells that have been activated in vitro and sustained in culture with the cytokine interleukin-2.


Biochimica et Biophysica Acta | 2003

VDAC is a conserved element of death pathways in plant and animal systems.

Ashwini Godbole; Jishy Varghese; Apurva Sarin; M. K. Mathew

Programmed cell death (PCD) is very much a part of plant life, although the underlying mechanisms are not so well understood as in animals. In animal cells, the voltage-dependent anion channel (VDAC), a major mitochondrial outer membrane transporter, plays an important role in apoptosis by participating in the release of intermembrane space proteins. To characterize plant PCD pathways by investigating the function of putative components in a mammalian apoptotic context, we have overexpressed a rice VDAC (osVDAC4) in the Jurkat T-cell line. Overexpression of osVDAC4 induces apoptosis, which can be blocked by Bcl-2 and the VDAC inhibitor DIDS. Modifying endogenous VDAC function by DIDS and hexokinase II (HxKII) in Jurkat cells inhibits mitochondria-mediated apoptotic pathways. Finally, we show that DIDS also abrogates heat-induced PCD in cucumber cotyledons. Our data suggest that VDAC is a conserved mitochondrial element of the death machinery in both plant and animal cells.


Developmental Cell | 2007

Role of Mitochondrial Remodeling in Programmed Cell Death in Drosophila melanogaster

Gaurav Goyal; Brennan Fell; Apurva Sarin; Richard J. Youle; V. Sriram

Summary The role of mitochondria in Drosophila programmed cell death remains unclear, although certain gene products that regulate cell death seem to be evolutionarily conserved. We find that developmental programmed cell death stimuli in vivo and multiple apoptotic stimuli ex vivo induce dramatic mitochondrial fragmentation upstream of effector caspase activation, phosphatidylserine exposure, and nuclear condensation in Drosophila cells. Unlike genotoxic stress, a lipid cell death mediator induced an increase in mitochondrial contiguity prior to fragmentation of the mitochondria. Using genetic mutants and RNAi-mediated knockdown of drp-1, we find that Drp-1 not only regulates mitochondrial fission in normal cells, but mediates mitochondrial fragmentation during programmed cell death. Mitochondria in drp-1 mutants fail to fragment, resulting in hyperplasia of tissues in vivo and protection of cells from multiple apoptotic stimuli ex vivo. Thus, mitochondrial remodeling is capable of modifying the propensity of cells to undergo death in Drosophila.


Cell Death & Differentiation | 2009

A hierarchical cascade activated by non-canonical Notch signaling and the mTOR-Rictor complex regulates neglect-induced death in mammalian cells

Lakshmi R. Perumalsamy; M Nagala; P Banerjee; Apurva Sarin

The regulation of cellular metabolism and survival by trophic factors is not completely understood. Here, we describe a signaling cascade activated by the developmental regulator Notch, which inhibits apoptosis triggered by neglect in mammalian cells. In this pathway, the Notch intracellular domain (NIC), which is released after interaction with ligand, converges on the kinase mammalian target of rapamycin (mTOR) and the substrate-defining protein rapamycin independent companion of mTOR (Rictor), culminating in the activation of the kinase Akt/PKB. Biochemical and molecular approaches using site-directed mutants identified AktS473 as a key downstream target in the antiapoptotic pathway activated by NIC. Despite the demonstrated requirement for Notch processing and its predominant nuclear localization, NIC function was independent of CBF1/RBP-J, an essential DNA-binding component required for canonical signaling. In experiments that placed spatial constraints on NIC, enforced nuclear retention abrogated antiapoptotic activity and a membrane-anchored form of NIC-blocked apoptosis through mTOR, Rictor and Akt-dependent signaling. We show that the NIC-mTORC2-Akt cascade blocks the apoptotic response triggered by removal of medium or serum deprivation. Consistently, membrane-tethered NIC, and AktS473 inhibited apoptosis triggered by cytokine deprivation in activated T cells. Thus, this study identifies a non-canonical signaling cascade wherein NIC integrates with multiple pathways to regulate cell survival.


Journal of Clinical Investigation | 2004

Inducible nitric oxide synthase in T cells regulates T cell death and immune memory.

Monika Vig; Smita Srivastava; Usha Kandpal; Hadassah Sade; Virginia Lewis; Apurva Sarin; Anna George; Vineeta Bal; Jeannine M. Durdik; Satyajit Rath

The progeny of T lymphocytes responding to immunization mostly die rapidly, leaving a few long-lived survivors functioning as immune memory. Thus, control of this choice of death versus survival is critical for immune memory. There are indications that reactive radicals may be involved in this death pathway. We now show that, in mice lacking inducible nitric oxide synthase (iNOS), higher frequencies of both CD4 and CD8 memory T cells persist in response to immunization, even when iNOS(+/+) APCs are used for immunization. Postactivation T cell death by neglect is reduced in iNOS(-/-) T cells, and levels of the antiapoptotic proteins Bcl-2 and Bcl-xL are increased. Inhibitors of the iNOS-peroxynitrite pathway also enhance memory responses and block postactivation death by neglect in both mouse and human T cells. However, early primary immune responses are not enhanced, which suggests that altered survival, rather than enhanced activation, is responsible for the persistent immunity observed. Thus, in primary immune responses, iNOS in activated T cells autocrinely controls their susceptibility to death by neglect to determine the level of persisting CD4 and CD8 T cell memory, and modulation of this pathway can enhance the persistence of immune memory in response to vaccination.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Notch-activated signaling cascade interacts with mitochondrial remodeling proteins to regulate cell survival

Lakshmi R. Perumalsamy; Manjula Nagala; Apurva Sarin

Survival of differentiated cells is one of several processes regulated by Notch activity, although the general principles underlying this function remain to be characterized. Here, we probe the mechanism underlying Notch-mediated survival, building on emerging evidence that apoptotic responses coordinated by specialized intermediates converge on mitochondria, identifying a core event in death pathways. The Bcl-2 family protein Bax is one such intermediate, which in a unifying response to diverse apoptotic stimuli nucleates multiprotein assemblies on mitochondria, committing cells to irrevocable damage. Using Bax as the prototype stimulus, we analyze Notch signaling for potential interactions with mitochondria, probe intrinsic properties of the Notch receptor, and describe key intermediates in the Notch-activated signaling cascade. Ligand-dependent processing was necessary to generate the Notch intracellular domain (NIC) although signaling was independent of canonical interactions with nuclear factors. Notably, antiapoptotic activity was recapitulated by NIC recombinants, localized outside the nucleus, and compromised by enforced nuclear sequestration. NIC signaled via the kinase Akt to prevent the loss of mitochondrial function, contiguity, and consequent nuclear damage, outcomes critically depend on mitochondrial remodeling proteins Mitofusins-(Mfn)-1 and 2. Thus, the NIC-Akt-Mfn signaling cascade identifies a pathway regulating cell-survival, independent of canonical functions associated with NIC activity.


Journal of Immunology | 2006

Evidence for a role for notch signaling in the cytokine-dependent survival of activated T cells

Geetha Bheeshmachar; Divya Purushotaman; Hadassah Sade; Vigneshkumar Gunasekharan; Annapoorni Rangarajan; Apurva Sarin

Peripheral T cell homeostasis results from a balance between factors promoting survival and those that trigger deletion of Ag-reactive cells. The cytokine IL-2 promotes T cell survival whereas reactive oxygen species (ROS) sensitize T cells to apoptosis. Two pathways of activated T cell apoptosis–one triggered by Fas ligand and the other by cytokine deprivation–depend on ROS, with the latter also regulated by members of the Bcl-2 family. Notch family proteins regulate several cell-fate decisions in metazoans. Ectopic expression of the Notch1 intracellular domain (NICD) in T cells inhibits Fas-induced apoptosis. The underlying mechanism is not known and the role, if any, of Notch in regulating apoptosis triggered by cytokine deprivation or neglect has not been examined. In this study, we use a Notch1/Fc chimera; a blocking Ab to Notch1 and chemical inhibitors of γ-secretase to investigate the role of Notch signaling in activated T cells of murine origin. We show that perturbing Notch signaling in activated CD4+/CD8+ T cells maintained in IL-2 results in the accumulation of ROS, reduced Akt/protein kinase B activity, and expression of the antiapoptotic protein Bcl-xL, culminating in apoptosis. A broad-spectrum redox scavenger inhibits apoptosis but T cells expressing mutant Fas ligand are sensitive to apoptosis. Activated T cells isolated on the basis of Notch expression (Notch+) are enriched for Bcl-xL expression and demonstrate reduced susceptibility to apoptosis triggered by neglect or oxidative stress. Furthermore, enforced expression of NICD protects activated T cells from apoptosis triggered by cytokine deprivation. Taken together, these data implicate Notch1 signaling in the cytokine-dependent survival of activated T cells.


European Journal of Immunology | 2001

The role of calpain in caspase activation during etoposide induced apoptosis in T cells

Jishy Varghese; G. Radhika; Apurva Sarin

T cells treated with the drug etoposide undergo apoptotic death characterized by early evidence of nuclear damage followed by loss of mitochondrial integrity and cell lysis. Calpains and caspases are cytoplasmic proteases and there is increasing evidence of cross‐talk between these proteases in death pathways. In this study we have investigated the role of calpain, in etoposide‐triggeredapoptosis in the 2B4 murine T cell hybridoma. Cell permeable inhibitors of calpain, ALLnM, E64 and calpeptin that block Fas ligand‐Fas‐mediated death in T cells, blocked etoposide‐induced nuclear damage, loss of mitochondrial integrity and cell lysis. A broad spectrum peptide inhibitor of caspases, ZVAD‐fmk, partially blocked nuclear damage but poorly inhibited mitochondrial damage or cell lysis triggered by etoposide. Etoposide‐induced expression of the cleaved, proteolytically active form of caspase 3, and DEVD‐ase activity, detected prior to nuclear damage, were blocked in the presence of calpain inhibitors. Collectively, these data describe a role for calpain in regulating etoposide‐induced apoptosis via a caspase‐dependent pathway in T cells.


Apoptosis | 2003

Caspase-3 activation is an early event and initiates apoptotic damage in a human leukemia cell line

Jishy Varghese; N. S. Khandre; Apurva Sarin

Many apoptotic pathways culminate in the activation of caspase cascades usually triggered by the apical caspases-8 or -9. We describe a paradigm where apoptosis is initiated by the effector caspase-3. Diethylmaleate (DEM)-induced apoptotic damage in Jurkat cells was blocked by the anti-apoptotic protein Bcl-2, whereas, a peptide inhibitor of caspase-3 but not caspase-9 blocked DEM-induced mitochondrial damage. Isogenic Jurkat cell lines deficient for caspase-8 or the adaptor FADD (Fas associated death domain) were not protected from DEM-induced apoptosis. Caspase-3 activation preceded that of caspase-9 and initial processing of caspase-3 was regulated independent of caspase-9 and Bcl-2. However, inhibitors of caspase-9 or caspase-6 regulated caspase-3 later in the pathway. We explored the mechanism by which caspase-3 processing is regulated in this system. DEM triggered a loss of Erk-1/2 phosphorylation and XIAP (X-linked inhibitor of apoptosis protein) expression. The phorbol ester PMA activated a MEK-dependent pathway to block caspase-3 processing and cell death. Constitutively active MEK-1 (CA-MEK) upregulated XIAP expression and exogenous XIAP inhibited DEM-induced apoptotic damage. Thus, we describe a pathway where caspase-3 functions to initiate apoptotic damage and caspase-9 and caspase-6 amplify the apoptotic cascade. Further, we show that MEK may regulate caspase-3 activation via the regulation of XIAP expression in these cells.


European Journal of Immunology | 2003

IL-7 inhibits dexamethasone-induced apoptosis via Akt/PKB in mature, peripheral T cells

Hadassah Sade; Apurva Sarin

We have investigated the mechanism of IL‐7‐mediated inhibition of dexamethasone‐induced apoptosis in T cells. Broad‐spectrum caspase inhibitors block dexamethasone‐triggered nuclear fragmentation, but not the loss of mitochondrial transmembrane potential or membrane integrity in CD3+ mature T cells isolated from adult mouse spleens. IL‐7 blocked dexamethasone‐induced apoptosis and the processing of caspase‐3 and caspase‐7. IL‐7 also blocked dexamethasone‐triggered dephosphorylation of the serine‐threonine kinase Akt/PKB and its target, the Ser136 residue in Bad. The loss of anti‐apoptotic proteins Bcl‐xL and inhibitor of apoptosis protein‐2 (IAP‐2) was also blocked by IL‐7. The protective effect was attenuated by pharmacological inhibitors of phosphatidylinositol‐3 kinase (PI3K) with one exception: inhibition of PI3K did not abrogate Bcl‐xL expression in the presence of IL‐7. The anti‐apoptotic role of Akt suggested by these experiments was tested by overexpression of constitutively active Akt, which blocked dexamethasone‐induced apoptosis and elevated IAP‐2 but not Bcl‐xL levels in a mature T cell line. Thus, IL‐7 regulates IAP‐2 expression and inhibits dexamethasone‐induced apoptosis by activating Akt via PI3K‐dependent signaling, but regulates Bcl‐xL expression via a PI3K‐independent pathway in mature T cells.

Collaboration


Dive into the Apurva Sarin's collaboration.

Top Co-Authors

Avatar

Hadassah Sade

National Centre for Biological Sciences

View shared research outputs
Top Co-Authors

Avatar

Lakshmi R. Perumalsamy

National Centre for Biological Sciences

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Pierre A. Henkart

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Jishy Varghese

National Centre for Biological Sciences

View shared research outputs
Top Co-Authors

Avatar

G. V. Shivashankar

National University of Singapore

View shared research outputs
Top Co-Authors

Avatar

Megha Garg

National Centre for Biological Sciences

View shared research outputs
Top Co-Authors

Avatar

Neha Parikh

National Centre for Biological Sciences

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge