Network


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

Hotspot


Dive into the research topics where Ashish Nagarsekar is active.

Publication


Featured researches published by Ashish Nagarsekar.


Journal of Immunology | 2005

Febrile-Range Hyperthermia Augments Neutrophil Accumulation and Enhances Lung Injury in Experimental Gram-Negative Bacterial Pneumonia

Penelope Rice; Erica Martin; Ju-Ren He; Mariah Frank; Louis J. DeTolla; Lisa Hester; Timothy O’Neill; Cheu Manka; Ivor J. Benjamin; Ashish Nagarsekar; Ishwar S. Singh; Jeffrey D. Hasday

We previously demonstrated that exposure to febrile-range hyperthermia (FRH) accelerates pathogen clearance and increases survival in murine experimental Klebsiella pneumoniae peritonitis. However, FRH accelerates lethal lung injury in a mouse model of pulmonary oxygen toxicity, suggesting that the lung may be particularly susceptible to injurious effects of FRH. In the present study, we tested the hypothesis that, in contrast with the salutary effect of FRH in Gram-negative peritonitis, FRH would be detrimental in multilobar Gram-negative pneumonia. Using a conscious, temperature-clamped mouse model and intratracheal inoculation with K. pneumoniae Caroli strain, we showed that FRH tended to reduce survival despite reducing the 3 day-postinoculation pulmonary pathogen burden by 400-fold. We showed that antibiotic treatment rescued the euthermic mice, but did not reduce lethality in the FRH mice. Using an intratracheal bacterial endotoxin LPS challenge model, we found that the reduced survival in FRH-treated mice was accompanied by increased pulmonary vascular endothelial injury, enhanced pulmonary accumulation of neutrophils, increased levels of IL-1β, MIP-2/CXCL213, GM-CSF, and KC/CXCL1 in the bronchoalveolar lavage fluid, and bronchiolar epithelial necrosis. These results suggest that FRH enhances innate host defense against infection, in part, by augmenting polymorphonuclear cell delivery to the site of infection. The ultimate effect of FRH is determined by the balance between accelerated pathogen clearance and collateral tissue injury, which is determined, in part, by the site of infection.


American Journal of Respiratory Cell and Molecular Biology | 2013

Interleukin-33 Potentiates Bleomycin-Induced Lung Injury

Irina G. Luzina; Pavel Kopach; Virginia Lockatell; Phillip H. Kang; Ashish Nagarsekar; Allen P. Burke; Jeffrey D. Hasday; Nevins W. Todd; Sergei P. Atamas

The mechanisms of interstitial lung disease (ILD) remain incompletely understood, although recent observations have suggested an important contribution by IL-33. Substantial elevations in IL-33 expression were found in the lungs of patients with idiopathic pulmonary fibrosis and scleroderma lung disease, as well as in the bleomycin injury mouse model. Most of the observed IL-33 expression was intracellular and intranuclear, suggesting involvement of the full-length (fl) protein, but not of the proteolytically processed mature IL-33 cytokine. The effects of flIL-33 on mouse lungs were assessed independently and in combination with bleomycin injury, using recombinant adenovirus-mediated gene delivery. Bleomycin-induced changes were not affected by gene deficiency of the IL-33 receptor T1/ST2. Combined flIL-33 expression and bleomycin injury exerted a synergistic effect on pulmonary lymphocyte and collagen accumulation, which could be explained by synergistic regulation of the cytokines transforming growth factor-β, IL-6, monocyte chemotactic protein-1, macrophage inflammatory protein\x{2013}1α, and tumor necrosis factor-α. By contrast, no increase in the levels of the Th2 cytokines IL-4, IL-5, or IL-13 was evident. Moreover, flIL-33 was found to increase the expression of several heat shock proteins (HSPs) significantly, and in particular HSP70, which is known to be associated with ILD. Thus, flIL-33 is a synergistic proinflammatory and profibrotic regulator that acts by stimulating the expression of several non-Th2 cytokines, and activates the expression of HSP70.


American Journal of Respiratory Cell and Molecular Biology | 2008

Heat Shock Co-Activates Interleukin-8 Transcription

Ishwar S. Singh; Aditi Gupta; Ashish Nagarsekar; Zachary A. Cooper; Cheu Manka; Lisa Hester; Ivor J. Benjamin; Ju Ren He; Jeffrey D. Hasday

The heat shock (HS) response is a phylogenetically ancient cellular response to stress, including heat, that shifts gene expression to a set of conserved HS protein (HSP) genes. In our earlier studies, febrile-range hyperthermia (FRH) not only activated HSP gene expression, but also increased expression of CXC chemokines in mice, leading us to hypothesize that the CXC chemokine family of genes might be HS-responsive. To address this hypothesis we analyzed the effect of HS on the expression of IL-8/CXCL-8, a member of the human CXC family of ELR(+) chemokines. HS markedly enhanced TNF-alpha-induced IL-8 secretion in human A549 respiratory epithelial-like cells and in primary human small airway epithelial cells. IL-8 mRNA was also up-regulated by HS, but the stability of IL-8 mRNA was not affected. TNF-alpha-induced reporter activity of an IL-8 promoter construct IL8(-1471/+44)-luc stably transfected in A549 cells was also enhanced by HS. Electrophoretic mobility and chromatin immunoprecipitation assays showed that the stress-activated transcription factor heat shock factor-1 (HSF-1) binds to at least two putative heat shock response elements (HSE) present in the IL-8 promoter. Deletional reporter constructs lacking either one or both of these sites showed reduced HS responsiveness. Furthermore, depletion of HSF-1 using siRNA also reduced the effects HS on TNF-alpha-induced IL-8 expression, demonstrating that HSF-1 could also act to regulate IL-8 gene transcription. We speculate that during evolution the CXC chemokine genes may have co-opted elements of the HS response to amplify their expression and enhance neutrophil delivery during febrile illnesses.


Immunological Investigations | 2005

CXC chemokines : A new family of heat-shock proteins?

Ashish Nagarsekar; Jeffrey D. Hasday; Ishwar S. Singh

The heat shock (HS) response is a generalized stress response that is characterized by the induced synthesis of a family of proteins referred to as heat shock proteins (HSPs). These proteins protect cells from a myriad of stressful insults in part by functioning as chaperones for denatured proteins. Increasing evidence suggests that the stress response is not limited to the HSP family of genes, but includes numerous other genes that are regulated by HS through the activation of the stress-activated transcription factor, heat shock factor-1 (HSF-1). Based on observations from our own in vivo hyperthermia models, we hypothesized that the CXC chemokine family of neutrophil activators and chemoattractants might be a previously unrecognized class of HS-responsive genes. Analysis of the promoters of the CXC family of chemokines in both human and mouse showed that they share a common promoter organization in which multiple copies of the HSF-1 binding sequence (heat shock response element, HRE) are present in the 5′-upstream flanking region of each of these genes. We have reviewed previous work from our own laboratory and others demonstrating a strong correlation between activation of HSPs and generation of CXC chemokines. Although rigorous experimental evidence is still required to support this hypothesis, this strong and consistent correlation between expression of HSPs and CXC chemokines in vivo and in vitro model systems suggests that the putative HREs present in the CXC chemokine genes are functionally active. We speculate that the activation of the HS response during febrile range hyperthermia, inflammation, infection and injury directly enhances expression of the CXC chemokines, thereby augmenting neutrophil delivery to sites of infection and injury during febrile illnesses.


Journal of Immunology | 2008

Febrile-range hyperthermia accelerates caspase-dependent apoptosis in human neutrophils

Ashish Nagarsekar; Rachel S. Greenberg; Nirav G. Shah; Ishwar S. Singh; Jeffrey D. Hasday

Human neutrophilic polymorphonuclear leukocytes (PMNs) are central to innate immunity and are responsible for clearance of pathogens. PMNs undergo a tightly regulated apoptosis program that allows for timely clearance of PMNs without extravasation of toxic intracellular contents. We investigated the rate of spontaneous apoptosis of human peripheral blood PMNs cultured at basal (37°C) and febrile-range (39.5°C) temperatures (FRT). We found that PMN apoptosis is accelerated at FRT, reaching ∼90% completion by 8 h at 39.5°C vs 18 h at 37°C based on morphologic criteria. Caspase-8 activation peaked within 15 min of PMN exposure to FRT, and subsequent activation of caspase-3 and -9, cleavage of the BH3 (Bcl-2 homology domain 3) only protein Bid, and mitochondrial release of cytochrome c were also greater in FRT-exposed PMNs. Inhibition of caspase-3, -8, and -9 conferred comparable protection from apoptosis in FRT-exposed PMNs. These results demonstrate that exposure to FRT enhances caspase-8 activation and subsequent mitochondrial-dependent and mitochondrial-independent apoptosis pathways. The PMN survival factors G-CSF, GM-CSF, and IL-8 each prolonged PMN survival at 37°C and 39.5°C, but did not reduce the difference in survival at the two temperatures. In a mouse model of intratracheal endotoxin-induced alveolitis, coexposure to FRT (core temperature ∼39.5°C) doubled the proportion of bronchoalveolar PMNs undergoing apoptosis compared with euthermic mice. This process may play an important role in limiting inflammation and tissue injury during febrile illnesses.


American Journal of Respiratory Cell and Molecular Biology | 2012

Hyperthermia Promotes and Prevents Respiratory Epithelial Apoptosis through Distinct Mechanisms

Ashish Nagarsekar; Mohan E. Tulapurkar; Ishwar S. Singh; Sergei P. Atamas; Nirav G. Shah; Jeffrey D. Hasday

Hyperthermia has been shown to confer cytoprotection and to augment apoptosis in different experimental models. We analyzed the mechanisms of both effects in the same mouse lung epithelial (MLE) cell line (MLE15). Exposing MLE15 cells to heat shock (HS; 42°C, 2 h) or febrile-range hyperthermia (39.5°C) concurrent with activation of the death receptors, TNF receptor 1 or Fas, greatly accelerated apoptosis, which was detectable within 30 minutes and was associated with accelerated activation of caspase-2, -8, and -10, and the proapoptotic protein, Bcl2-interacting domain (Bid). Caspase-3 activation and cell death were partially blocked by inhibitors targeting all three initiator caspases. Cells expressing the IκB superrepessor were more susceptible than wild-type cells to TNF-α-induced apoptosis at 37°C, but HS and febrile-range hyperthermia still increased apoptosis in these cells. Delaying HS for 3 hours after TNF-α treatment abrogated its proapoptotic effect in wild-type cells, but not in IκB superrepressor-expression cells, suggesting that TNF-α stimulates delayed resistance to the proapoptotic effects of HS through an NF-κB-dependent mechanism. Pre-exposure to 2-hour HS beginning 6 to16 hours before TNF-α treatment or Fas activation reduced apoptosis in MLE15 cells. The antiapoptotic effects of HS pretreatment were reduced in TNF-α-treated embryonic fibroblasts from heat shock factor-1 (HSF1)-deficient mice, but the proapoptotic effects of concurrent HS were preserved. Thus, depending on the temperature and timing relative to death receptor activation, hyperthermia can exert pro- and antiapoptotic effects through distinct mechanisms.


American Journal of Physiology-lung Cellular and Molecular Physiology | 2016

Activation of heat shock response augments fibroblast growth factor-1 expression in wounded lung epithelium

Rachel G. Scheraga; Christopher Thompson; Mohan E. Tulapurkar; Ashish Nagarsekar; Mark J. Cowan; Ratnakar Potla; Junfeng Sun; Rongman Cai; Carolea Logun; James H. Shelhamer; Nevins W. Todd; Ishwar S. Singh; Irina G. Luzina; Sergei P. Atamas; Jeffrey D. Hasday

We previously showed that coincident exposure to heat shock (HS; 42°C for 2 h) and TNF-α synergistically induces apoptosis in mouse lung epithelium. We extended this work by analyzing HS effects on human lung epithelial responses to clinically relevant injury. Cotreatment with TNF-α and HS induced little caspase-3 and poly(ADP-ribose) polymerase cleavage in human small airway epithelial cells, A549 cells, and BEAS2B cells. Scratch wound closure rates almost doubled when A549 and BEAS2B cells and air-liquid interface cultures of human bronchial epithelial cells were heat shocked immediately after wounding. Microarray, qRT-PCR, and immunoblotting showed fibroblast growth factor 1 (FGF1) to be synergistically induced by HS and wounding. Enhanced FGF1 expression in HS/wounded A549 was blocked by inhibitors of p38 MAPK (SB203580) or HS factor (HSF)-1 (KNK-437) and in HSF1 knockout BEAS2B cells. PCR demonstrated FGF1 to be expressed from the two most distal promoters in wounded/HS cells. Wound closure in HS A549 and BEAS2B cells was reduced by FGF receptor-1/3 inhibition (SU-5402) or FGF1 depletion. Exogenous FGF1 accelerated A549 wound closure in the absence but not presence of HS. In the presence of exogenous FGF1, HS slowed wound closure, suggesting that it increases FGF1 expression but impairs FGF1-stimulated wound closure. Frozen sections from normal and idiopathic pulmonary fibrosis (IPF) lung were analyzed for FGF1 and HSP70 by immunofluorescence confocal microscopy and qRT-PCR. FGF1 and HSP70 mRNA levels were 7.5- and 5.9-fold higher in IPF than normal lung, and the proteins colocalized to fibroblastic foci in IPF lung. We conclude that HS signaling may have an important impact on gene expression contributing to lung injury, healing, and fibrosis.


Biomacromolecules | 2003

Genetic engineering of stimuli-sensitive silkelastin-like protein block copolymers.

Ashish Nagarsekar; John Crissman; Mary Crissman; Franco A. Ferrari; Joseph Cappello; Hamidreza Ghandehari


Journal of Biomedical Materials Research | 2002

Genetic synthesis and characterization of pH- and temperature-sensitive silk-elastinlike protein block copolymers

Ashish Nagarsekar; John Crissman; Mary Crissman; Franco A. Ferrari; Joseph Cappello; Hamidreza Ghandehari


Transactions of the American Clinical and Climatological Association | 2011

FEVER, HYPERTHERMIA, AND THE LUNG: IT'S ALL ABOUT CONTEXT AND TIMING

Jeffrey D. Hasday; Nirav G. Shah; Phillip A. Mackowiak; Mohan E. Tulapurkar; Ashish Nagarsekar; Ishwar S. Singh

Collaboration


Dive into the Ashish Nagarsekar's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Cheu Manka

University of Maryland

View shared research outputs
Researchain Logo
Decentralizing Knowledge