Network


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

Hotspot


Dive into the research topics where Dheeraj Soni is active.

Publication


Featured researches published by Dheeraj Soni.


Nature Immunology | 2014

The transcription factor DREAM represses the deubiquitinase A20 and mediates inflammation

Chinnaswamy Tiruppathi; Dheeraj Soni; Dong Mei Wang; Jiaping Xue; Vandana Singh; Prabhakar B. Thippegowda; Bopaiah P. Cheppudira; Rakesh K. Mishra; Auditi DebRoy; Zhijian Qian; Kurt Bachmaier; You Yang Zhao; John W. Christman; Stephen M. Vogel; Averil Ma; Asrar B. Malik

Here we found that the transcription repressor DREAM bound to the promoter of the gene encoding A20 to repress expression of this deubiquitinase that suppresses inflammatory NF-κB signaling. DREAM-deficient mice displayed persistent and unchecked A20 expression in response to endotoxin. DREAM functioned by transcriptionally repressing A20 through binding to downstream regulatory elements (DREs). In contrast, binding of the transcription factor USF1 to the DRE-associated E-box domain in the gene encoding A20 activated its expression in response to inflammatory stimuli. Our studies define the critical opposing functions of DREAM and USF1 in inhibiting and inducing A20 expression, respectively, and thereby the strength of NF-κB signaling. Targeting of DREAM to induce USF1-mediated A20 expression is therefore a potential anti-inflammatory strategy for the treatment of diseases associated with unconstrained NF-κB activity, such as acute lung injury.


Journal of Biological Chemistry | 2014

Cooperative signaling via transcription factors NF-κB and AP1/c-Fos mediates endothelial cell STIM1 expression and hyperpermeability in response to endotoxin.

Auditi DebRoy; Stephen M. Vogel; Dheeraj Soni; Premanand Sundivakkam; Asrar B. Malik; Chinnaswamy Tiruppathi

Background: STIM1 activates store-operated Ca2+ entry (SOCE). The transcriptional regulation of STIM1 during sepsis is not known. Results: STIM1 expression occurs downstream of TLR4 via activation of NF-κB and p38α-mediated c-Fos expression in endothelial cells. Conclusion: Cooperative signaling of NF-κB and p38 MAPK downstream of TLR4 is essential for STIM1 expression during sepsis. Significance: Selective p38α inhibitors may represent a potential therapeutic strategy to prevent sepsis-mediated lung vascular leaks. Stromal interacting molecule 1 (STIM1) regulates store-operated Ca2+ entry (SOCE). Here we show that STIM1 expression in endothelial cells (ECs) is increased during sepsis and, therefore, contributes to hyperpermeability. LPS induced STIM1 mRNA and protein expression in human and mouse lung ECs. The induced STIM1 expression was associated with augmented SOCE as well as a permeability increase in both in vitro and in vivo models. Because activation of both the NF-κB and p38 MAPK signaling pathways downstream of TLR4 amplifies vascular inflammation, we studied the influence of these two pathways on LPS-induced STIM1 expression. Inhibition of either NF-κB or p38 MAPK activation by pharmacological agents prevented LPS-induced STIM1 expression. Silencing of the NF-κB proteins (p65/RelA or p50/NF-κB1) or the p38 MAPK isoform p38α prevented LPS-induced STIM1 expression and increased SOCE in ECs. In support of these findings, we found NF-κB and AP1 binding sites in the 5′-regulatory region of human and mouse STIM1 genes. Further, we demonstrated that LPS induced time-dependent binding of the transcription factors NF-κB (p65/RelA) and AP1 (c-Fos/c-Jun) to the STIM1 promoter. Interestingly, silencing of c-Fos, but not c-Jun, markedly reduced LPS-induced STIM1 expression in ECs. We also observed that silencing of p38α prevented c-Fos expression in response to LPS in ECs, suggesting that p38α signaling mediates the expression of c-Fos. These results support the proposal that cooperative signaling of both NF-κB and AP1 (via p38α) amplifies STIM1 expression in ECs and, thereby, contributes to the lung vascular hyperpermeability response during sepsis.


Circulation Research | 2017

Neutrophil Activation of Endothelial Cell-Expressed TRPM2 Mediates Transendothelial Neutrophil Migration and Vascular Injury.

Manish Mittal; Saroj Nepal; Yoshikazu Tsukasaki; Claudie Hecquet; Dheeraj Soni; Jalees Rehman; Chinnaswamy Tiruppathi; Asrar B. Malik

Rationale: TRPM2 (transient receptor potential melastatin-2) expressed in endothelial cells (ECs) is a cation channel mediating Ca2+ entry in response to intracellular generation of adenosine diphosphoribose—the TRPM2 ligand. Objective: Because polymorphonuclear neutrophils (PMN) interaction with ECs generates reactive oxygen species, we addressed the possible role of TRPM2 expressed in ECs in the mechanism of transendothelial migration of PMNs. Methods and Results: We observed defective PMN transmigration in response to lipopolysaccharide challenge in adult mice in which the EC expressed TRPM2 is conditionally deleted (Trpm2i&Dgr;EC). PMN interaction with ECs induced the entry of Ca2+ in ECs via the EC-expressed TRPM2. Prevention of generation of adenosine diphosphoribose in ECs significantly reduced Ca2+ entry in response to PMN activation of TRPM2 in ECs. PMNs isolated from gp91phox−/− mice significantly reduced Ca2+ entry in ECs via TRPM2 as compared with wild-type PMNs and failed to induce PMN transmigration. Overexpression of the adenosine diphosphoribose insensitive TRPM2 mutant channel (C1008→A) in ECs suppressed the Ca2+ entry response. Further, the forced expression of TRPM2 mutant channel (C1008→A) or silencing of poly ADP-ribose polymerase in ECs of mice prevented PMN transmigration. Conclusions: Thus, endotoxin-induced transmigration of PMNs was secondary to TRPM2-activated Ca2+ signaling and VE-cadherin phosphorylation resulting in the disassembly of adherens junctions and opening of the paracellular pathways. These results suggest blocking TRPM2 activation in ECs is a potentially important means of therapeutically modifying PMN-mediated vascular inflammation.


Proceedings of the National Academy of Sciences of the United States of America | 2016

TNFα-stimulated gene-6 (TSG6) activates macrophage phenotype transition to prevent inflammatory lung injury.

Manish Mittal; Chinnaswamy Tiruppathi; Saroj Nepal; You Yang Zhao; Dagmara Grzych; Dheeraj Soni; Darwin J. Prockop; Asrar B. Malik

Significance We found that TNFα-stimulated gene-6 (TSG6), a secreted 30-kDa immunomodulatory protein, resolved LPS-induced inflammatory lung injury by shifting macrophages from a proinflammatory to an anti-inflammatory phenotype. Macrophages from mice genetically deficient in TSG6 failed to transition, demonstrating the essential role of TSG6 in mediating macrophage plasticity. The finding that TSG6 induced the marked transition in anti-inflammatory macrophages lays the foundation for its therapeutic application. TNFα-stimulated gene-6 (TSG6), a 30-kDa protein generated by activated macrophages, modulates inflammation; however, its mechanism of action and role in the activation of macrophages are not fully understood. Here we observed markedly augmented LPS-induced inflammatory lung injury and mortality in TSG6−/− mice compared with WT (TSG6+/+) mice. Treatment of mice with intratracheal instillation of TSG6 prevented LPS-induced lung injury and neutrophil sequestration, and increased survival in mice. We found that TSG6 inhibited the association of TLR4 with MyD88, thereby suppressing NF-κB activation. TSG6 also prevented the expression of proinflammatory proteins (iNOS, IL-6, TNFα, IL-1β, and CXCL1) while increasing the expression of anti-inflammatory proteins (CD206, Chi3l3, IL-4, and IL-10) in macrophages. This shift was associated with suppressed activation of proinflammatory transcription factors STAT1 and STAT3. In addition, we observed that LPS itself up-regulated the expression of TSG6 in TSG6+/+ mice, suggesting an autocrine role for TSG6 in transitioning macrophages. Thus, TSG6 functions by converting macrophages from a proinflammatory to an anti-inflammatory phenotype secondary to suppression of TLR4/NF-κB signaling and STAT1 and STAT3 activation.


Biophysical Journal | 2017

Molecular-Scale Biophysical Modulation of an Endothelial Membrane by Oxidized Phospholipids

Elizabeth LeMaster; Tzu Pin Shentu; Dev K. Singh; Nicolas Barbera; Dheeraj Soni; Chinnaswamy Tiruppathi; Papasani V. Subbaiah; Evgeny Berdyshev; Irina Bronova; Michael Cho; Belinda S. Akpa; Irena Levitan

The influence of two bioactive oxidized phospholipids on model bilayer properties, membrane packing, and endothelial cell biomechanics was investigated computationally and experimentally. The truncated tail phospholipids, 1-palmitoyl-2-(5-oxovaleroyl)-sn-glycero-3-phosphocholine (POVPC) and 1-palmitoyl-2-glutaroyl-sn-glycero-3-phosphocholine (PGPC), are two major oxidation products of the unsaturated phospholipid 1-palmitoyl-2-arachidonoyl-sn-glycero-phosphocholine. A combination of coarse-grained molecular dynamics simulations, Laurdan multiphoton imaging, and atomic force microscopy microindentation experiments was used to determine the impact of POVPC and PGPC on the structure of a multicomponent phospholipid bilayer and to assess the consequences of their incorporation on membrane packing and endothelial cell stiffness. Molecular simulations predicted differential bilayer perturbation effects of the two oxidized phospholipids based on the chemical identities of their truncated tails, including decreased bilayer packing, decreased bilayer bending modulus, and increased water penetration. Disruption of lipid order was consistent with Laurdan imaging results indicating that POVPC and PGPC decrease the lipid packing of both ordered and disordered membrane domains. Computational predictions of a larger membrane perturbation effect by PGPC correspond to greater stiffness of PGPC-treated endothelial cells observed by measuring cellular elastic moduli using atomic force microscopy. Our results suggest that disruptions in membrane structure by oxidized phospholipids play a role in the regulation of overall endothelial cell stiffness.


American Journal of Physiology-lung Cellular and Molecular Physiology | 2017

Pyk2 phosphorylation of VE-PTP downstream of STIM1-induced Ca2+ entry regulates disassembly of adherens junctions

Dheeraj Soni; Sushil C. Regmi; Dong Mei Wang; Auditi DebRoy; You Yang Zhao; Stephen M. Vogel; Asrar B. Malik; Chinnaswamy Tiruppathi

Vascular endothelial protein tyrosine phosphatase (VE-PTP) stabilizes endothelial adherens junctions (AJs) through constitutive dephosphorylation of VE-cadherin. Here we investigated the role of stromal interaction molecule 1 (STIM1) activation of store-operated Ca2+ entry (SOCE) in regulating AJ assembly. We observed that SOCE induced by STIM1 activated Pyk2 in human lung microvascular endothelial cells (ECs) and induced tyrosine phosphorylation of VE-PTP at Y1981. Pyk2-induced tyrosine phosphorylation of VE-PTP promoted Src binding to VE-PTP, Src activation, and subsequent VE-cadherin phosphorylation and thereby increased the endothelial permeability response. The increase in permeability was secondary to disassembly of AJs. Pyk2-mediated responses were blocked in EC-restricted Stim1 knockout mice, indicating the requirement for STIM1 in initiating the signaling cascade. A peptide derived from the Pyk2 phosphorylation site on VE-PTP abolished the STIM1/SOCE-activated permeability response. Thus Pyk2 activation secondary to STIM1-induced SOCE causes tyrosine phosphorylation of VE-PTP, and VE-PTP, in turn, binds to and activates Src, thereby phosphorylating VE-cadherin to increase endothelial permeability through disassembly of AJs. Our results thus identify a novel signaling mechanism by which STIM1-induced Ca2+ signaling activates Pyk2 to inhibit the interaction of VE-PTP and VE-cadherin and hence increase endothelial permeability. Therefore, targeting the Pyk2 activation pathway may be a potentially important anti-inflammatory strategy.


Cell death discovery | 2018

Deubiquitinase function of A20 maintains and repairs endothelial barrier after lung vascular injury

Dheeraj Soni; DongMei Wang; Sushil C. Regmi; Manish Mittal; Stephen M. Vogel; Dirk Schlüter; Chinnaswamy Tiruppathi

Vascular endothelial cadherin (VE-cad) expression at endothelial adherens junctions (AJs) regulates vascular homeostasis. Here we show that endothelial A20 is required for VE-cad expression at AJs to maintain and repair the injured endothelial barrier. In endothelial cell (EC)-restricted Tnfaip3 (A20) knockout (A20∆EC) mice, LPS challenge caused uncontrolled lung vascular leak and persistent sequestration of polymorphonuclear neutrophil (PMNs). Importantly, A20∆EC mice exhibited drastically reduced VE-cad expression in lungs compared with wild-type counterparts. Endothelial expression of wild-type A20 but not the deubiquitinase-inactive A20 mutant (A20C103A) prevented VE-cad ubiquitination, restored VE-cad expression, and suppressed lung vascular leak in A20∆EC mice. Interestingly, IRAK-M-mediated nuclear factor-κB (NF-κB) signaling downstream of TLR4 was required for A20 expression in ECs. interleukin-1 receptor-associated kinase M (IRAK-M) knockdown suppressed basal and LPS-induced A20 expression in ECs. Further, in vivo silencing of IRAK-M in mouse lung vascular ECs through the CRISPR-Cas9 system prevented expression of A20 and VE-cad while augmenting lung vascular leak. These results suggest that targeting of endothelial A20 is a potential therapeutic strategy to restore endothelial barrier integrity in the setting of acute lung injury.


Circulation Research | 2017

Response by Mittal et al to Letter Regarding Article, “Neutrophil Activation of Endothelial Cell-Expressed TRPM2 Mediates Transendothelial Neutrophil Migration and Vascular Injury”

Manish Mittal; Saroj Nepal; Yoshikazu Tsukasaki; Claudie Hecquet; Dheeraj Soni; Chinnaswamy Tiruppathi; Asrar B. Malik; Jalees Rehman

If the intent of the letter by Gagat and Grzamka is to point out that actin cytoskeleton remodeling induces vascular barrier disruption is an important phenomenon, we fully agree.1 Its clear tight junctions have a role in regulating endothelial barrier in the brain endothelial cells, although their role is less important in the continuous endothelium such as in the heart and lungs.1 Recent knock out mouse model studies2,3 (including ours4) have provided compelling evidence that adherens junctions or VE-cadherin junctions are important in regulating leukocyte transmigration and …


The FASEB Journal | 2016

Ca2+-dependent Pyk2 Activation Destabilizes Endothelial Adherens Junctions by Disrupting Interaction between VE-PTP and VE-Cadherin

Dheeraj Soni; Auditi DebRoy; DongMei Wang; Stephen M. Vogel; Chinnaswamy Tiruppathi


The FASEB Journal | 2015

TAK1 Signaling Downstream of PAR-1 in Endothelial Cells Restores Lung Vascular Barrier Integrity

Dheeraj Soni; Auditi DebRoy; DongMei Wang; Stephen M. Vogel; Chinnaswamy Tiruppathi

Collaboration


Dive into the Dheeraj Soni's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Stephen M. Vogel

University of Illinois at Chicago

View shared research outputs
Top Co-Authors

Avatar

Auditi DebRoy

University of Illinois at Chicago

View shared research outputs
Top Co-Authors

Avatar

Manish Mittal

University of Illinois at Chicago

View shared research outputs
Top Co-Authors

Avatar

DongMei Wang

University of Illinois at Chicago

View shared research outputs
Top Co-Authors

Avatar

Saroj Nepal

University of Illinois at Chicago

View shared research outputs
Top Co-Authors

Avatar

You Yang Zhao

University of Illinois at Chicago

View shared research outputs
Top Co-Authors

Avatar

Claudie Hecquet

University of Illinois at Chicago

View shared research outputs
Top Co-Authors

Avatar

Dong Mei Wang

University of Illinois at Chicago

View shared research outputs
Researchain Logo
Decentralizing Knowledge