Network


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

Hotspot


Dive into the research topics where Ranjeet K. Sinha is active.

Publication


Featured researches published by Ranjeet K. Sinha.


Blood | 2012

Biased agonism of protease-activated receptor 1 by activated protein C caused by noncanonical cleavage at Arg46

Laurent O. Mosnier; Ranjeet K. Sinha; Laurent Burnier; Eveline A. Bouwens

Activated protein C (APC) exerts endothelial cytoprotective actions that require protease-activated receptor 1 (PAR1), whereas thrombin acting via PAR1 causes endothelial disruptive, proinflammatory actions. APCs activities, but not thrombins, require PAR1 located in caveolae. PAR1 is a biased 7-transmembrane receptor because G proteins mediate thrombins signaling, whereas β-arrestin 2 mediates APCs signaling. Here we elucidate novel mechanisms for APCs initiation of signaling. Biochemical studies of APCs protease specificity showed that APC cleaved PAR1 sequences at both Arg41 and Arg46. That PAR1 cleavage at Arg46 can occur on cells was supported by APCs cleavage of N-terminal-SEAP-tagged R41Q-PAR1 but not R41Q/R46Q-PAR1 mutants transfected into cells and by anti-PAR1 epitope mapping of APC-treated endothelial cells. A synthetic peptide composing PAR1 residues 47-66, TR47, stimulated protective signaling in endothelial cells as reflected in Akt and glycogen synthase kinase 3β phosphorylation, Ras-related C3 botulinum toxin substrate 1 activation, and barrier stabilization effects. In mice, the TR47 peptide reduced VEGF-induced vascular leakage. These in vitro and in vivo data imply that the novel PAR1 N-terminus beginning at residue Asn47, which is generated by APC cleavage at Arg46, mediates APCs cytoprotective signaling and that this unique APC-generated N-terminal peptide tail is a novel biased agonist for PAR1.


Carcinogenesis | 2015

Disruptive environmental chemicals and cellular mechanisms that confer resistance to cell death

Kannan Badri Narayanan; Manaf Ali; Barry J. Barclay; Qiang Shawn Cheng; Leandro S. D’Abronzo; Rita Dornetshuber-Fleiss; Paramita M. Ghosh; Michael J. Gonzalez Guzman; Tae-Jin Lee; Po Sing Leung; Lin Li; Suidjit Luanpitpong; Edward Ratovitski; Yon Rojanasakul; Maria Fiammetta Romano; Simona Romano; Ranjeet K. Sinha; Clement Yedjou; Fahd Al-Mulla; Rabeah Al-Temaimi; Amedeo Amedei; Dustin G. Brown; Elizabeth P. Ryan; Annamaria Colacci; Roslida A. Hamid; Chiara Mondello; Jayadev Raju; Hosni K. Salem; Jordan Woodrick; A.Ivana Scovassi

Cell death is a process of dying within biological cells that are ceasing to function. This process is essential in regulating organism development, tissue homeostasis, and to eliminate cells in the body that are irreparably damaged. In general, dysfunction in normal cellular death is tightly linked to cancer progression. Specifically, the up-regulation of pro-survival factors, including oncogenic factors and antiapoptotic signaling pathways, and the down-regulation of pro-apoptotic factors, including tumor suppressive factors, confers resistance to cell death in tumor cells, which supports the emergence of a fully immortalized cellular phenotype. This review considers the potential relevance of ubiquitous environmental chemical exposures that have been shown to disrupt key pathways and mechanisms associated with this sort of dysfunction. Specifically, bisphenol A, chlorothalonil, dibutyl phthalate, dichlorvos, lindane, linuron, methoxychlor and oxyfluorfen are discussed as prototypical chemical disruptors; as their effects relate to resistance to cell death, as constituents within environmental mixtures and as potential contributors to environmental carcinogenesis.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2016

Apolipoprotein E Receptor 2 Mediates Activated Protein C–Induced Endothelial Akt Activation and Endothelial Barrier Stabilization

Ranjeet K. Sinha; Xia V. Yang; José A. Fernández; Xiao Xu; Laurent O. Mosnier

Objective— Activated protein C (APC), a plasma serine protease, initiates cell signaling that protects endothelial cells from apoptosis and endothelial barrier disruption. Apolipoprotein E receptor 2 (ApoER2; LRP8) is a receptor known for mediating signaling initiated by reelin in neurons. ApoER2 contributes to APC-initiated signaling in monocytic U937 cells. The objective was to determine whether ApoER2 is required for APC’s beneficial signaling in the endothelial cell surrogate EA.hy926 line. Approach and Results— We used small interfering RNA and inhibitors to probe requirements for specific receptors for APC’s antiapoptotic activity and for phosphorylation of disabled-1 by Src family kinases and of Akt. When small interfering RNA for ApoER2 or endothelial cell protein C receptor or protease activated receptor 1 was used, APC’s antiapoptotic activity was ablated, indicating that each of these receptors was required. In EA.hy926 cells, APC induced a 2- to 3-fold increased phosphorylation of Ser473-Akt and Tyr232-disabled-1, a phosphorylation known to trigger disabled-1–mediated signaling in other cell types. Ser473-Akt phosphorylation was inhibited by ApoER2 small interfering RNA or by inhibitors of Src (PP2), phosphatidylinositol-3 kinase (LY303511), and protease activated receptor 1 (SCH79797). ApoER2 small interfering RNA blocked the ability of APC to prevent thrombin-induced endothelial barrier disruption in TransEndothelial Resistance assays. Binding studies using purified APC and purified immobilized wild-type and mutated ApoER2 ectodomains suggested that APC binding involves Lys49, Asp50, and Trp64 on the surface of the N-terminal LA1 domain of ApoER2. Conclusions— ApoER2 contributes cooperatively with endothelial cell protein C receptor and protease activated receptor 1 to APC-initiated endothelial antiapoptotic and barrier protective signaling.


Blood | 2018

PAR1 biased signaling is required for activated protein C in vivo benefits in sepsis and stroke

Ranjeet K. Sinha; Yaoming Wang; Zhen Zhao; Xiao Xu; Laurent Burnier; Naveen Gupta; José A. Fernández; Greg Martin; Sergey Kupriyanov; Laurent O. Mosnier; Berislav V. Zlokovic

Activated protein C (APC) cleaves protease-activated receptor 1 (PAR1) in vitro at R46 to initiate beneficial cell signaling; however, thrombin and APC can cleave at R41. To elucidate PAR1-dependent aspects of the pharmacologic in vivo mechanisms of APC, we generated C57BL/6 mouse strains carrying QQ41 or QQ46 point mutations in PAR1 (F2r gene). Using these strains, we determined whether or not recombinant murine signaling-selective APC mutants would reduce septic death or provide neuroprotection against ischemic stroke when mice carried PAR1-homozygous mutations that prevent cleavage at either R41 or R46. Intercrossing PAR1+/R46Q mice generated expected numbers of PAR1+/+, PAR1+/R46Q, and R46Q/R46Q offspring whereas intercrossing PAR1+/R41Q mice gave decreased R41Q/R41Q homozygotes (resembling intercrossing PAR1+/PAR1-knockout mice). QQ41-PAR1 and QQ46-PAR1 brain endothelial cells showed the predicted retention or loss of cellular responses to thrombin receptor-activating peptide, thrombin, or APC for each PAR1 mutation. In sepsis studies, exogenous APC reduced mortality from 50% to 10% in Escherichia coli-induced pneumonia for wild-type (Wt) PAR1 and QQ41-PAR1 mice (P < .01) but had no benefit for QQ46-PAR1 mice. In transient distal middle cerebral artery occlusion stroke studies, exogenous APC significantly reduced infarct size, edema, and neuronal apoptosis for Wt mice and QQ41-PAR1 mice but had no detectable benefits for mice carrying QQ46-PAR1. In functional studies of forelimb-asymmetry and foot-fault tests at 24 hours after stroke induction, signaling-selective APC was beneficial for Wt and QQ41-PAR1 mice but not QQ46-PAR1 mice. These results support the concept that APC-induced, PAR1-dependent biased signaling following R46 cleavage is central to the in vivo benefits of APC.


Blood Cells Molecules and Diseases | 2013

Neurotoxicity of the anticoagulant-selective E149A-activated protein C variant after focal ischemic stroke in mice.

Yaoming Wang; Ranjeet K. Sinha; Laurent O. Mosnier; Berislav V. Zlokovic

Wild type (WT) activated protein C (APC) and cytoprotective-selective APC variants such as 3K3A-APC (<10% anticoagulant but normal cytoprotective activity) are neuroprotective in murine focal ischemic stroke models. Here we compared the neuroprotective effects of the anticoagulant-selective E149A-APC variant (>3-fold increased anticoagulant activity but defective cytoprotective activities) to those of the cytoprotective-selective 5A-APC variant (<10% anticoagulant activity). After transient distal middle cerebral artery occlusion, mice received a vehicle, E149A-APC or 5A-APC at 0.2mg/kg at 4h after stroke. Treatment with 5A-APC was neuroprotective, as it improved performance on forelimb use asymmetry test and foot fault test (P<0.05), reduced by 48% and 50% the infarct and edema volumes, respectively (P<0.05), and was not associated with an increased risk of bleeding as indicated by normal hemoglobin levels in the ischemic brain at day 7. In contrast, E149A-APC treatment worsened neurological outcome determined by foot fault tests and forelimb use asymmetry tests, and increased significantly by 44% and 60% infarct and edema volume, respectively (P<0.05). At 7days after treatment, E149A-APC compared to vehicle or 5A-APC notably increased by ~5-fold the hemoglobin level in the ischemic hemisphere suggesting it provoked significant intracerebral bleeding. Thus, the enhanced anticoagulant activity of E149A-APC increased post-ischemic accumulation of neurotoxic erythrocyte-derived hemoglobin which likely worsened the neurological and neuropathological outcomes after stroke. Our data emphasize that APCs cytoprotective activities, but not its anticoagulant activity, are key for APC neuroprotection after transient ischemic stroke.


Blood | 2016

Prothrombotic skeletal muscle myosin directly enhances prothrombin activation by binding factors Xa and Va.

Hiroshi Deguchi; Ranjeet K. Sinha; Patrizia Marchese; Zaverio M. Ruggeri; Jevgenia Zilberman-Rudenko; Owen J. T. McCarty; Mitchell J. Cohen

To test the hypothesis that skeletal muscle myosins can directly influence blood coagulation and thrombosis, ex vivo studies of the effects of myosin on thrombogenesis in fresh human blood were conducted. Addition of myosin to blood augmented the thrombotic responses of human blood flowing over collagen-coated surfaces (300 s-1 shear rate). Perfusion of human blood over myosin-coated surfaces also caused fibrin and platelet deposition, evidencing myosins thrombogenicity. Myosin markedly enhanced thrombin generation in both platelet-rich plasma and platelet-poor plasma, indicating that myosin promoted thrombin generation in plasma primarily independent of platelets. In purified reaction mixtures composed only of factor Xa, factor Va, prothrombin, and calcium ions, myosin greatly enhanced prothrombinase activity. The Gla domain of factor Xa was not required for myosins prothrombinase enhancement. When binding of purified clotting factors to immobilized myosin was monitored using biolayer interferometry, factors Xa and Va each showed favorable binding interactions. Factor Va reduced by 100-fold the apparent Kd of myosin for factor Xa (Kd ∼0.48 nM), primarily by reducing koff, indicating formation of a stable ternary complex of myosin:Xa:Va. In studies to assess possible clinical relevance for this discovery, we found that antimyosin antibodies inhibited thrombin generation in acute trauma patient plasmas more than in control plasmas (P = .0004), implying myosin might contribute to acute trauma coagulopathy. We posit that myosin enhancement of thrombin generation could contribute either to promote hemostasis or to augment thrombosis risk with consequent implications for myosins possible contributions to pathophysiology in the setting of acute injuries.


Stem Cells | 2018

The TLR4-PAR1 Axis Regulates Bone Marrow Mesenchymal Stromal Cell Survival and Therapeutic Capacity in Murine E. coli Pneumonia

Naveen Gupta; Ranjeet K. Sinha; Anna Krasnodembskaya; Xiao Xu; Victor Nizet; Michael A. Matthay; G Griffin

Bone marrow derived mesenchymal stromal cells have been shown to have significant therapeutic effects in experimental models of pneumonia and lung injury. The current study examined the roles of the toll like receptor 4 (TLR4) and protease activated receptor 1 (PAR1) pathways on mesenchymal stromal cell (MSC) survival and therapeutic activity in a murine model of pneumonia. MSCs from TLR4 ‐/‐ and R41Q‐PAR1 mutated mice were isolated to test the effect of mutating these specific pathways on MSC survival when exposed to cytotoxic stimuli in vitro. An Escherichia coli pneumonia model was used to assess the effect of these specific pathways on MSC therapeutic activity in vivo. Our results showed that mutation of either the TLR4 or PAR1 pathways in MSCs impaired cell survival under conditions of inflammatory stress in vitro, and eliminated their therapeutic efficacy in vivo. Also, stimulation of the TLR4 pathway on MSCs led to secretion of low levels of prothrombin by MSCs, while disrupting the TLR4 pathway impaired canonical signaling through PAR1 in response to thrombin. Therefore, this study demonstrates that both TLR4 and PAR1 are required for MSC survival under inflammatory conditions in vitro and therapeutic capacity in vivo, and that the TLR4 pathway regulates signaling through PAR1 on MSCs. Stem Cells 2018;36:796–806


Stem Cells | 2018

The TLR4‐PAR1 Axis Regulates Bone Marrow Mesenchymal Stromal Cell Survival and Therapeutic Capacity in Experimental Bacterial Pneumonia

Naveen Gupta; Ranjeet K. Sinha; Anna Krasnodembskaya; Xiao Xu; Victor Nizet; Michael A. Matthay

Bone marrow derived mesenchymal stromal cells have been shown to have significant therapeutic effects in experimental models of pneumonia and lung injury. The current study examined the roles of the toll like receptor 4 (TLR4) and protease activated receptor 1 (PAR1) pathways on mesenchymal stromal cell (MSC) survival and therapeutic activity in a murine model of pneumonia. MSCs from TLR4 ‐/‐ and R41Q‐PAR1 mutated mice were isolated to test the effect of mutating these specific pathways on MSC survival when exposed to cytotoxic stimuli in vitro. An Escherichia coli pneumonia model was used to assess the effect of these specific pathways on MSC therapeutic activity in vivo. Our results showed that mutation of either the TLR4 or PAR1 pathways in MSCs impaired cell survival under conditions of inflammatory stress in vitro, and eliminated their therapeutic efficacy in vivo. Also, stimulation of the TLR4 pathway on MSCs led to secretion of low levels of prothrombin by MSCs, while disrupting the TLR4 pathway impaired canonical signaling through PAR1 in response to thrombin. Therefore, this study demonstrates that both TLR4 and PAR1 are required for MSC survival under inflammatory conditions in vitro and therapeutic capacity in vivo, and that the TLR4 pathway regulates signaling through PAR1 on MSCs. Stem Cells 2018;36:796–806


Journal of Antimicrobial Chemotherapy | 2018

SCH79797 improves outcomes in experimental bacterial pneumonia by boosting neutrophil killing and direct antibiotic activity

Naveen Gupta; Roland Liu; Stephanie Shin; Ranjeet K. Sinha; Joe Pogliano; Kit Pogliano; Victor Nizet; Ross Corriden

Objectives The role of protease-activated receptor 1 (PAR1) in the pathogenesis of pneumonia and sepsis is ambiguous given the existing literature. As PAR1 is classically activated by the coagulation-based protease thrombin and leads to vascular leakage, our hypothesis was that PAR1 blockade with SCH79797 would be therapeutically beneficial in an experimental model of murine Gram-negative pneumonia. Methods In this study, we administered SCH79797 via the intrapulmonary route 6 h after the establishment of Escherichia coli pneumonia and observed a significant improvement in survival, lung injury, bacterial clearance and inflammation. We focused on neutrophils as a potential target of the PAR1 antagonist, since they are the predominant inflammatory cell type in the infected lung. Results Neutrophils appear to express PAR1 at low levels and the PAR1 antagonist SCH79797 enhanced neutrophil killing. Part of this effect may be explained by alterations in the generation of reactive oxygen species (ROS). SCH79797 also led to robust neutrophil extracellular trap (NET) generation and cathelicidin-related antimicrobial peptide (CRAMP) release by neutrophils. Surprisingly, SCH79797 also had a potent, direct antibiotic effect with disruption of the E. coli cell membrane. However, the newer-generation PAR1 antagonist, vorapaxar (SCH530348), had no appreciable effect on neutrophil activity or direct bacterial killing, which suggests the effects seen with SCH79797 may be PAR1 independent. Conclusions In summary, we observed that intrapulmonary treatment with SCH79797 has significant therapeutic effects in a model of E. coli pneumonia that appear to be due, in part, to both neutrophil-stimulating and direct antibacterial effects of SCH79797.


Blood Advances | 2017

Activated protein C light chain provides an extended binding surface for its anticoagulant cofactor, protein S

José A. Fernández; Xiao Xu; Ranjeet K. Sinha; Laurent O. Mosnier; Michel F. Sanner

Protein S anticoagulant cofactor sensitivity and PAR1 cleavage activity were assayed for 9 recombinant APC mutants.Residues L38, K43, I73, F95, and W115 on one face of the APC light chain define an extended surface containing the protein S binding site.

Collaboration


Dive into the Ranjeet K. Sinha's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Xiao Xu

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Laurent Burnier

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Naveen Gupta

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Greg Martin

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Hiroshi Deguchi

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Sergey Kupriyanov

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Victor Nizet

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge