Network


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

Hotspot


Dive into the research topics where J. Vidya Sarma is active.

Publication


Featured researches published by J. Vidya Sarma.


Nature Medicine | 2006

Generation of C5a in the absence of C3: a new complement activation pathway.

Markus Huber-Lang; J. Vidya Sarma; Firas S. Zetoune; Daniel Rittirsch; Thomas A. Neff; Stephanie R. McGuire; John D. Lambris; Roscoe L. Warner; Michael A. Flierl; Laszlo M. Hoesel; Florian Gebhard; John G. Younger; Scott M. Drouin; Rick A. Wetsel; Peter A. Ward

Complement-mediated tissue injury in humans occurs upon deposition of immune complexes, such as in autoimmune diseases and acute respiratory distress syndrome. Acute lung inflammatory injury in wild-type and C3−/− mice after deposition of IgG immune complexes was of equivalent intensity and was C5a dependent, but injury was greatly attenuated in Hc−/− mice (Hc encodes C5). Injury in lungs of C3−/− mice and C5a levels in bronchoalveolar lavage (BAL) fluids from these mice were greatly reduced in the presence of antithrombin III (ATIII) or hirudin but were not reduced in similarly treated C3+/+ mice. Plasma from C3−/− mice contained threefold higher levels of thrombin activity compared to plasma from C3+/+ mice. There were higher levels of F2 mRNA (encoding prothrombin) as well as prothrombin and thrombin protein in liver of C3−/− mice compared to C3+/+ mice. A potent solid-phase C5 convertase was generated using plasma from either C3+/+ or C3−/− mice. Human C5 incubated with thrombin generated C5a that was biologically active. These data suggest that, in the genetic absence of C3, thrombin substitutes for the C3-dependent C5 convertase. This linkage between the complement and coagulation pathways may represent a new pathway of complement activation.


Cell and Tissue Research | 2011

The Complement System

J. Vidya Sarma; Peter A. Ward

The complement system consists of a tightly regulated network of proteins that play an important role in host defense and inflammation. Complement activation results in opsonization of pathogens and their removal by phagocytes, as well as cell lysis. Inappropriate complement activation and complement deficiencies are the underlying cause of the pathophysiology of many diseases such as systemic lupus erythematosus and asthma. This review represents an overview of the complement system in an effort to understand the beneficial as well as harmful roles it plays during inflammatory responses.


Nature | 2007

Phagocyte-derived catecholamines enhance acute inflammatory injury.

Michael A. Flierl; Daniel Rittirsch; Brian A. Nadeau; Anthony J. Chen; J. Vidya Sarma; Firas S. Zetoune; Stephanie R. McGuire; Rachel P. List; Danielle E. Day; L. Marco Hoesel; Hongwei Gao; Nico van Rooijen; Markus Huber-Lang; Richard R. Neubig; Peter A. Ward

It is becoming increasingly clear that the autonomic nervous system and the immune system demonstrate cross-talk during inflammation by means of sympathetic and parasympathetic pathways. We investigated whether phagocytes are capable of de novo production of catecholamines, suggesting an autocrine/paracrine self-regulatory mechanism by catecholamines during inflammation, as has been described for lymphocytes. Here we show that exposure of phagocytes to lipopolysaccharide led to a release of catecholamines and an induction of catecholamine-generating and degrading enzymes, indicating the presence of the complete intracellular machinery for the generation, release and inactivation of catecholamines. To assess the importance of these findings in vivo, we chose two models of acute lung injury. Blockade of α2-adrenoreceptors or catecholamine-generating enzymes greatly suppressed lung inflammation, whereas the opposite was the case either for an α2-adrenoreceptor agonist or for inhibition of catecholamine-degrading enzymes. We were able to exclude T cells or sympathetic nerve endings as sources of the injury-modulating catecholamines. Our studies identify phagocytes as a new source of catecholamines, which enhance the inflammatory response.


Journal of Immunology | 2003

Protective Effects of IL-6 Blockade in Sepsis Are Linked to Reduced C5a Receptor Expression

Niels C. Riedemann; Thomas A. Neff; Ren Feng Guo; Kurt D. Bernacki; Ines J. Laudes; J. Vidya Sarma; John D. Lambris; Peter A. Ward

IL-6 is known to be an important pro- and anti-inflammatory cytokine, which is up-regulated during sepsis. Our previous work has suggested a role for IL-6 in the up-regulation of C5aR in sepsis. We reported earlier that interception of C5a or C5aR results in improved outcomes in experimental sepsis. Using the cecal ligation/puncture (CLP) model in mice, we now demonstrate that treatment with anti-IL-6 Ab (anti-IL-6) results in significantly improved survival, dependent on the amount of Ab infused. CLP animals showed significantly increased binding of 125I-labeled anti-C5aR to organs when compared to either control mice at 0 h or CLP animals infused with normal rabbit 125I-labeled IgG. Binding of 125I-labeled anti-C5aR to lung, liver, kidney, and heart was significantly decreased in anti-IL-6-treated animals 6 h after CLP. RT-PCR experiments with mRNA isolated from various organs obtained 3, 6, and 12 h after CLP demonstrated increased C5aR mRNA expression during the onset of sepsis, which was greatly suppressed in CLP mice treated with anti-IL-6. These data suggest that IL-6 plays an important role in the increased expression of C5aR in lung, liver, kidney, and heart during the development of sepsis in mice and that interception of IL-6 leads to reduced expression of C5aR and improved survival.


American Journal of Pathology | 2002

Generation of C5a by phagocytic cells

Markus Huber-Lang; Ellen M. Younkin; J. Vidya Sarma; Niels C. Riedemann; Stephanie R. McGuire; Kristina T. Lu; Robin G. Kunkel; John G. Younger; Firas S. Zetoune; Peter A. Ward

The complement activation product, C5a, is a powerful phlogistic factor. Using antibodies to detect human or rat C5a, incubation at pH 7.4 of human blood neutrophils or rat alveolar macrophages (AMs) with C5 in the presence of phorbol 12-myristate 13-acetate (PMA) led to generation of C5a. Rat AMs activated with lipopolysaccharide also generated C5a from C5. With activated neutrophils, extensive cleavage of C5 occurred, whereas activated macrophages had much more selective proteolytic activity for C5. Peripheral blood human or rat mononuclear cells and rat alveolar epithelial cells when stimulated with phorbol ester all failed to demonstrate an ability to cleave C5, suggesting a specificity of C5 cleavage by phagocytic cells. With rat AMs, C5a generation was time-dependent and was blocked if AMs were pretreated with inhibitors of transcription or protein synthesis (actinomycin D or cycloheximide). Similar treatment of activated human polymorphonuclear leukocytes only partially reduced C5a generation after addition of C5. C5a generated by activated AMs was biologically (chemotactically) active. This generation was sensitive to serine protease inhibitors but not to other classes of inhibitors. These data indicate that phagocytic cells, especially lung macrophages, can generate C5a from C5. In the context of the lung, this may represent an important C5a-generating pathway that is independent of the plasma complement system.


Journal of Immunology | 2002

Complement-Induced Impairment of Innate Immunity During Sepsis

Markus Huber-Lang; Ellen M. Younkin; J. Vidya Sarma; Stephanie R. McGuire; Kristina T. Lu; Ren Feng Guo; Vaishalee A. Padgaonkar; John T. Curnutte; Richard W. Erickson; Peter A. Ward

This study defines the molecular basis for defects in innate immunity involving neutrophils during cecal ligation/puncture (CLP)-induced sepsis in rats. Blood neutrophils from CLP rats demonstrated defective phagocytosis and defective assembly of NADPH oxidase, the latter being due to the inability of p47phox to translocate from the cytosol to the cell membrane of neutrophils after cell stimulation by phorbol ester (PMA). The appearance of these defects was prevented by in vivo blockade of C5a in CLP rats. In vitro exposure of neutrophils to C5a led to reduced surface expression of C5aR and defective assembly of NADPH oxidase, as defined by failure in phosphorylation of p47phox and its translocation to the cell membrane, together with failure in phosphorylation of p42/p44 mitogen-activated protein kinases. These data identify a molecular basis for defective innate immunity involving neutrophils during sepsis.


The FASEB Journal | 2008

Adverse functions of IL-17A in experimental sepsis

Michael A. Flierl; Daniel Rittirsch; Hongwei Gao; Laszlo M. Hoesel; Brian A. Nadeau; Danielle E. Day; Firas S. Zetoune; J. Vidya Sarma; Markus Huber-Lang; James L.M. Ferrara; Peter A. Ward

IL‐17A is a proinflammatory cytokine produced by a variety of cells. In the current study, we examined the role of IL‐17A in sepsis induced in mice by cecal ligation and puncture (CLP). IL‐17A levels, which rose time‐dependently in plasma after CLP, were not affected in the absence of αβ T cells or neutrophils. In sharp contrast, γδ T cell‐knockout or γδ T cell‐depleted mice displayed baseline IL‐17A plasma levels after CLP. Neutralization of IL‐17A by two different antibodies improved sepsis (survival from ~10% to nearly 60%). Unexpectedly, antibody treatment was protective, even when administration of anti‐IL‐17A was delayed for up to 12 h after CLP. These protective effects of IL‐17A blockade were associated with substantially reduced levels of bacteremia together with significant reductions of systemic proinflammatory cytokines and chemokines in plasma. In vitro incubation of mouse peritoneal macrophages with lipopolysaccharide (LPS) in the copresence of IL‐17A substantially increased the production of TNF‐α, IL‐1β, and IL‐6 by these cells. These data suggest that, during experimental sepsis, γδ T cell‐derived IL‐17A promotes high levels of proinflammatory mediators and bacteremia, resulting in enhanced lethality. IL‐17A may be a potential therapeutic target in sepsis.—Flierl, M. A., Rittirsch, D., Gao, H., Hoesel, L. M., Nadeau, B. A., Day, D. E., Zetoune, F. S., Sarma, J. V., Huber‐Lang, M. S., Ferrara, J. L. M., Ward, P. A. Adverse functions of IL‐17A in experimental sepsis. FASEB J. 22, 2198–2205 (2008)


The FASEB Journal | 2002

Protection of innate immunity by C5aR antagonist in septic mice

Markus Huber-Lang; Niels C. Riedeman; J. Vidya Sarma; Ellen M. Younkin; Stephanie R. McGuire; Ines J. Laudes; Kristina T. Lu; Ren Feng Guo; Thomas A. Neff; Vaishalee A. Padgaonkar; John D. Lambris; Lynn A. Spruce; Dimitrios Mastellos; Firas S. Zetoune; Peter A. Ward

Innate immune functions are known to be compromised during sepsis, often with lethal consequences. There is also evidence in rats that sepsis is associated with excessive complement activation and generation of the potent anaphylatoxin C5a. In the presence of a cyclic peptide antagonist (C5aRa) to the C5a receptor (C5aR), the binding of murine 125I‐C5a to murine neutrophils was reduced, the in vitro chemotactic responses of mouse neutrophils to mouse C5a were markedly diminished, the acquired defect in hydrogen peroxide (H2O2) production of C5a‐exposed neutrophils was reversed, and the lung permeability index (extravascular leakage of albumin) in mice after intrapulmonary deposition of IgG immune complexes was markedly diminished. Mice that developed sepsis after cecal ligation/puncture (CLP) and were treated with C5aRa had greatly improved survival rates. These data suggest that C5aRa interferes with neutrophil responses to C5a, preventing C5a‐induced compromise of innate immunity during sepsis, with greatly improved survival rates after CLP.—Huber‐Lang, M. S., Riedeman, N. C., Sarma, J. V., Younkin, E. M., McGuire, S. R., Laudes, I. J., Lu, K. T., Guo, R.‐F., Neff, T. A., Padgaonkar, V. A., Lambris, J. D., Spruce, L., Mastellos, D., Zetoune, F. S., Ward, P. A. Protection of innate immunity by C5aR antagonist in septic mice. FASEB J. 16, 1567–1574 (2002)


American Journal of Pathology | 2003

Regulatory Effects of iNOS on Acute Lung Inflammatory Responses in Mice

Cecilia L. Speyer; Thomas A. Neff; Roscoe L. Warner; Ren Feng Guo; J. Vidya Sarma; Niels C. Riedemann; Megan E. Murphy; Hedwig S. Murphy; Peter A. Ward

The role of endogenous NO in the regulation of acute lung injury is not well defined. We investigated the effects of inducible nitric oxide synthase (iNOS) and endothelial NOS (eNOS) on the acute inflammatory response in mouse lungs. Acute lung injury was induced by intratracheal instillation of bacterial lipopolysaccharide (LPS) into wild-type (WT) mice and mice deficient in iNOS (iNOS(-/-)) or eNOS (eNOS(-/-)). Endpoints of inflammatory injury were myeloperoxidase (MPO) content and leak of albumin into lung. Inflammatory injury was similar in WT and eNOS(-/-) mice but was substantially increased in iNOS(-/-) mice. Bronchoalveolar lavage (BAL) fluids of iNOS(-/-) and WT mice showed similar levels of CXC chemokines (MIP-2, KC) but enhanced levels of CC chemokines (MCP-1, MCP-3). Increased lung content of MPO in iNOS(-/-) mice was reduced by anti-MCP-1 to values found in WT mice. In vitro stimulation of microvascular endothelial cells with LPS and IFN gamma revealed elevated production of CXC and CC chemokines in cells from iNOS(-/-) mice when compared to endothelial cells from iNOS(+/+) mice. Peritoneal macrophages from iNOS(-/-) donors also revealed increased production of CC chemokines after stimulation with LPS and interferon (IFN gamma). These data indicate that absence of iNOS causes enhanced lung inflammatory responses in mice which may be related to enhanced production of MCP-1 by endothelial cells and macrophages. It appears that iNOS affects the lung inflammatory response by regulating chemokine production.


Journal of Immunology | 2002

Expression and Function of C5a Receptor in Mouse Microvascular Endothelial Cells

Ines J. Laudes; Jeffrey C. Chu; Markus Huber-Lang; Ren Feng Guo; Niels C. Riedemann; J. Vidya Sarma; Fakhri Mahdi; Hedwig S. Murphy; Cecilia L. Speyer; Kristina T. Lu; John D. Lambris; Firas S. Zetoune; Peter A. Ward

The complement-derived anaphylatoxin, C5a, is a potent phlogistic molecule that mediates its effects by binding to C5a receptor (C5aR; CD88). We now demonstrate specific binding of radiolabeled recombinant mouse C5a to mouse dermal microvascular endothelial cells (MDMEC) with a Kd50 of 3.6 nM and to ∼15,000–20,000 receptors/cell. Recombinant mC5a competed effectively with binding of [125I]rmC5a to MDMEC. Enhanced binding of C5a occurred, as well as increased mRNA for C5aR, after in vitro exposure of MDMEC to LPS, IFN-γ, or IL-6 in a time- and dose-dependent manner. By confocal microscopy, C5aR could be detected on surfaces of MDMEC using anti-C5aR Ab. In vitro expression of macrophage inflammatory protein-2 (MIP-2) and monocyte chemoattractant protein-1 (MCP-1) by MDMEC was also measured. Exposure of MDMEC to C5a or IL-6 did not result in changes in MIP-2 or MCP-1 production, but initial exposure of MDMEC to IL-6, followed by exposure to C5a, resulted in significantly enhanced production of MIP-2 and MCP-1 (but not TNF-α and MIP-1α). Although LPS or IFN-γ alone induced some release of MCP-1 and MIP-2, pre-exposure of these monolayers to LPS or IFN-γ, followed by addition of C5a, resulted in synergistic production of MIP-2 and MCP-1. Following i.v. infusion of LPS into mice, up-regulation of C5aR occurred in the capillary endothelium of mouse lung, as determined by immunostaining. These results support the hypothesis that C5aR expression on MDMEC and on the microvascular endothelium of lung can be up-regulated, suggesting that C5a in the co-presence of additional agonists may mediate pro-inflammatory effects of endothelial cells.

Collaboration


Dive into the J. Vidya Sarma's collaboration.

Top Co-Authors

Avatar

Peter A. Ward

Natural Environment Research Council

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Peter A. Ward

Natural Environment Research Council

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michael A. Flierl

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Hongwei Gao

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge