Network


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

Hotspot


Dive into the research topics where Markus Huber-Lang is active.

Publication


Featured researches published by Markus Huber-Lang.


Nature Protocols | 2009

Immunodesign of experimental sepsis by cecal ligation and puncture

Daniel Rittirsch; Markus Huber-Lang; Michael A. Flierl; Peter A. Ward

Sepsis remains a prevalent clinical challenge and the underlying pathophysiology is still poorly understood. To investigate the complex molecular mechanisms of sepsis, various animal models have been developed, the most frequently used being the cecal ligation and puncture (CLP) model in rodents. In this model, sepsis originates from a polymicrobial infectious focus within the abdominal cavity, followed by bacterial translocation into the blood compartment, which then triggers a systemic inflammatory response. A requirement of this model is that it is performed with high consistency to obtain reproducible results. Evidence is now emerging that the accompanying inflammatory response varies with the severity grade of sepsis, which is highly dependent on the extent of cecal ligation. In this protocol, we define standardized procedures for inducing sepsis in mice and rats by applying defined severity grades of sepsis through modulation of the position of cecal ligation. The CLP procedure can be performed in as little as 10 min for each animal by an experienced user, with additional time required for subsequent postoperative care and data collection.


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.


Journal of Immunology | 2010

Molecular Intercommunication between the Complement and Coagulation Systems

Umme Amara; Michael A. Flierl; Daniel Rittirsch; Andreas Klos; Hui Chen; Barbara Acker; Uwe B. Brückner; Bo Nilsson; Florian Gebhard; John D. Lambris; Markus Huber-Lang

The complement system as well as the coagulation system has fundamental clinical implications in the context of life-threatening tissue injury and inflammation. Associations between both cascades have been proposed, but the precise molecular mechanisms remain unknown. The current study reports multiple links for various factors of the coagulation and fibrinolysis cascades with the central complement components C3 and C5 in vitro and ex vivo. Thrombin, human coagulation factors (F) XIa, Xa, and IXa, and plasmin were all found to effectively cleave C3 and C5. Mass spectrometric analyses identified the cleavage products as C3a and C5a, displaying identical molecular weights as the native anaphylatoxins C3a and C5a. Cleavage products also exhibited robust chemoattraction of human mast cells and neutrophils, respectively. Enzymatic activity for C3 cleavage by the investigated clotting and fibrinolysis factors is defined in the following order: FXa > plasmin > thrombin > FIXa > FXIa > control. Furthermore, FXa-induced cleavage of C3 was significantly suppressed in the presence of the selective FXa inhibitors fondaparinux and enoxaparin in a concentration-dependent manner. Addition of FXa to human serum or plasma activated complement ex vivo, represented by the generation of C3a, C5a, and the terminal complement complex, and decreased complement hemolytic serum activity that defines exact serum concentration that results in complement-mediated lysis of 50% of sensitized sheep erythrocytes. Furthermore, in plasma from patients with multiple injuries (n = 12), a very early appearance and correlation of coagulation (thrombin–antithrombin complexes) and the complement activation product C5a was found. The present data suggest that coagulation/fibrinolysis proteases may act as natural C3 and C5 convertases, generating biologically active anaphylatoxins, linking both cascades via multiple direct interactions in terms of a complex serine protease system.


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.


Advances in Experimental Medicine and Biology | 2008

Interaction Between the Coagulation and Complement System

Umme Amara; Daniel Rittirsch; Michael A. Flierl; Uwe B. Brückner; Andreas Klos; Florian Gebhard; John D. Lambris; Markus Huber-Lang

The complement system as a main column of innate immunity and the coagulation system as a main column in hemostasis undergo massive activation early after injury. Interactions between the two cascades have often been proposed but the precise molecular pathways of this interplay are still in the dark. To elucidate the mechanisms involved, the effects of various coagulation factors on complement activation and generation of anaphylatoxins were investigated and summarized in the light of the latest literature. Own in vitro findings suggest, that the coagulation factors FXa, FXIa and plasmin may cleave both C5 and C3, and robustly generate C5a and C3a (as detected by immunoblotting and ELISA). The produced anaphylatoxins were found to be biologically active as shown by a dose-dependent chemotactic response of neutrophils and HMC-1 cells, respectively. Thrombin did not only cleave C5 (Huber-Lang et al. 2006) but also in vitro-generated C3a when incubated with native C3. The plasmin-induced cleavage activity could be dose-dependently blocked by the serine protease inhibitor aprotinin and leupeptine. These findings suggest that various serine proteases belonging to the coagulation system are able to activate the complement cascade independently of the established pathways. Moreover, functional C5a and C3a are generated, both of which are known to be crucially involved in the inflammatory response.


Journal of Immunology | 2001

Role of C5a in Multiorgan Failure During Sepsis

Markus Huber-Lang; Vidya Sarma; Kristina T. Lu; Stephanie R. McGuire; Vaishalee A. Padgaonkar; Renfeng Guo; Ellen M. Younkin; Robin G. Kunkel; Jiabing Ding; Richard W. Erickson; John T. Curnutte; Peter A. Ward

In humans with sepsis, the onset of multiorgan failure (MOF), especially involving liver, lungs, and kidneys, is a well known complication that is associated with a high mortality rate. Our previous studies with the cecal ligation/puncture (CLP) model of sepsis in rats have revealed a C5a-induced defect in the respiratory burst of neutrophils. In the current CLP studies, MOF occurred during the first 48 h with development of liver dysfunction and pulmonary dysfunction (falling arterial partial pressure of O2, rising partial pressure of CO2). In this model an early respiratory alkalosis developed, followed by a metabolic acidosis with increased levels of blood lactate. During these events, blood neutrophils lost their chemotactic responsiveness both to C5a and to the bacterial chemotaxin, fMLP. Neutrophil dysfunction was associated with virtually complete loss in binding of C5a, but binding of fMLP remained normal. If CLP animals were treated with anti-C5a, indicators of MOF and lactate acidosis were greatly attenuated. Under the same conditions, C5a binding to blood neutrophils remained intact; in tandem, in vitro chemotactic responses to C5a and fMLP were retained. These data suggest that, in the CLP model of sepsis, treatment with anti-C5a prevents development of MOF and the accompanying onset of blood neutrophil dysfunction. This may explain the protective effects of anti-C5a in the CLP model of sepsis.


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)

Collaboration


Dive into the Markus Huber-Lang's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Peter A. Ward

University of Louisville

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michael A. Flierl

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge