Network


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

Hotspot


Dive into the research topics where Martin Hegen is active.

Publication


Featured researches published by Martin Hegen.


Journal of Immunology | 2006

MAPKAP Kinase 2-Deficient Mice Are Resistant to Collagen-Induced Arthritis

Martin Hegen; Matthias Gaestel; Cheryl Nickerson-Nutter; Lih-Ling Lin; Jean-Baptiste Telliez

TNF-α is a pleiotropic cytokine considered a primary mediator of immune regulation and inflammatory response and has been shown to play a central role in rheumatoid arthritis (RA). MAPKAP kinase 2 (MK2) is a serine/threonine kinase that is regulated through direct phosphorylation by p38 MAPK, and has been shown to be an essential component in the inflammatory response that regulates the biosynthesis of TNF-α at a posttranscriptional level. The murine model of collagen-induced arthritis (CIA) is an established disease model to study pathogenic mechanisms relevant to RA. In this study, we report that deletion of the MK2 gene in DBA/1LacJ mice confers protection against CIA. Interestingly, the MK2 heterozygous mutants display an intermediate level of protection when compared with homozygous mutant and wild-type littermates. We show that MK2−/− and MK2+/− mice exhibit decreased disease incidence and severity in the CIA disease model and reduced TNF-α and IL-6 serum levels following LPS/d-Gal treatment compared with wild-type mice. Additionally, we show that levels of IL-6 mRNA in paws of mice with CIA correlate with the disease status. These findings suggest that an MK2 inhibitor could be of great therapeutic value to treat inflammatory diseases like RA.


Journal of Experimental Medicine | 2003

Cytosolic phospholipase A2α-deficient mice are resistant to collagen-induced arthritis

Martin Hegen; Linhong Sun; Naonori Uozumi; Kazuhiko Kume; Mary E. Goad; Cheryl Nickerson-Nutter; Takao Shimizu; James D. Clark

Pathogenic mechanisms relevant to rheumatoid arthritis occur in the mouse model of collagen-induced arthritis (CIA). Cytosolic phospholipase A2α (cPLA2α) releases arachidonic acid from cell membranes to initiate the production of prostaglandins and leukotrienes. These inflammatory mediators have been implicated in the development of CIA. To test the hypothesis that cPLA2α plays a key role in the development of CIA, we backcrossed cPLA2α-deficient mice on the DBA/1LacJ background that is susceptible to CIA. The disease severity scores and the incidence of disease were markedly reduced in cPLA2α-deficient mice compared with wild-type littermates. At completion of the study, >90% of the wild-type mice had developed disease whereas none of the cPLA2α-deficient mice had more than one digit inflamed. Furthermore, visual disease scores correlated with severity of disease determined histologically. Pannus formation, articular fibrillation, and ankylosis were all dramatically reduced in the cPLA2α-deficient mice. Although the disease scores differed significantly between cPLA2α mutant and wild-type mice, anti-collagen antibody levels were similar in the wild-type mice and mutant littermates. These data demonstrate the critical role of cPLA2α in the pathogenesis of CIA.


Immunology | 1997

Cross‐linking of CD26 by antibody induces tyrosine phosphorylation and activation of mitogen‐activated protein kinase

Martin Hegen; J Kameoka; Rui-Ping Dong; Stuart F. Schlossman; Chikao Morimoto

CD26, a T‐cell activation antigen that has dipeptidyl peptidase IV activity in its extracellular domain and has also been shown to play an important role in T‐cell activation. The earliest biochemical events seen in stimulated T lymphocytes activated through the engagement of the T‐cell receptor (TCR) is the tyrosine phosphorylation of a panel of cellular proteins. In this study we demonstrate that antibody‐induced cross‐linking of CD26 in CD26‐transfected Jurkat cells induced tyrosine phosphorylation of several intracellular proteins with a similar pattern to that seen after TCR/CD3 stimulation. Herbimycin A, an inhibitor of the src family protein tyrosine kinases dramatically inhibited this CD26‐mediated effect on tyrosine phosphorylation. Major tyrosine phosphorylated proteins were identified by immunoblotting, and included p56lck, p59fyn, zeta associated protein‐tyrosine kinase of 70 000 MW (ZAP‐70), mitogen‐activated protein (MAP) kinase, c‐Cbl, and phospholipase Cγ. CD26‐induced tyrosine phosphorylation of MAP kinase correlated with increased MAP kinase activity. In addition, CD26 was costimulatory to CD3 signal transduction since co‐cross‐linking of CD26 and CD3 antigens induced prolonged and increased tyrosine phosphorylation in comparison with CD3 activation alone. We therefore conclude that CD26 is a true costimulatory entity that can up‐regulate the signal transducing properties of the TCR.


Journal of Medicinal Chemistry | 2008

Indole Cytosolic Phospholipase A2 α Inhibitors: Discovery and in Vitro and in Vivo Characterization of 4-{3-[5-Chloro-2-(2-{[(3,4-dichlorobenzyl)sulfonyl]amino}ethyl)-1-(diphenylmethyl)-1H-indol-3-yl]propyl}benzoic Acid, Efipladib

John C. McKew; Katherine L. Lee; Marina W.H. Shen; Paresh Thakker; Megan A. Foley; Mark L. Behnke; Baihua Hu; Fuk-Wah Sum; Steve Tam; Yonghan Hu; Lihren Chen; Steven J. Kirincich; Ronald S. Michalak; Jennifer R. Thomason; Manus Ipek; Kun Wu; Lane Wooder; Manjunath K. Ramarao; Elizabeth Murphy; Debra G. Goodwin; Leo M. Albert; Xin Xu; Frances Donahue; M. Sherry Ku; James C. Keith; Cheryl Nickerson-Nutter; William M. Abraham; Cara Williams; Martin Hegen; James D. Clark

The optimization of a class of indole cPLA 2 alpha inhibitors is described herein. The importance of the substituent at C3 and the substitution pattern of the phenylmethane sulfonamide region are highlighted. Optimization of these regions led to the discovery of 111 (efipladib) and 121 (WAY-196025), which are shown to be potent, selective inhibitors of cPLA 2 alpha in a variety of isolated enzyme assays, cell based assays, and rat and human whole blood assays. The binding of these compounds has been further examined using isothermal titration calorimetry. Finally, these compounds have shown efficacy when dosed orally in multiple acute and chronic prostaglandin and leukotriene dependent in vivo models.


Annals of the Rheumatic Diseases | 2011

Animal models for arthritis: innovative tools for prevention and treatment

George Kollias; P. Papadaki; Florence Apparailly; Margriet J. Vervoordeldonk; Rikard Holmdahl; V. Baumans; C. Desaintes; J. Di Santo; J. Distler; Paul Garside; Martin Hegen; T. W. J. Huizinga; Astrid Jüngel; L Klareskog; Iain B. McInnes; I. Ragoussis; Georg Schett; B.t. Hart; P P Tak; René E. M. Toes; W.B. van den Berg; W. Wurst

The development of novel treatments for rheumatoid arthritis (RA) requires the interplay between clinical observations and studies in animal models. Given the complex molecular pathogenesis and highly heterogeneous clinical picture of RA, there is an urgent need to dissect its multifactorial nature and to propose new strategies for preventive, early and curative treatments. Research on animal models has generated new knowledge on RA pathophysiology and aetiology and has provided highly successful paradigms for innovative drug development. Recent focus has shifted towards the discovery of novel biomarkers, with emphasis on presymptomatic and emerging stages of human RA, and towards addressing the pathophysiological mechanisms and subsequent efficacy of interventions that underlie different disease variants. Shifts in the current paradigms underlying RA pathogenesis have also led to increased demand for new (including humanised) animal models. There is therefore an urgent need to integrate the knowledge on human and animal models with the ultimate goal of creating a comprehensive ‘pathogenesis map’ that will guide alignment of existing and new animal models to the subset of disease they mimic. This requires full and standardised characterisation of all models at the genotypic, phenotypic and biomarker level, exploiting recent technological developments in ‘omics’ profiling and computational biology as well as state of the art bioimaging. Efficient integration and dissemination of information and resources as well as outreach to the public will be necessary to manage the plethora of data accumulated and to increase community awareness and support for innovative animal model research in rheumatology.


Arthritis & Rheumatism | 2010

Selective functional inhibition of JAK‐3 is sufficient for efficacy in collagen‐induced arthritis in mice

Tsung H. Lin; Martin Hegen; Elizabeth Quadros; Cheryl Nickerson-Nutter; Kenneth C. Appell; Andrew G. Cole; Yuefei Shao; Steve Tam; Michael Ohlmeyer; Bojing Wang; Debra G. Goodwin; Earl F. Kimble; Jorge Quintero; Min Gao; Peter Symanowicz; Christopher Wrocklage; Jennifer Lussier; Scott H. Schelling; Dejun Xuan; Rustem Krykbaev; Jenny Togias; Xin Xu; Richard Harrison; Tarek S. Mansour; Mary Collins; James D. Clark; Maria L. Webb; Katherine J. Seidl

OBJECTIVE All gamma-chain cytokines signal through JAK-3 and JAK-1 acting in tandem. We undertook this study to determine whether the JAK-3 selective inhibitor WYE-151650 would be sufficient to disrupt cytokine signaling and to ameliorate autoimmune disease pathology without inhibiting other pathways mediated by JAK-1, JAK-2, and Tyk-2. METHODS JAK-3 kinase selective compounds were characterized by kinase assay and JAK-3-dependent (interleukin-2 [IL-2]) and -independent (IL-6, granulocyte-macrophage colony-stimulating factor [GM-CSF]) cell-based assays measuring proliferation or STAT phosphorylation. In vivo, off-target signaling was measured by IL-22- and erythropoietin (EPO)-mediated models, while on-target signaling was measured by IL-2-mediated signaling. Efficacy of JAK-3 inhibitors was determined using delayed-type hypersensitivity (DTH) and collagen-induced arthritis (CIA) models in mice. RESULTS In vitro, WYE-151650 potently suppressed IL-2-induced STAT-5 phosphorylation and cell proliferation, while exhibiting 10-29-fold less activity against JAK-3-independent IL-6- or GM-CSF-induced STAT phosphorylation. Ex vivo, WYE-151650 suppressed IL-2-induced STAT phosphorylation, but not IL-6-induced STAT phosphorylation, as measured in whole blood. In vivo, WYE-151650 inhibited JAK-3-mediated IL-2-induced interferon-gamma production and decreased the natural killer cell population in mice, while not affecting IL-22-induced serum amyloid A production or EPO-induced reticulocytosis. WYE-151650 was efficacious in mouse DTH and CIA models. CONCLUSION In vitro, ex vivo, and in vivo assays demonstrate that WYE-151650 is efficacious in mouse CIA despite JAK-3 selectivity. These data question the need to broadly inhibit JAK-1-, JAK-2-, or Tyk-2-dependent cytokine pathways for efficacy.


Arthritis & Rheumatism | 2012

JAK inhibition with tofacitinib suppresses arthritic joint structural damage through decreased RANKL production

Timothy P. LaBranche; Michael I. Jesson; Zaher A. Radi; Chad E. Storer; Julia A. Guzova; Sheri L. Bonar; Janice M. Thompson; Fernando A. Happa; Zachary S. Stewart; Yutian Zhan; Chris S. Bollinger; Prashant N. Bansal; Jeremy W. Wellen; Dean Wilkie; Steven A. Bailey; Peter Symanowicz; Martin Hegen; Richard D. Head; Nandini Kishore; Gabriel Mbalaviele; Debra M. Meyer

OBJECTIVE The mechanistic link between Janus kinase (JAK) signaling and structural damage to arthritic joints in rheumatoid arthritis (RA) is poorly understood. This study was undertaken to investigate how selective inhibition of JAK with tofacitinib (CP-690,550) affects osteoclast-mediated bone resorption in a rat adjuvant-induced arthritis (AIA) model, as well as human T lymphocyte RANKL production and human osteoclast differentiation and function. METHODS Hind paw edema, inflammatory cell infiltration, and osteoclast-mediated bone resorption in rat AIA were assessed using plethysmography, histopathologic analysis, and immunohistochemistry; plasma and hind paw tissue levels of cytokines and chemokines (including RANKL) were also assessed. In vitro RANKL production by activated human T lymphocytes was evaluated by immunoassay, while human osteoclast differentiation and function were assessed via quantitative tartrate-resistant acid phosphatase staining and degradation of human bone collagen, respectively. RESULTS Edema, inflammation, and osteoclast-mediated bone resorption in rats with AIA were dramatically reduced after 7 days of treatment with the JAK inhibitor, which correlated with reduced numbers of CD68/ED-1+, CD3+, and RANKL+ cells in the paws; interleukin-6 (transcript and protein) levels were rapidly reduced in paw tissue within 4 hours of the first dose, whereas it took 4-7 days of therapy for RANKL levels to decrease. Tofacitinib did not impact human osteoclast differentiation or function, but did decrease human T lymphocyte RANKL production in a concentration-dependent manner. CONCLUSION These results suggest that the JAK inhibitor tofacitinib suppresses osteoclast-mediated structural damage to arthritic joints, and this effect is secondary to decreased RANKL production.


Bioorganic & Medicinal Chemistry Letters | 2011

Imidazo(1,5-a)quinoxalines as irreversible BTK inhibitors for the treatment of rheumatoid arthritis

Kyung-Hee Kim; Andreas Maderna; Mark E. Schnute; Martin Hegen; Shashi Mohan; Joy S. Miyashiro; Laura Lin; Evelyn Li; Sean Keegan; Jennifer Lussier; Christopher Wrocklage; Cheryl Nickerson-Nutter; Arthur J. Wittwer; Holly Soutter; Nicole Caspers; Seungil Han; Ravi G. Kurumbail; Kyri Dunussi-Joannopoulos; John Douhan; Allan Wissner

Imidazo[1,5-a]quinoxalines were synthesized that function as irreversible Brutons tyrosine kinase (BTK) inhibitors. The syntheses and SAR of this series of compounds are presented as well as the X-ray crystal structure of the lead compound 36 in complex with a gate-keeper variant of ITK enzyme. The lead compound showed good in vivo efficacy in preclinical RA models.


Molecular Immunology | 1998

CORRELATION OF THE EPITOPES DEFINED BY ANTI-CD26 MABS AND CD26 FUNCTION

Rui-Ping Dong; Kouichi Tachibana; Martin Hegen; S. Scharpé; Donny Cho; Stuart F. Schlossman; Chikao Morimoto

To clarify the different anti-CD26 mAbs corresponding different functions of CD26, the correlation of the epitopes defined by anti-CD26 mAbs and the functions of CD26 have been studied. Using truncated, human-rat CD26 swap mutants and cross-blocking studies, 13 anti-CD26 mAbs were divided into 5 separate groups. These 5 epitopes were localized between the 1-247th, 248-358th, 359-449th (closer to the 359th amino acid), 450-577th and 359 653th amino acid regions. MAbs against two of these five epitopes, the 248-358th and 359-449th amino acid regions, were associated with inducing modulation of CD26 and T-cell costimulation through the CD3 pathway. Furthermore, mAbs against one of these epitopes, the 359-449th amino acid region, appeared to encompass the ADA binding domain. Analysing the avidity of each mAb to the CD26 molecule using DPPIV enzymatic activity as an indicator, we found that the function of CD26 had little correlation with the avidity of anti-CD26 mAbs, suggesting that distinct epitopes defined by anti-CD26 mAbs appeared to be associated with different functions of CD26. These results will be very useful in the further definition of the functional domains of CD26.


Bioconjugate Chemistry | 2012

Pharmacokinetic, biodistribution, and biophysical profiles of TNF nanobodies conjugated to linear or branched poly(ethylene glycol).

Yulia Vugmeyster; Clifford Entrican; Alison Joyce; Rosemary Lawrence-Henderson; Beth Leary; Christopher S. Mahoney; Himakshi K. Patel; Stephen W. Raso; Stephane Olland; Martin Hegen; Xin Xu

Covalent attachment of poly(ethylene glycol) (PEG) to therapeutic proteins has been used to prolong in vivo exposure of therapeutic proteins. We have examined pharmacokinetic, biodistribution, and biophysical profiles of three different tumor necrosis factor alpha (TNF) Nanobody-40 kDa PEG conjugates: linear 1 × 40 KDa, branched 2 × 20 kDa, and 4 × 10 kDa conjugates. In accord with earlier reports, the superior PK profile was observed for the branched versus linear PEG conjugates, while all three conjugates had similar potency in a cell-based assay. Our results also indicate that (i) a superior PK profile of branched versus linear PEGs is likely to hold across species, (ii) for a given PEG size, the extent of PEG branching affects the PK profile, and (iii) tissue penetration may differ between linear and branched PEG conjugates in a tissue-specific manner. Biophysical analysis (R(g)/R(h) ratio) demonstrated that among the three protein-PEG conjugates the linear PEG conjugate had the most extended time-average conformation and the most exposed surface charges. We hypothesized that these biophysical characteristics of the linear PEG conjugate accounts for relatively less optimal masking of sites involved in elimination of the PEGylated Nanobodies (e.g., intracellular uptake and proteolysis), leading to lower in vivo exposure compared to the branched PEG conjugates. However, additional studies are needed to test this hypothesis.

Collaboration


Dive into the Martin Hegen's collaboration.

Top Co-Authors

Avatar

René E. M. Toes

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Xin Xu

University of Minnesota

View shared research outputs
Top Co-Authors

Avatar

Mary Collins

University College London

View shared research outputs
Researchain Logo
Decentralizing Knowledge