Network


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

Hotspot


Dive into the research topics where Henning Walczak is active.

Publication


Featured researches published by Henning Walczak.


Nature Medicine | 1999

Tumoricidal activity of tumor necrosis factor-related apoptosis-inducing ligand in vivo.

Henning Walczak; Robert E. Miller; Kiley Ariail; Brian Gliniak; Thomas S. Griffith; Marek Kubin; Wilson Chin; Jon Jones; Anne Woodward; Tiep Le; Craig A. Smith; Pam Smolak; Raymond G. Goodwin; Charles Rauch; JoAnn C. L. Schuh; David H. Lynch

To evaluate the utility of tumor necrosis factor–related apoptosis–inducing ligand (TRAIL) as a cancer therapeutic, we created leucine zipper (LZ) forms of human (hu) and murine (mu) TRAIL to promote and stabilize the formation of trimers. Both were biologically active, inducing apoptosis of both human and murine target cells in vitro with similar specific activities. In contrast to the fulminant hepatotoxicity of LZ–huCD95L in vivo, administration of either LZ–huTRAIL or LZ–muTRAIL did not seem toxic to normal tissues of mice. Finally, repeated treatments with LZ–huTRAIL actively suppressed growth of the TRAIL–sensitive human mammary adenocarcinoma cell line MDA–231 in CB.17 (SCID) mice, and histologic examination of tumors from SCID mice treated with LZ–huTRAIL demonstrated clear areas of apoptotic necrosis within 9–12 hours of injection.


The EMBO Journal | 1997

TRAIL‐R2: a novel apoptosis‐mediating receptor for TRAIL

Henning Walczak; Mariapia A. Degli-Esposti; Richard S. Johnson; Pam Smolak; Jennifer Y. Waugh; Norman Boiani; Martin S. Timour; Mary Gerhart; Kenneth A. Schooley; Craig A. Smith; Raymond G. Goodwin; Charles Rauch

TRAIL is a member of the tumor necrosis factor (TNF) family of cytokines and induces apoptosis in a wide variety of cells. Based on homology searching of a private database, a receptor for TRAIL (DR4 or TRAIL‐R1) was recently identified. Here we report the identification of a distinct receptor for TRAIL, TRAIL‐R2, by ligand‐based affinity purification and subsequent molecular cloning. TRAIL‐R2 was purified independently as the only receptor for TRAIL detectable on the surface of two different human cell lines that undergo apoptosis upon stimulation with TRAIL. TRAIL‐R2 contains two extracellular cysteine‐rich repeats, typical for TNF receptor (TNFR) family members, and a cytoplasmic death domain. TRAIL binds to recombinant cell‐surface‐expressed TRAIL‐R2, and TRAIL‐induced apoptosis is inhibited by a TRAIL‐R2–Fc fusion protein. TRAIL‐R2 mRNA is widely expressed and the gene encoding TRAIL‐R2 is located on human chromosome 8p22‐21. Like TRAIL‐R1, TRAIL‐R2 engages a caspase‐dependent apoptotic pathway but, in contrast to TRAIL‐R1, TRAIL‐R2 mediates apoptosis via the intracellular adaptor molecule FADD/MORT1. The existence of two distinct receptors for the same ligand suggests an unexpected complexity to TRAIL biology, reminiscent of dual receptors for TNF, the canonical member of this family.


Immunity | 2000

FADD/MORT1 and Caspase-8 Are Recruited to TRAIL Receptors 1 and 2 and Are Essential for Apoptosis Mediated by TRAIL Receptor 2

Martin R. Sprick; Markus A. Weigand; Eva Rieser; Charles Rauch; Peter Juo; John Blenis; Peter H. Krammer; Henning Walczak

Apoptosis induced by tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL/APO-2L) has been shown to exert important functions during various immunological processes. The involvement of the death adaptor proteins FADD/MORT1, TRADD, and RIP and the apoptosis-initiating caspases-8 and -10 in death signaling by the two death-inducing TRAIL receptors 1 and 2 (TRAIL-R1 and TRAIL-R2) are controversial. Analysis of the native TRAIL death-inducing signaling complex (DISC) revealed ligand-dependent recruitment of FADD/MORT1 and caspase-8. Differential precipitation of ligand-stimulated TRAIL receptors demonstrated that FADD/MORT1 and caspase-8 were recruited to TRAIL-R1 and TRAIL-R2 independently of each other. FADD/MORT1- and caspase-8-deficient Jurkat cells expressing only TRAIL-R2 were resistant to TRAIL-induced apoptosis. Thus, FADD/MORT1 and caspase-8 are essential for apoptosis induction via TRAIL-R2.


Journal of Clinical Investigation | 1997

Drug-induced apoptosis in hepatoma cells is mediated by the CD95 (APO-1/Fas) receptor/ligand system and involves activation of wild-type p53.

M Müller; Susanne Strand; Hubert Hug; Eva Maria Heinemann; Henning Walczak; Walter J. Hofmann; W Stremmel; Peter H. Krammer; Peter R. Galle

Chemotherapeutic drugs are cytotoxic by induction of apoptosis in drug-sensitive cells. We investigated the mechanism of bleomycin-induced cytotoxicity in hepatoma cells. At concentrations present in the sera of patients during therapy, bleomycin induced transient accumulation of nuclear wild-type (wt) p53 and upregulated expression of cell surface CD95 (APO-1/Fas) receptor in hepatoma cells carrying wt p53 (HepG2). Bleomycin did not increase CD95 in hepatoma cells with mutated p53 (Huh7) or in hepatoma cells which were p53-/- (Hep3B). In addition, sensitivity towards CD95-mediated apoptosis was also increased in wt p53 positive HepG2 cells. Microinjection of wt p53 cDNA into HepG2 cells had the same effect. In contrast, bleomycin did not enhance susceptibility towards CD95-mediated apoptosis in Huh7 and in Hep3B cells. Furthermore, bleomycin treatment of HepG2 cells increased CD95 ligand (CD95L) mRNA expression. Most notably, bleomycin-induced apoptosis in HepG2 cells was almost completely inhibited by antibodies which interfere with CD95 receptor/ligand interaction. These data suggest that apoptosis induced by bleomycin is mediated, at least in part, by p53-dependent stimulation of the CD95 receptor/ligand system. The same applies to other anti-cancer drugs such as cisplatin and methotrexate. These data may have major consequences for drug treatment of cancer and the explanation of drug sensitivity and resistance.


Nature Reviews Molecular Cell Biology | 2014

Regulated necrosis: the expanding network of non-apoptotic cell death pathways

Tom Vanden Berghe; Andreas Linkermann; Sandrine Jouan-Lanhouet; Henning Walczak; Peter Vandenabeele

Cell death research was revitalized by the understanding that necrosis can occur in a highly regulated and genetically controlled manner. Although RIPK1 (receptor-interacting protein kinase 1)- and RIPK3–MLKL (mixed lineage kinase domain-like)-mediated necroptosis is the most understood form of regulated necrosis, other examples of this process are emerging, including cell death mechanisms known as parthanatos, oxytosis, ferroptosis, NETosis, pyronecrosis and pyroptosis. Elucidating how these pathways of regulated necrosis are interconnected at the molecular level should enable this process to be therapeutically targeted.


Journal of Clinical Investigation | 2003

CD28-dependent Rac1 activation is the molecular target of azathioprine in primary human CD4+ T lymphocytes

Imke Tiede; Gerhard Fritz; Susanne Strand; Daniela Poppe; Radovan Dvorsky; Dennis Strand; Hans A. Lehr; Stefan Wirtz; Christoph Becker; Raja Atreya; Jonas Mudter; Kai Hildner; Brigitte Bartsch; Martin H. Holtmann; Richard S. Blumberg; Henning Walczak; Heiko Iven; Peter R. Galle; Mohammad Reza Ahmadian; Markus F. Neurath

Azathioprine and its metabolite 6-mercaptopurine (6-MP) are immunosuppressive drugs that are used in organ transplantation and autoimmune and chronic inflammatory diseases such as Crohn disease. However, their molecular mechanism of action is unknown. In the present study, we have identified a unique and unexpected role for azathioprine and its metabolites in the control of T cell apoptosis by modulation of Rac1 activation upon CD28 costimulation. We found that azathioprine and its metabolites induced apoptosis of T cells from patients with Crohn disease and control patients. Apoptosis induction required costimulation with CD28 and was mediated by specific blockade of Rac1 activation through binding of azathioprine-generated 6-thioguanine triphosphate (6-Thio-GTP) to Rac1 instead of GTP. The activation of Rac1 target genes such as mitogen-activated protein kinase kinase (MEK), NF-kappaB, and bcl-x(L) was suppressed by azathioprine, leading to a mitochondrial pathway of apoptosis. Azathioprine thus converts a costimulatory signal into an apoptotic signal by modulating Rac1 activity. These findings explain the immunosuppressive effects of azathioprine and suggest that 6-Thio-GTP derivates may be useful as potent immunosuppressive agents in autoimmune diseases and organ transplantation.


Nature Reviews Cancer | 2011

Rethinking ovarian cancer: recommendations for improving outcomes.

Sebastian Vaughan; Jermaine Coward; Robert C. Bast; Andrew Berchuck; Jonathan S. Berek; James D. Brenton; George Coukos; Christopher C. Crum; Ronny Drapkin; Dariush Etemadmoghadam; Michael Friedlander; Hani Gabra; Stan B. Kaye; Christopher J. Lord; Ernst Lengyel; Douglas A. Levine; Iain A. McNeish; Usha Menon; Gordon B. Mills; Kenneth P. Nephew; Amit M. Oza; Anil K. Sood; Euan A. Stronach; Henning Walczak; David Bowtell; Frances R. Balkwill

There have been major advances in our understanding of the cellular and molecular biology of the human malignancies that are collectively referred to as ovarian cancer. At a recent Helene Harris Memorial Trust meeting, an international group of researchers considered actions that should be taken to improve the outcome for women with ovarian cancer. Nine major recommendations are outlined in this Opinion article.


Nature | 2011

Linear ubiquitination prevents inflammation and regulates immune signalling

Björn Gerlach; Stefanie M. Cordier; Anna C. Schmukle; Christoph H. Emmerich; Eva Rieser; Tobias Haas; Andrew I. Webb; James A Rickard; Holly Anderton; W. Wei-Lynn Wong; Ueli Nachbur; Lahiru Gangoda; Uwe Warnken; Anthony W. Purcell; John Silke; Henning Walczak

Members of the tumour necrosis factor (TNF) receptor superfamily have important functions in immunity and inflammation. Recently linear ubiquitin chains assembled by a complex containing HOIL-1 and HOIP (also known as RBCK1 and RNF31, respectively) were implicated in TNF signalling, yet their relevance in vivo remained uncertain. Here we identify SHARPIN as a third component of the linear ubiquitin chain assembly complex, recruited to the CD40 and TNF receptor signalling complexes together with its other constituents, HOIL-1 and HOIP. Mass spectrometry of TNF signalling complexes revealed RIP1 (also known as RIPK1) and NEMO (also known as IKKγ or IKBKG) to be linearly ubiquitinated. Mutation of the Sharpin gene (Sharpincpdm/cpdm) causes chronic proliferative dermatitis (cpdm) characterized by inflammatory skin lesions and defective lymphoid organogenesis. Gene induction by TNF, CD40 ligand and interleukin-1β was attenuated in cpdm-derived cells which were rendered sensitive to TNF-induced death. Importantly, Tnf gene deficiency prevented skin lesions in cpdm mice. We conclude that by enabling linear ubiquitination in the TNF receptor signalling complex, SHARPIN interferes with TNF-induced cell death and, thereby, prevents inflammation. Our results provide evidence for the relevance of linear ubiquitination in vivo in preventing inflammation and regulating immune signalling.


Molecular Cell | 2009

Recruitment of the linear ubiquitin chain assembly complex stabilizes the TNF-R1 signaling complex and is required for TNF-mediated gene induction.

Tobias Haas; Christoph H. Emmerich; Björn Gerlach; Anna C. Schmukle; Stefanie M. Cordier; Eva Rieser; Rebecca Feltham; James E. Vince; Uwe Warnken; Till Wenger; Ronald Koschny; David Komander; John Silke; Henning Walczak

TNF is a key inflammatory cytokine. Using a modified tandem affinity purification approach, we identified HOIL-1 and HOIP as functional components of the native TNF-R1 signaling complex (TNF-RSC). Together, they were shown to form a linear ubiquitin chain assembly complex (LUBAC) and to ubiquitylate NEMO. We show that LUBAC binds to ubiquitin chains of different linkage types and that its recruitment to the TNF-RSC is impaired in TRADD-, TRAF2-, and cIAP1/2- but not in RIP1- or NEMO-deficient MEFs. Furthermore, the E3 ligase activity of cIAPs, but not TRAF2, is required for HOIL-1 recruitment to the TNF-RSC. LUBAC enhances NEMO interaction with the TNF-RSC, stabilizes this protein complex, and is required for efficient TNF-induced activation of NF-kappaB and JNK, resulting in apoptosis inhibition. Finally, we demonstrate that sustained stability of the TNF-RSC requires LUBACs enzymatic activity, thereby adding a third form of ubiquitin linkage to the triggering of TNF signaling by the TNF-RSC.


The EMBO Journal | 2002

Caspase‐10 is recruited to and activated at the native TRAIL and CD95 death‐inducing signalling complexes in a FADD‐dependent manner but can not functionally substitute caspase‐8

Martin R. Sprick; Eva Rieser; Heiko Stahl; Anne Grosse-Wilde; Markus A. Weigand; Henning Walczak

The involvement of the death adaptor protein FADD and the apoptosis‐initiating caspase‐8 in CD95 and TRAIL death signalling has recently been demonstrated by the analysis of the native death‐inducing signalling complex (DISC) that forms upon ligand‐induced receptor cross‐linking. However, the role of caspase‐10, the other death‐effector‐domain‐containing caspase besides caspase‐8, in death receptor signalling has been controversial. Here we show that caspase‐10 is recruited not only to the native TRAIL DISC but also to the native CD95 DISC, and that FADD is necessary for its recruitment to and activation at these two protein complexes. With respect to the function of caspase‐10, we show that it is not required for apoptosis induction. In addition, caspase‐10 can not substitute for caspase‐8, as the defect in apoptosis induction observed in caspase‐8‐deficient cells could not be rescued by overexpression of caspase‐10. Finally, we demonstrate that caspase‐10 is cleaved during CD95‐induced apoptosis of activated T cells. These results show that caspase‐10 activation occurs in primary cells, but that its function differs from that of caspase‐8.

Collaboration


Dive into the Henning Walczak's collaboration.

Top Co-Authors

Avatar

Peter H. Krammer

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Martin R. Sprick

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Tom M. Ganten

University Hospital Heidelberg

View shared research outputs
Top Co-Authors

Avatar

Jaromir Sykora

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Maurice Darding

University College London

View shared research outputs
Top Co-Authors

Avatar

Jens Dhein

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Eva Rieser

University College London

View shared research outputs
Top Co-Authors

Avatar

Peter Draber

University College London

View shared research outputs
Researchain Logo
Decentralizing Knowledge