Network


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

Hotspot


Dive into the research topics where Martin Herrmann is active.

Publication


Featured researches published by Martin Herrmann.


Nature | 1997

Immunosuppressive effects of apoptotic cells

Reinhard E. Voll; Martin Herrmann; Edith Roth; Christian Stach; Joachim R. Kalden; Irute Girkontaite

Apoptotic cell death is important in the development and homeostasis of multicellular organisms and is a highly controlled means of eliminating dangerous, damaged or unnecessary cells without causing an inflammatory response or tissue damage,. We now show that the presence of apoptotic cells during monocyte activation increases their secretion of the anti-inflammatory and immunoregulatory cytokine interleukin 10 (IL-10) and decreases secretion of the proinflammatory cytokines tumour necrosis factor-α (TNF-α), IL-1 and IL-12. This may inhibit inflammation and contribute to impaired cell-mediated immunity in conditions associated with increased apoptosis, such as viral infections, pregnancy, cancer and exposure to radiation.


Arthritis & Rheumatism | 1998

Impaired phagocytosis of apoptotic cell material by monocyte-derived macrophages from patients with systemic lupus erythematosus.

Martin Herrmann; Reinhard E. Voll; Otmar Zoller; Manuela Hagenhofer; Botond B. Ponner; Joachim R. Kalden

OBJECTIVE To investigate whether the established impaired phagocyte function in systemic lupus erythematosus (SLE) patients also affects apoptotic cell clearance. Accumulation of apoptotic waste as a source for autoantigens that induce and maintain autoimmune responses is discussed. METHODS Apoptosis was detected by morphology and propidium iodide staining. In vitro phagocytosis of autologous apoptotic cells in cultured peripheral blood mononuclear cells was evaluated microscopically. Cross-feeding experiments were performed to investigate phagocytosis of heterologous apoptotic cells by in vitro-differentiated macrophages. Furthermore, the effect of annexin V on the phagocytosis of apoptotic cells was investigated. RESULTS Reduced clearance of apoptotic cells in SLE patients was observed. The defective clearance appeared to reflect phagocyte dysfunction and not an abnormal execution of apoptosis. A similar picture was seen when in vitro-differentiated macrophages from control populations were treated with annexin V. CONCLUSION Noninflammatory engulfment phagocytosis of apoptotic cells is decreased in SLE patients. Persistently circulating apoptotic waste may encounter inflammatory removal pathways and serve as immunogen for the induction of autoreactive lymphocytes and as antigen for immune complex formation.


Arthritis & Rheumatism | 2002

Impaired uptake of apoptotic cells into tingible body macrophages in germinal centers of patients with systemic lupus erythematosus

Irith Baumann; Wasilis Kolowos; Reinhard E. Voll; Bernhard Manger; Udo S. Gaipl; Winfried Neuhuber; Thomas Kirchner; Joachim R. Kalden; Martin Herrmann

OBJECTIVE To investigate the fate of apoptotic cells in the germinal centers (GCs) of patients with systemic lupus erythematosus (SLE). METHODS Lymph node biopsy specimens obtained from 7 SLE patients with benign follicular hyperplasia, 5 non-SLE patients with benign follicular hyperplasia (non-SLE), 5 patients with malignant follicular lymphoma, and 3 patients with dermatopathic lymphadenitis were stained with monoclonal antibodies against macrophages (CD68) and follicular dendritic cells (CR2/CD21). TUNEL staining and transmission electron microscopy were performed to detect apoptotic cells. Confocal microscopy was used to evaluate the in vivo capacity of tingible body macrophages to remove apoptotic cell material. RESULTS In a subgroup of patients with SLE, apoptotic cells accumulated in the GCs of the lymph nodes. The number of tingible body macrophages, which usually contained engulfed apoptotic nuclei, was significantly reduced in these patients. In contrast to what was observed in all controls, TUNEL-positive apoptotic material from SLE patients was observed to be directly associated with the surfaces of follicular dendritic cells (FDCs). CONCLUSION Our findings suggest that in a sub-group of SLE patients, apoptotic cells are not properly cleared by tingible body macrophages of the GCs. Consequently, nuclear autoantigens bind to FDCs and may thus provide survival signals for autoreactive B cells. This action may override an important control mechanism for B cell development, resulting in the loss of tolerance for nuclear antigens.


Journal of Immunology | 2005

Release of High Mobility Group Box 1 by Dendritic Cells Controls T Cell Activation via the Receptor for Advanced Glycation End Products

Ingrid E. Dumitriu; Paramita Baruah; Barbara Valentinis; Reinhard E. Voll; Martin Herrmann; Peter P. Nawroth; Bernd Arnold; Marco Bianchi; Angelo A. Manfredi; Patrizia Rovere-Querini

High mobility group box 1 (HMGB1) is an abundant and conserved nuclear protein that is released by necrotic cells and acts in the extracellular environment as a primary proinflammatory signal. In this study we show that human dendritic cells, which are specialized in Ag presentation to T cells, actively release their own HMGB1 into the extracellular milieu upon activation. This secreted HMGB1 is necessary for the up-regulation of CD80, CD83, and CD86 surface markers of human dendritic cells and for IL-12 production. The HMGB1 secreted by dendritic cells is also required for the clonal expansion, survival, and functional polarization of naive T cells. Using neutralizing Abs and receptor for advanced glycation end product-deficient (RAGE−/−) cells, we demonstrate that RAGE is required for the effect of HMGB1 on dendritic cells. HMGB1/RAGE interaction results in downstream activation of MAPKs and NF-κB. The use of an ancient signal of necrosis, HMGB1, by dendritic cells to sustain their own maturation and for activation of T lymphocytes represents a profitable evolutionary mechanism.


Nature Reviews Rheumatology | 2010

The role of defective clearance of apoptotic cells in systemic autoimmunity

Luis E. Munoz; Kirsten Lauber; Martin Schiller; Angelo A. Manfredi; Martin Herrmann

The inefficient clearance of dying cells can result in the accumulation of apoptotic cell remnants. This occurrence is considered an intrinsic defect that can cause the permanent presence of cellular debris responsible for the initiation of systemic autoimmunity in diseases such as systemic lupus erythematosus (SLE). If postapoptotic debris accumulates in germinal centers, activates complement and functions as a survival signal for B cells that have become autoreactive by somatic hypermutation, autoimmunity could arise (etiology). The accumulation of postapoptotic remnants and fragments derived from secondary necrotic cells in the presence of autoantibodies against apoptotic cells or adaptor molecules obliges their pathological elimination and maintains autoinflammation. The autoimmunity that occurs in patients with SLE involves complex antigens that contain nucleic acids, which can function as virus mimetics. Complexes of autoantibodies, proteins and nucleic acids are likely to be mistaken by the immune system for opsonized viruses, resulting in the production of type I interferons, a hallmark of SLE (pathogenesis). The pathogenicity of autoantibodies is thought to strongly increase if autoantigens are accessible for immune-complex formation. The immune complex could be considered a binary pyrogen formed from less proinflammatory components. The accessibility of cognate autoantigens, in turn, is likely to be related to impaired or delayed clearance of apoptotic cells.


Arthritis & Rheumatism | 2009

Altered skeletal expression of sclerostin and its link to radiographic progression in ankylosing spondylitis

Heiner Appel; G. Ruiz-Heiland; Joachim Listing; Jochen Zwerina; Martin Herrmann; Ruediger B. Mueller; Hildrun Haibel; Xenofon Baraliakos; Axel Hempfing; Martin Rudwaleit; Joachim Sieper; Georg Schett

OBJECTIVE Osteocytes are considered to be sensors of bone damage and regulators of bone mass by specifically expressing sclerostin, an inhibitor of bone formation. The contribution of osteocytes in regulating local bone remodeling in arthritis is unknown. The aim of this study was to investigate the role of osteocytes as contributors to bone remodeling in ankylosing spondylitis (AS). METHODS Sclerostin expression and osteocyte death were assessed by immunohistochemistry in joints derived from patients with AS, patients with rheumatoid arthritis (RA), and patients with osteoarthritis (OA), as well as from control subjects. In addition, the serum level of sclerostin was assessed by enzyme-linked immunosorbent assay in healthy subjects and patients with AS; this assessment included the longitudinal correlation of sclerostin serum levels and radiographic progression in the spine of patients with AS. RESULTS Sclerostin expression was confined exclusively to osteocytes. Whereas the majority of osteocytes in healthy individuals and patients with RA were sclerostin positive, expression was significantly reduced in patients with OA and was virtually absent in patients with AS. Moreover, serum levels of sclerostin were significantly lower in patients with AS than in healthy individuals. Importantly, low serum sclerostin levels in patients with AS were significantly associated with the formation of new syndesmophytes (P = 0.007). CONCLUSION Sclerostin expression is impaired in patients with AS, suggesting a specific alteration of osteocyte function in this disease. A low serum level of sclerostin in the setting of AS is linked to increased structural damage, emphasizing the role of sclerostin in the suppression of bone formation.


OncoImmunology | 2014

Consensus guidelines for the detection of immunogenic cell death

Oliver Kepp; Laura Senovilla; Ilio Vitale; Erika Vacchelli; Sandy Adjemian; Patrizia Agostinis; Lionel Apetoh; Fernando Aranda; Vincenzo Barnaba; Norma Bloy; Laura Bracci; Karine Breckpot; David Brough; Aitziber Buqué; Maria G. Castro; Mara Cirone; María I. Colombo; Isabelle Cremer; Sandra Demaria; Luciana Dini; Aristides G. Eliopoulos; Alberto Faggioni; Silvia C. Formenti; Jitka Fucikova; Lucia Gabriele; Udo S. Gaipl; Jérôme Galon; Abhishek D. Garg; François Ghiringhelli; Nathalia A. Giese

Apoptotic cells have long been considered as intrinsically tolerogenic or unable to elicit immune responses specific for dead cell-associated antigens. However, multiple stimuli can trigger a functionally peculiar type of apoptotic demise that does not go unnoticed by the adaptive arm of the immune system, which we named “immunogenic cell death” (ICD). ICD is preceded or accompanied by the emission of a series of immunostimulatory damage-associated molecular patterns (DAMPs) in a precise spatiotemporal configuration. Several anticancer agents that have been successfully employed in the clinic for decades, including various chemotherapeutics and radiotherapy, can elicit ICD. Moreover, defects in the components that underlie the capacity of the immune system to perceive cell death as immunogenic negatively influence disease outcome among cancer patients treated with ICD inducers. Thus, ICD has profound clinical and therapeutic implications. Unfortunately, the gold-standard approach to detect ICD relies on vaccination experiments involving immunocompetent murine models and syngeneic cancer cells, an approach that is incompatible with large screening campaigns. Here, we outline strategies conceived to detect surrogate markers of ICD in vitro and to screen large chemical libraries for putative ICD inducers, based on a high-content, high-throughput platform that we recently developed. Such a platform allows for the detection of multiple DAMPs, like cell surface-exposed calreticulin, extracellular ATP and high mobility group box 1 (HMGB1), and/or the processes that underlie their emission, such as endoplasmic reticulum stress, autophagy and necrotic plasma membrane permeabilization. We surmise that this technology will facilitate the development of next-generation anticancer regimens, which kill malignant cells and simultaneously convert them into a cancer-specific therapeutic vaccine.


Journal of Experimental Medicine | 2004

Inhibition of Phosphatidylserine Recognition Heightens the Immunogenicity of Irradiated Lymphoma Cells In Vivo

Attilio Bondanza; Valérie S. Zimmermann; Patrizia Rovere-Querini; Javier Turnay; Ingrid E. Dumitriu; Christian Stach; Reinhard E. Voll; Udo S. Gaipl; Wolf Bertling; Ernst Pöschl; Joachim R. Kalden; Angelo A. Manfredi; Martin Herrmann

Strategies to enhance the immunogenicity of tumors are urgently needed. Although vaccination with irradiated dying lymphoma cells recruits a tumor-specific immune response, its efficiency as immunogen is poor. Annexin V (AxV) binds with high affinity to phosphatidylserine on the surface of apoptotic and necrotic cells and thereby impairs their uptake by macrophages. Here, we report that AxV preferentially targets irradiated lymphoma cells to CD8+ dendritic cells for in vivo clearance, elicits the release of proinflammatory cytokines and dramatically enhances the protection elicited against the tumor. The response was endowed with both memory, because protected animals rejected living lymphoma cells after 72 d, and specificity, because vaccinated animals failed to reject unrelated neoplasms. Finally, AxV–coupled irradiated cells induced the regression of growing tumors. These data indicate that endogenous adjuvants that bind to dying tumor cells can be exploited to target tumors for immune rejection.


Immunity | 2012

12/15-Lipoxygenase Orchestrates the Clearance of Apoptotic Cells and Maintains Immunologic Tolerance

Stefan Uderhardt; Martin Herrmann; Olga Oskolkova; Susanne Aschermann; Wolfgang Bicker; Natacha Ipseiz; Kerstin Sarter; Benjamin Frey; Tobias Rothe; Reinhard E. Voll; Falk Nimmerjahn; Valery N. Bochkov; Georg Schett; Gerhard Krönke

Noninflammatory clearance of apoptotic cells (ACs) is crucial to maintain self-tolerance. Here, we have reported a role for the enzyme 12/15-lipoxygenase (12/15-LO) as a central factor governing the sorting of ACs into differentially activated monocyte subpopulations. During inflammation, uptake of ACs was confined to a population of 12/15-LO-expressing, alternatively activated resident macrophages (resMΦ), which blocked uptake of ACs into freshly recruited inflammatory Ly6C(hi) monocytes in a 12/15-LO-dependent manner. ResMΦ exposed 12/15-LO-derived oxidation products of phosphatidylethanolamine (oxPE) on their plasma membranes and thereby generated a sink for distinct soluble receptors for ACs such as milk fat globule-EGF factor 8, which were essential for the uptake of ACs into inflammatory monocytes. Loss of 12/15-LO activity, in turn, resulted in an aberrant phagocytosis of ACs by inflammatory monocytes, subsequent antigen presentation of AC-derived antigens, and a lupus-like autoimmune disease. Our data reveal an unexpected key role for enzymatic lipid oxidation during the maintenance of self-tolerance.


Autoimmunity Reviews | 2010

Autoimmunity and chronic inflammation — Two clearance-related steps in the etiopathogenesis of SLE

Luis E. Munoz; Christina Janko; Connie Schulze; Christine Schorn; Kerstin Sarter; Georg Schett; Martin Herrmann

Systemic lupus erythematosus (SLE) is an autoimmune disease with very prominent chronic inflammatory aspects that render into multiple symptoms and clinical signs. The precise etiology of SLE remains elusive; however, it is known that its etiopathogenesis is of multifactorial nature. The production of autoantibodies (AAb) targeting double stranded DNA (dsDNA) and other nuclear autoantigens is the main characteristic of this disease. These target antigens are often modified and/or translocated when apoptotic cells undergo secondary necrosis as a consequence of the clearance deficiency in patients with SLE. In healthy individuals, dead and dying cells are rapidly removed by macrophages in an anti-inflammatory context; this does not elicit immune responses. In SLE, apoptotic cells are often not properly cleared; autoantigens leak out, and are subsequently presented to B cells by follicular dendritic cells (FDC) in secondary lymphoid tissues. This defect challenges the peripheral self-tolerance. Autoreactive B cell activation and production of anti-nuclear AAb result as the first step in the etiopathogenesis of SLE. The second step is the formation of immune complexes (IC) with apoptotic cell-derived nuclear remnants either in situ or deposited in various tissues. Nucleic acid-containing IC may also be ingested by phagocytes, which subsequently produce pro-inflammatory cytokines. Both processes result in chronic organ and tissue damage, development and maintenance of the systemic autoimmune disease. In conclusion, clearance deficiency may contribute to SLE in two ways: first, in germinal centres it enables the affinity maturation of autoreactive B cells and second, in peripheral tissues it leads to the accumulation of accessible nuclear autoantigens. Chronic inflammation in SLE is consequently promoted by the persistently binding of AAb with their cognate autoantigens forming a binary weapon: the nucleic acid-containing IC.

Collaboration


Dive into the Martin Herrmann's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Luis E. Munoz

University of Erlangen-Nuremberg

View shared research outputs
Top Co-Authors

Avatar

Udo S. Gaipl

University of Erlangen-Nuremberg

View shared research outputs
Top Co-Authors

Avatar

Georg Schett

University of Erlangen-Nuremberg

View shared research outputs
Top Co-Authors

Avatar

Joachim R. Kalden

University of Erlangen-Nuremberg

View shared research outputs
Top Co-Authors

Avatar

Christina Janko

University of Erlangen-Nuremberg

View shared research outputs
Top Co-Authors

Avatar

Rostyslav Bilyy

Danylo Halytsky Lviv National Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

J. R. Kalden

University of Erlangen-Nuremberg

View shared research outputs
Top Co-Authors

Avatar

Peter Kern

University of Erlangen-Nuremberg

View shared research outputs
Researchain Logo
Decentralizing Knowledge