Network


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

Hotspot


Dive into the research topics where Nadine Hövelmeyer is active.

Publication


Featured researches published by Nadine Hövelmeyer.


Nature Medicine | 2005

Experimental autoimmune encephalomyelitis repressed by microglial paralysis

Frank L. Heppner; Melanie Greter; Denis Marino; Jeppe Falsig; Gennadij Raivich; Nadine Hövelmeyer; Ari Waisman; Thomas Rülicke; Marco Prinz; Josef Priller; Burkhard Becher; Adriano Aguzzi

Although microglial activation occurs in inflammatory, degenerative and neoplastic central nervous system (CNS) disorders, its role in pathogenesis is unclear. We studied this question by generating CD11b-HSVTK transgenic mice, which express herpes simplex thymidine kinase in macrophages and microglia. Ganciclovir treatment of organotypic brain slice cultures derived from CD11b-HSVTK mice abolished microglial release of nitrite, proinflammatory cytokines and chemokines. Systemic ganciclovir administration to CD11b-HSVTK mice elicited hematopoietic toxicity, which was prevented by transfer of wild-type bone marrow. In bone marrow chimeras, ganciclovir blocked microglial activation in the facial nucleus upon axotomy and repressed the development of experimental autoimmune encephalomyelitis. We conclude that microglial paralysis inhibits the development and maintenance of inflammatory CNS lesions. The microglial compartment thus provides a potential therapeutic target in inflammatory CNS disorders. These results validate CD11b-HSVTK mice as a tool to study the impact of microglial activation on CNS diseases in vivo.


Journal of Immunology | 2005

Apoptosis of Oligodendrocytes via Fas and TNF-R1 Is a Key Event in the Induction of Experimental Autoimmune Encephalomyelitis

Nadine Hövelmeyer; Zhenyue Hao; Ksanthi Kranidioti; George Kassiotis; Thorsten Buch; Friederike Frommer; Lennart von Hoch; Dana Krämer; Liliana Minichiello; George Kollias; Hans Lassmann; Ari Waisman

In experimental autoimmune encephalomyelitis (EAE), an animal model for multiple sclerosis, immunization with myelin Ags leads to demyelination and paralysis. To investigate which molecules are crucial for the pathogenesis of EAE, we specifically assessed the roles of the death receptors Fas and TNF-R1. Mice lacking Fas expression in oligodendrocytes (ODCs) were generated and crossed to TNF-R1-deficient mice. To achieve specific deletion of a loxP-flanked fas allele in ODCs, we generated a new insertion transgene, expressing the Cre recombinase specifically in ODCs. Fas inactivation alone as well as the complete absence of TNF-R1 protected mice partially from EAE induced by the immunization with myelin ODC glycoprotein. The double-deficient mice, however, showed almost no clinical signs of EAE after immunization. Histological analysis revealed that demyelination was suppressed in CNS tissue and that lymphocyte infiltration was notably reduced. We conclude that the death receptors Fas and TNF-R1 are major initiators of ODC apoptosis in EAE. Although only moderate reduction of lymphocyte infiltration into CNS tissue was observed, the absence of these receptors appears to confer protection from demyelination and development of clinical disease.


Journal of Experimental Medicine | 2007

Regulation of B cell homeostasis and activation by the tumor suppressor gene CYLD

Nadine Hövelmeyer; F. Thomas Wunderlich; Ramin Massoumi; Charlotte G. Jakobsen; Jian Song; Marcus A. Wörns; Carsten Merkwirth; Andrew Kovalenko; Monique Aumailley; Dennis Strand; Jens C. Brüning; Peter R. Galle; David Wallach; Reinhard Fässler; Ari Waisman

B cell homeostasis is regulated by multiple signaling processes, including nuclear factor-κB (NF-κB), BAFF-, and B cell receptor signaling. Conditional disruption of genes involved in these pathways has shed light on the mechanisms governing signaling from the cell surface to the nucleus. We describe a novel mouse strain that expresses solely and excessively a naturally occurring splice variant of CYLD (CYLDex7/8 mice), which is a deubiquitinating enzyme that is integral to NF-κB signaling. This shorter CYLD protein lacks the TRAF2 and NEMO binding sites present in full-length CYLD. A dramatic expansion of mature B lymphocyte populations in all peripheral lymphoid organs occurs in this strain. The B lymphocytes themselves exhibit prolonged survival and manifest a variety of signaling disarrangements that do not occur in mice with a complete deletion of CYLD. Although both the full-length and the mutant CYLD are able to interact with Bcl-3, a predominant nuclear accumulation of Bcl-3 occurs in the CYLD mutant B cells. More dramatic, however, is the accumulation of the NF-κB proteins p100 and RelB in CYLDex7/8 B cells, which, presumably in combination with nuclear Bcl-3, results in increased levels of Bcl-2 expression. These findings suggest that CYLD can both positively and negatively regulate signal transduction and homeostasis of B cells in vivo, depending on the expression of CYLD splice variants.


European Journal of Immunology | 2011

A20 deficiency in B cells enhances B-cell proliferation and results in the development of autoantibodies

Nadine Hövelmeyer; Sonja Reissig; Nguyen Thi Xuan; Petra Adams-Quack; Dominika Lukas; Alexei Nikolaev; Dirk Schlüter; Ari Waisman

A20/TNFAIP3 is an ubiquitin‐editing enzyme, important for the regulation of the NF‐κB pathway. Mutations in the TNFAIP3 gene have been linked to different human autoimmune disorders. In human B‐cell lymphomas, the inactivation of A20 results in constitutive NF‐κB activation. Recent studies demonstrate that in mice the germline inactivation of A20 leads to early lethality, due to inflammation in multiple organs of the body. In this report, we describe a new mouse strain allowing for the tissue‐specific deletion of A20. We show that B‐cell‐specific deletion of A20 results in a dramatic reduction in marginal zone B cells. Furthermore, A20‐deficient B cells display a hyperactive phenotype represented by enhanced proliferation upon activation. Finally, these mice develop higher levels of serum immunoglobulins, resulting in an excessive production of self‐reactive autoantibodies.


Brain | 2010

Smad7 in T cells drives T helper 1 responses in multiple sclerosis and experimental autoimmune encephalomyelitis.

Ingo Kleiter; Jian Song; Dominika Lukas; Maruf Hasan; Bernhard Neumann; Andrew L. Croxford; Xiomara Pedré; Nadine Hövelmeyer; Nir Yogev; Alexander Mildner; Marco Prinz; Elena Wiese; Kurt Reifenberg; Stefan Bittner; Heinz Wiendl; Lawrence Steinman; Christoph Becker; Ulrich Bogdahn; Markus F. Neurath; Andreas Steinbrecher; Ari Waisman

Autoreactive CD4+ T lymphocytes play a vital role in the pathogenesis of multiple sclerosis and its animal model, experimental autoimmune encephalomyelitis. Since the discovery of T helper 17 cells, there is an ongoing debate whether T helper 1, T helper 17 or both subtypes of T lymphocytes are important for the initiation of autoimmune neuroinflammation. We examined peripheral blood CD4+ cells from patients with active and stable relapsing–remitting multiple sclerosis, and used mice with conditional deletion or over-expression of the transforming growth factor-β inhibitor Smad7, to delineate the role of Smad7 in T cell differentiation and autoimmune neuroinflammation. We found that Smad7 is up-regulated in peripheral CD4+ cells from patients with multiple sclerosis during relapse but not remission, and that expression of Smad7 strongly correlates with T-bet, a transcription factor defining T helper 1 responses. Concordantly, mice with transgenic over-expression of Smad7 in T cells developed an enhanced disease course during experimental autoimmune encephalomyelitis, accompanied by elevated infiltration of inflammatory cells and T helper 1 responses in the central nervous system. On the contrary, mice with a T cell-specific deletion of Smad7 had reduced disease and central nervous system inflammation. Lack of Smad7 in T cells blunted T cell proliferation and T helper 1 responses in the periphery but left T helper 17 responses unaltered. Furthermore, frequencies of regulatory T cells were increased in the central nervous system of mice with a T cell-specific deletion and reduced in mice with a T cell-specific over-expression of Smad7. Downstream effects of transforming growth factor-β on in vitro differentiation of naïve T cells to T helper 1, T helper 17 and regulatory T cell phenotypes were enhanced in T cells lacking Smad7. Finally, Smad7 was induced during T helper 1 differentiation and inhibited during T helper 17 differentiation. Taken together, the level of Smad7 in T cells determines T helper 1 polarization and regulates inflammatory cellular responses. Since a Smad7 deletion in T cells leads to immunosuppression, Smad7 may be a potential new therapeutic target in multiple sclerosis.


Journal of Immunology | 2012

The Tumor Suppressor CYLD Controls the Function of Murine Regulatory T Cells

Sonja Reissig; Nadine Hövelmeyer; Benno Weigmann; Alexei Nikolaev; Thomas Wunderlich; Matthias Hahn; Marcus F. Neurath; Ari Waisman

CYLD was originally identified as a tumor suppressor gene mutated in familial cylindromatosis, an autosomal dominant predisposition to multiple benign neoplasms of the skin known as cylindromas. The CYLD protein is a deubiquitinating enzyme that acts as a negative regulator of NF-κB and JNK signaling through its interaction with NEMO and TNFR-associated factor 2. We have previously described a novel mouse strain that expresses solely and excessively a naturally occurring splice variant of CYLD (CYLDex7/8). In this study, we demonstrate that CYLD plays a critical role in Treg development and function. T cells of CYLDex7/8 mice had a hyperactive phenotype manifested by increased production of inflammatory cytokines and constitutive activation of the NF-κB pathway. Furthermore, the amount of Foxp3+ regulatory T cells in these mice was markedly enhanced in thymus and peripheral organs. Importantly, these regulatory T cells displayed decreased expression levels of CD25 and CTLA-4 associated with impaired suppressive capacity. Hence, our data emphasize an essential role of CYLD in maintaining T cell homeostasis as well as normal T regulatory cell function, thereby controlling abnormal T cell responses.


Journal of Hepatology | 2012

Liver specific deletion of CYLDexon7/8 induces severe biliary damage, fibrosis and increases hepatocarcinogenesis in mice.

Toni Urbanik; Rj Boger; Thomas Longerich; Katharina Becker; Karl Roland Ehrenberg; Nadine Hövelmeyer; Matthias Hahn; Marcus Schuchmann; Dirk Jäger; Ari Waisman; Marcus A. Wörns; Henning Schulze-Bergkamen

BACKGROUND & AIMSnCYLD is a tumor suppressor gene that is mutated in familial cylindromatosis, an autosomal dominant predisposition to tumors of skin appendages. Reduced CYLD expression has been observed in other tumor entities, including hepatocellular carcinoma. In the present study, we analyzed the role of CYLD in liver homeostasis and hepatocarcinogenesis in vivo.nnnMETHODSnMice with liver-specific deletion of CYLDexon7/8 (CYLD(FF)xAlbCre) were generated. Liver tissues were histologically analyzed and oval cell activation was investigated. Hepatocarcinogenesis was induced by diethylnitrosamine/phenobarbital (DEN/PB). Microarray expression profiling of livers was performed in untreated as well as DEN/PB-treated mice. NF-κB signaling was assessed by ELISA, quantitative real-time PCR, and Western blotting.nnnRESULTSnCYLD(FF)xAlbCre hepatocytes and cholangiocytes did not express full-length CYLD (FL-CYLD) protein but showed increased expression of the naturally occurring short-CYLD splice variant (s-CYLD). CYLD(FF)xAlbCre mice exhibited a prominent biliary phenotype with ductular reaction and biliary-type fibrosis. In addition, CYLD(FF)xAlbCre mice showed a significantly increased sensitivity towards DEN/PB-induced hepatocarcinogenesis. Moreover, we could observe the development of cholangiocellular carcinoma, in line with enhanced oval cell activity. NF-κB-signaling was increased in livers of CYLD(FF)xAlbCre mice and likely contributed to the inflammatory and fibrotic response.nnnCONCLUSIONSnThe deletion of exon7/8 of the CYLD gene activates oval cells, leads to a biliary phenotype, and increases the susceptibility towards carcinogenesis in the liver. Thus, our study presents a novel model of biliary damage and liver fibrosis, followed by cancer development.


Blood | 2009

Naturally occurring short splice variant of CYLD positively regulates dendritic cell function

Cathy Cecilia Srokowski; Joumana Masri; Nadine Hövelmeyer; Anna Katharina Krembel; Christine Tertilt; Dennis Strand; Karsten Mahnke; Ramin Massoumi; Ari Waisman; Hansjörg Schild

Deubiquitination of NF-kappaB members by CYLD is crucial in controlling the magnitude and nature of cell activation. The role of the naturally occurring CYLD splice variant in dendritic cell (DC) function was analyzed using CYLD(ex7/8) mice, which lack the full-length CYLD (flCYLD) transcript and overexpress the short splice variant (sCYLD). Bone marrow-derived DCs from CYLD(ex7/8) mice display a hyperactive phenotype in vitro and in vivo and have a defect in establishing tolerance with the use of DEC-205-mediated antigen targeting to resting DCs. The combination of sCYLD overexpression and lack of flCYLD in CYLD(ex7/8) DCs leads to enhanced NF-kappaB activity accompanied by an increased nuclear translocation of the IkappaB molecule Bcl-3, along with nuclear p50 and p65. This suggests that, in contrast to flCYLD, sCYLD is a positive regulator of NF-kappaB activity, and its overexpression induces a hyperactive phenotype in DCs.


European Journal of Immunology | 2014

Overexpression of Bcl-3 inhibits the development of marginal zone B cells.

Nadine Hövelmeyer; Marcus A. Wörns; Sonja Reissig; Petra Adams-Quack; Jennifer Leclaire; Matthias Hahn; Simone Wörtge; Alexei Nikolaev; Peter R. Galle; Ari Waisman

The transcription factor Bcl‐3 functions as a proto‐oncogene via regulation of cell proliferation and apoptosis. Bcl‐3 is an atypical member of the IκB family and plays a central role in the immune response through interactions with the NF‐κB subunits p50 and p52. To investigate the impact of Bcl‐3 on B‐cell maturation and regulation, we generated mice that overexpress Bcl‐3 specifically in B cells. Interestingly, these mice lack marginal zone B cells and exhibit a significant reduction in the number of B‐1 B cells. Further, B cells from these mice are impaired in their proliferative capacity. Our data demonstrate that the overexpression of the transcription factor Bcl‐3 inhibits germinal center formation, marginal zone B‐cell development, and affects the B‐1 B‐cell compartment.


European Journal of Immunology | 2016

NF-κB-inducing kinase is essential for B-cell maintenance in mice.

Matthias Hahn; Anna Macht; Ari Waisman; Nadine Hövelmeyer

NF‐κB‐inducing kinase (NIK) is a key mediator of the noncanonical NF‐κB signaling pathway, which is critical for normal B‐cell development and function. It is well established that the complete deletion of NIK in mice results in defective B cells and impaired secondary lymphoid organogenesis. To address the role of NIK deficiency specifically in B cells, we generated a new mouse strain for the conditional deletion of this kinase. Deletion of NIK during B‐cell development results in a drastic reduction of mature B cells from the transitional 2 stage on, while B‐1 B cells are less affected. Moreover, deletion of NIK in the germinal centers decreases the numbers of germinal center B cells and impairs the ability of NIK‐deficient B cells to develop into class‐switched cells in vivo. This new mouse strain will be helpful for studying the role of NIK in different cell types of the body.

Collaboration


Dive into the Nadine Hövelmeyer's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge