Network


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

Hotspot


Dive into the research topics where Gopala Nishanth is active.

Publication


Featured researches published by Gopala Nishanth.


Journal of Immunology | 2011

Gp130-Dependent Astrocytic Survival Is Critical for the Control of Autoimmune Central Nervous System Inflammation

Fahad Haroon; Katrin Drögemüller; Ulrike Händel; Anna Brunn; Dirk Reinhold; Gopala Nishanth; Werner Mueller; C Trautwein; Matthias Ernst; Martina Deckert; Dirk Schlüter

Astrocytes are activated in experimental autoimmune encephalomyelitis (EAE) and have been suggested to either aggravate or ameliorate EAE. However, the mechanisms leading to an adverse or protective effect of astrocytes on the course of EAE are incompletely understood. To gain insight into the astrocyte-specific function of gp130 in EAE, we immunized mice lacking cell surface expression of gp130, the signal-transducing receptor for cytokines of the IL-6 family, with myelin oligodendrocyte glycoprotein35–55 peptide. These glial fibrillary acid protein (GFAP)-Cre gp130fl/fl mice developed clinically a significantly more severe EAE than control mice and succumbed to chronic EAE. Loss of astrocytic gp130 expression resulted in apoptosis of astrocytes in inflammatory lesions of GFAP-Cre gp130fl/fl mice, whereas gp130fl/fl control mice developed astrogliosis. Astrocyte loss of GFAP-Cre gp130fl/fl mice was paralleled by significantly larger areas of demyelination and significantly increased numbers of CD4 T cells in the CNS. Additionally, loss of astrocytes in GFAP-Cre gp130fl/fl mice resulted in a reduction of CNS regulatory Foxp3+ CD4 T cells and an increase of IL-17–, IFN-γ–, and TNF-producing CD4 as well as IFN-γ– and TNF-producing CD8 T cells, illustrating that astrocytes regulate the phenotypic composition of T cells. An analysis of mice deficient in either astrocytic gp130– Src homology region 2 domain-containing phosphatase 2/Ras/ERK or gp130–STAT1/3 signaling revealed that prevention of astrocyte apoptosis, restriction of demyelination, and T cell infiltration were dependent on the astrocytic gp130–Src homology region 2 domain-containing phosphatase 2/Ras/ERK, but not on the gp130–STAT1/3 pathway, further demonstrating that gp130-dependent astrocyte activation is crucial to ameliorate EAE.


Acta Neuropathologica | 2013

Astrocytic A20 ameliorates experimental autoimmune encephalomyelitis by inhibiting NF‑κB‑ and STAT1‑dependent chemokine production in astrocytes

Xu Wang; Martina Deckert; Nguyen Thi Xuan; Gopala Nishanth; Sissy Just; Ari Waisman; Michael Naumann; Dirk Schlüter

Single-nucleotide polymorphisms in the tumor necrosis factor, alpha-induced protein 3 gene, which encodes the ubiquitin-modifying protein A20, are linked to susceptibility to multiple sclerosis (MS), a demyelinating autoimmune disease of the central nervous system (CNS). Since it is unresolved how A20 regulates MS pathogenesis, we examined its function in a murine model of MS, namely experimental autoimmune encephalomyelitis (EAE). Deletion of A20 in neuroectodermal cells (astrocytes, neurons, and oligodendrocytes; Nestin-Cre A20fl/fl mice) or selectively in astrocytes (GFAP-Cre A20fl/fl mice) resulted in more severe EAE as compared to control animals. In Nestin-Cre A20fl/fl and GFAP-Cre A20fl/fl mice demyelination and recruitment of inflammatory leukocytes were increased as compared to A20fl/fl control mice. Importantly, numbers of encephalitogenic CD4+ T cells producing interferon (IFN)-γ, interleukin (IL)-17, and granulocyte–macrophage colony-stimulating factor (GM-CSF), respectively, as well as mRNA production of IFN-γ, IL-17, tumor necrosis factor (TNF), GM-CSF, IL-6, CXCL1, CCL2, and CXCL10 were significantly increased in spinal cords of Nestin-Cre A20fl/fl and GFAP-Cre A20fl/fl mice, respectively. Compared to A20-sufficient astrocytes, A20-deficient astrocytes displayed stronger activation of nuclear factor kappa-light-chain enhancer of activated B cells (NF-κB) in response to TNF, IL-17, and GM-CSF, and of signal transducer and activator of transcription 1 (STAT1) upon IFN-γ stimulation. Due to NF-κB and STAT1 hyperactivation, A20-deficient astrocytes produced significantly more chemokines in response to these key encephalitogenic cytokines of autoimmune CD4+ T cells resulting in an amplification of CD4+ T cell recruitment to the CNS. Thus, astrocytic A20 is an important inhibitor of autoimmune-mediated demyelination in the CNS.


Journal of Immunology | 2008

Protein Kinase C-θ Critically Regulates the Proliferation and Survival of Pathogen-Specific T Cells in Murine Listeriosis

Monika Sakowicz-Burkiewicz; Gopala Nishanth; Ulrike Helmuth; Katrin Drögemüller; Dirk H. Busch; Olaf Utermöhlen; Michael Naumann; Martina Deckert; Dirk Schlüter

Protein kinase C-θ (PKC-θ) is essential for the activation of T cells in autoimmune disorders, but not in viral infections. To study the role of PKC-θ in bacterial infections, PKC-θ−/− and wild-type mice were infected with Listeria monocytogenes (LM). In primary and secondary listeriosis, the numbers of LM-specific CD8 and CD4 T cells were drastically reduced in PKC-θ−/− mice, resulting in increased CFUs in spleen and liver of both PKC-θ−/− C57BL/6 and BALB/c mice. Furthermore, immunization with peptide-loaded wild-type dendritic cells induced LM-specific CD4 and CD8 T cells in wild-type but not in PKC-θ−/− mice. In listeriosis, transfer of wild-type T cells into PKC-θ−/− mice resulted in a normal control of Listeria, and, additionally, a selective expression of PKC-θ in LM-specific T cells was sufficient to drive a normal proliferation and survival of these T cells in LM-infected PKC-θ−/− recipients, illustrating a cell-autonomous function of PKC-θ in LM-specific T cells. Conversely, adoptively transferred PKC-θ−/− T cells were partially rescued from cell death and proliferated in LM-infected wild-type recipients, demonstrating that a PKC-θ deficiency of LM-specific T cells can be partially compensated for by a wild-type environment. Additionally, in vitro experiments showed that only the addition of IL-2, but not an inhibition of caspase-3, induced proliferation and prevented death of PKC-θ−/− T cells stimulated with LM-infected wild-type dendritic cells, further demonstrating that the impaired proliferation and survival of PKC-θ−/− T cells in listeriosis is not intrinsically fixed and can be experimentally improved.


Infection and Immunity | 2010

Protective Toxoplasma gondii-Specific T-Cell Responses Require T-Cell-Specific Expression of Protein Kinase C-Theta

Gopala Nishanth; Monika Sakowicz-Burkiewicz; Ulrike Händel; Stefanie Kliche; Xiaoqian Wang; Michael Naumann; Martina Deckert; Dirk Schlüter

ABSTRACT Protein kinase C-theta (PKC-θ) is important for the activation of autoreactive T cells but is thought to be of minor importance for T-cell responses in infectious diseases, suggesting that PKC-θ may be a target for the treatment of T-cell-mediated autoimmune diseases. To explore the function of PKC-θ in a chronic persisting infection in which T cells are crucial for pathogen control, we infected BALB/c PKC-θ−/− and PKC-θ+/+ wild-type mice with Toxoplasma gondii. The PKC-θ−/− mice succumbed to necrotizing Toxoplasma encephalitis due to an insufficient parasite control up to day 40, whereas the wild-type mice survived. The number of T. gondii-specific CD4 and CD8 T cells was significantly reduced in the PKC-θ−/− mice, resulting in the impaired production of protective cytokines (gamma interferon, tumor necrosis factor) and antiparasitic effector molecules (inducible nitric oxide, gamma interferon-induced GTPase) in the spleen and brain. In addition, Th2-cell numbers were reduced in infected the PKC-θ−/− mice, paralleled by the diminished GATA3 expression of PKC-θ−/− CD4 T cells and reduced T. gondii-specific IgG production in serum and cerebrospinal fluid. Western blot analysis of splenic CD4 and CD8 T cells revealed an impaired activation of the NF-κB, AP-1, and MAPK pathways in T. gondii-infected PKC-θ−/− mice. Adoptive transfer of wild-type CD4 plus CD8 T cells significantly protected PKC-θ−/− mice from death by increasing the numbers of gamma interferon-producing T. gondii-specific CD4 and CD8 T cells, illustrating a cell-autonomous, protective function of PKC-θ in T cells. These findings imply that PKC-θ inhibition drastically impairs T. gondii-specific T-cell responses with fatal consequences for intracerebral parasite control and survival.


Journal of Immunology | 2014

IFN-γ Regulates CD8+ Memory T Cell Differentiation and Survival in Response to Weak, but Not Strong, TCR Signals

Diana Stoycheva; Katrin Deiser; Lilian Stärck; Gopala Nishanth; Dirk Schlüter; Wolfgang Uckert; Thomas Schüler

In response to primary Ag contact, naive mouse CD8+ T cells undergo clonal expansion and differentiate into effector T cells. After pathogen clearance, most effector T cells die, and only a small number of memory T cell precursors (TMPs) survive to form a pool of long-lived memory T cells (TMs). Although high- and low-affinity CD8+ T cell clones are recruited into the primary response, the TM pool consists mainly of high-affinity clones. It remains unclear whether the more efficient expansion of high-affinity clones and/or cell-intrinsic processes exclude low-affinity T cells from the TM pool. In this article, we show that the lack of IFN-γR signaling in CD8+ T cells promotes TM formation in response to weak, but not strong, TCR agonists. The IFN-γ–sensitive accumulation of TMs correlates with reduced mammalian target of rapamycin activation and the accumulation of long-lived CD62LhiBcl-2hiEomeshi TMPs. Reconstitution of mammalian target of rapamycin or IFN-γR signaling is sufficient to block this process. Hence, our data suggest that IFN-γR signaling actively blocks the formation of TMPs responding to weak TCR agonists, thereby promoting the accumulation of high-affinity T cells finally dominating the TM pool.


European Journal of Immunology | 2015

A20 expression in dendritic cells protects mice from LPS‐induced mortality

Nguyen Thi Xuan; Xu Wang; Gopala Nishanth; Ari Waisman; Katrin Borucki; Berend Isermann; Michael Naumann; Martina Deckert; Dirk Schlüter

DCs contribute to immune homeostasis under physiological conditions and regulate the immune activation during infection. The deubiquitinase A20 inhibits the activation of NF‐κB‐dependent immune reactions, and prevents the hyperactivation of DCs under steady‐state conditions. However, the role of DC‐specific A20 under pathological conditions is unknown. Here, we demonstrate that upon injection of low‐dose LPS, mice with DC‐specific A20 deletion (CD11c‐Cre A20fl/fl) died within 6 h, whereas A20fl/fl controls survived. LPS‐induced mortality in CD11c‐Cre A20fl/fl mice was characterized by increased serum levels of IL‐2, IL‐10, IL‐12, IFN‐γ, and TNF. Upon LPS stimulation, the activation of NF‐κB and ERK‐NFATc3 pathways were enhanced in A20‐deficient DCs, resulting in an increased production of IL‐2, IL‐12, and TNF both in vitro and in vivo. Targeted inhibition of ERK in A20‐deficient DCs abolished the increased production of IL‐2. A20‐deficient DCs failed to induce LPS tolerance, which was independent of T cells and the intestinal flora, since T‐cell depletion and decolonization of CD11c‐Cre A20fl/fl mice could not prevent death of LPS‐challenged CD11c‐Cre A20fl/fl mice. In conclusion, these findings show that DC‐specific A20 preserves immune homeostasis in steady‐state conditions and is also required for LPS tolerance.


PLOS Pathogens | 2013

CYLD Enhances Severe Listeriosis by Impairing IL-6/STAT3-Dependent Fibrin Production

Gopala Nishanth; Martina Deckert; Katharina Wex; Ramin Massoumi; Katrin Schweitzer; Michael Naumann; Dirk Schlüter

The facultative intracellular bacterium Listeria monocytogenes (Lm) may cause severe infection in humans and livestock. Control of acute listeriosis is primarily dependent on innate immune responses, which are strongly regulated by NF-κB, and tissue protective factors including fibrin. However, molecular pathways connecting NF-κB and fibrin production are poorly described. Here, we investigated whether the deubiquitinating enzyme CYLD, which is an inhibitor of NF-κB-dependent immune responses, regulated these protective host responses in murine listeriosis. Upon high dose systemic infection, all C57BL/6 Cyld−/− mice survived, whereas 100% of wildtype mice succumbed due to severe liver pathology with impaired pathogen control and hemorrhage within 6 days. Upon in vitro infection with Lm, CYLD reduced NF-κB-dependent production of reactive oxygen species, interleukin (IL)-6 secretion, and control of bacteria in macrophages. Furthermore, Western blot analyses showed that CYLD impaired STAT3-dependent fibrin production in cultivated hepatocytes. Immunoprecipitation experiments revealed that CYLD interacted with STAT3 in the cytoplasm and strongly reduced K63-ubiquitination of STAT3 in IL-6 stimulated hepatocytes. In addition, CYLD diminished IL-6-induced STAT3 activity by reducing nuclear accumulation of phosphorylated STAT3. In vivo, CYLD also reduced hepatic STAT3 K63-ubiquitination and activation, NF-κB activation, IL-6 and NOX2 mRNA production as well as fibrin production in murine listeriosis. In vivo neutralization of IL-6 by anti-IL-6 antibody, STAT3 by siRNA, and fibrin by warfarin treatment, respectively, demonstrated that IL-6-induced, STAT3-mediated fibrin production significantly contributed to protection in Cyld−/− mice. In addition, in vivo Cyld siRNA treatment increased STAT3 phosphorylation, fibrin production, pathogen control and survival of Lm-infected WT mice illustrating that therapeutic inhibition of CYLD augments the protective NF-κB/IL-6/STAT3 pathway and fibrin production.


European Journal of Immunology | 2014

CTLA-4 (CD152) enhances the Tc17 differentiation program.

Jonas Pick; Aditya Arra; Holger Lingel; J. Kolja Hegel; Magdalena Huber; Gopala Nishanth; Gerhard Jorch; Klaus-Dieter Fischer; Dirk Schlüter; Kerry Tedford; Monika C. Brunner-Weinzierl

Although CD8+ T cells that produce IL‐17 (Tc17 cells) have been linked to host defense, Tc17 cells show reduced cytotoxic activity, which is the characteristic function of CD8+ T cells. Here, we show that CTLA‐4 enhances the frequency of IL‐17 in CD8+ T cells, indicating that CTLA‐4 (CD152) specifically promotes Tc17 differentiation. Simultaneous stimulation of CTLA‐4+/+ and CTLA‐4−/− T cells in cocultures and agonistic CTLA‐4 stimulation unambiguously revealed a cell‐intrinsic mechanism for IL‐17 control by CTLA‐4. The quality of CTLA‐4‐induced Tc17 cells was tested in vivo, utilizing infection with the facultative intracellular bacterium Listeria monocytogenes (LM). Unlike CTLA‐4+/+ Tc17 cells, CTLA‐4−/− were nearly as efficient as Tc1 CTLA‐4+/+ cells in LM clearance. Additionally, adoptively transferred CTLA‐4−/− Tc17 cells expressed granzyme B after rechallenge, and produced Tc1 cytokines such as IFN‐γ and TNF‐α, which strongly correlate with bacterial clearance. CTLA‐4+/+ Tc17 cells demonstrated a high‐quality Tc17 differentiation program ex vivo, which was also evident in isolated IL‐17‐secreting Tc17 cells, with CTLA‐4‐mediated enhanced upregulation of Tc17‐related molecules such as IL‐17A, RORγt, and IRF‐4. Our results show that CTLA‐4 promotes Tc17 differentiation that results in robust Tc17 responses. Its inactivation might therefore represent a central therapeutic target to enhance clearance of infection.


Scientific Reports | 2016

A20 Curtails Primary but Augments Secondary CD8(+) T Cell Responses in Intracellular Bacterial Infection.

Sissy Just; Gopala Nishanth; Jörn H. Buchbinder; Xu Wang; Michael Naumann; Inna N. Lavrik; Dirk Schlüter

The ubiquitin-modifying enzyme A20, an important negative feedback regulator of NF-κB, impairs the expansion of tumor-specific CD8+ T cells but augments the proliferation of autoimmune CD4+ T cells. To study the T cell-specific function of A20 in bacterial infection, we infected T cell-specific A20 knockout (CD4-Cre A20fl/fl) and control mice with Listeria monocytogenes. A20-deficient pathogen-specific CD8+ T cells expanded stronger resulting in improved pathogen control at day 7 p.i. Imaging flow cytometry revealed that A20-deficient Listeria-specific CD8+ T cells underwent increased apoptosis and necroptosis resulting in reduced numbers of memory CD8+ T cells. In contrast, the primary CD4+ T cell response was A20-independent. Upon secondary infection, the increase and function of pathogen-specific CD8+ T cells, as well as pathogen control were significantly impaired in CD4-Cre A20fl/fl mice. In vitro, apoptosis and necroptosis of Listeria-specific A20-deficient CD8+ T cells were strongly induced as demonstrated by increased caspase-3/7 activity, RIPK1/RIPK3 complex formation and more morphologically apoptotic and necroptotic CD8+ T cells. In vitro, A20 limited CD95L and TNF-induced caspase3/7 activation. In conclusion, T cell-specific A20 limited the expansion but reduced apoptosis and necroptosis of Listeria-specific CD8+ T cells, resulting in an impaired pathogen control in primary but improved clearance in secondary infection.


Frontiers in Immunology | 2017

The Deubiquitinating Enzyme Cylindromatosis Dampens CD8+ T Cell Responses and Is a Critical Factor for Experimental Cerebral Malaria and Blood–Brain Barrier Damage

Ursula Schmid; Werner Stenzel; Josephin Koschel; Maria Raptaki; Xu Wang; Michael Naumann; Kai Matuschewski; Dirk Schlüter; Gopala Nishanth

Cerebral malaria is a severe complication of human malaria and may lead to death of Plasmodium falciparum-infected individuals. Cerebral malaria is associated with sequestration of parasitized red blood cells within the cerebral microvasculature resulting in damage of the blood–brain barrier and brain pathology. Although CD8+ T cells have been implicated in the development of murine experimental cerebral malaria (ECM), several other studies have shown that CD8+ T cells confer protection against blood-stage infections. Since the role of host deubiquitinating enzymes (DUBs) in malaria is yet unknown, we investigated how the DUB cylindromatosis (CYLD), an important inhibitor of several cellular signaling pathways, influences the outcome of ECM. Upon infection with Plasmodium berghei ANKA (PbA) sporozoites or PbA-infected red blood cells, at least 90% of Cyld−/− mice survived the infection, whereas all congenic C57BL/6 mice displayed signatures of ECM, impaired parasite control, and disruption of the blood–brain barrier integrity. Cyld deficiency prevented brain pathology, including hemorrhagic lesions, enhanced activation of astrocytes and microglia, infiltration of CD8+ T cells, and apoptosis of endothelial cells. Furthermore, PbA-specific CD8+ T cell responses were augmented in the blood of Cyld−/− mice with increased production of interferon-γ and granzyme B and elevated activation of protein kinase C-θ and nuclear factor “kappa light-chain enhancer” of activated B cells. Importantly, accumulation of CD8+ T cells in the brain of Cyld−/− mice was significantly reduced compared to C57BL/6 mice. Bone marrow chimera experiments showed that the absence of ECM signatures in infected Cyld−/− mice could be attributed to hematopoietic and radioresistant parenchymal cells, most likely endothelial cells that did not undergo apoptosis. Together, we were able to show that host deubiqutinating enzymes play an important role in ECM and that CYLD promotes ECM supporting it as a potential therapeutic target for adjunct therapy to prevent cerebral complications of severe malaria.

Collaboration


Dive into the Gopala Nishanth's collaboration.

Top Co-Authors

Avatar

Dirk Schlüter

Otto-von-Guericke University Magdeburg

View shared research outputs
Top Co-Authors

Avatar

Michael Naumann

Otto-von-Guericke University Magdeburg

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Xu Wang

Otto-von-Guericke University Magdeburg

View shared research outputs
Top Co-Authors

Avatar

Sissy Just

Otto-von-Guericke University Magdeburg

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Katharina Wex

Otto-von-Guericke University Magdeburg

View shared research outputs
Top Co-Authors

Avatar

Katrin Drögemüller

Otto-von-Guericke University Magdeburg

View shared research outputs
Top Co-Authors

Avatar

Ulrike Händel

Otto-von-Guericke University Magdeburg

View shared research outputs
Top Co-Authors

Avatar

Ursula Schmid

Otto-von-Guericke University Magdeburg

View shared research outputs
Researchain Logo
Decentralizing Knowledge