Network


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

Hotspot


Dive into the research topics where Dietrich Conze is active.

Publication


Featured researches published by Dietrich Conze.


Immunity | 1998

DIFFERENTIATION OF CD4+ T CELLS TO TH1 CELLS REQUIRES MAP KINASE JNK2

Derek D. Yang; Dietrich Conze; Alan J. Whitmarsh; Tamera Barrett; Roger J. Davis; Mercedes Rincon; Richard A. Flavell

Precursor CD4+ T cells develop into effector Th1 and Th2 cells that play a central role in the immune response. We show that the JNK MAP kinase pathway is induced in Th1 but not in Th2 effector cells upon antigen stimulation. Further, the differentiation of precursor CD4+ T cells into effector Th1 but not Th2 cells is impaired in JNK2-deficient mice. The inability of IL-12 to differentiate JNK2-deficient CD4+ T cells fully into effector Th1 cells is caused by a defect in IFNgamma production during the early stages of differentiation. The addition of exogenous IFNgamma during differentiation restores IL-12-mediated Th1 polarization in the JNK2-deficient mice. The JNK MAP kinase signaling pathway, therefore, plays an important role in the balance of Th1 and Th2 immune responses.


Immunity | 2002

Salp15, an Ixodes scapularis Salivary Protein, Inhibits CD4+ T Cell Activation

Juan Anguita; Nandhini Ramamoorthi; Joppe W.R. Hovius; Subrata Das; Venetta Thomas; Rafal Persinski; Dietrich Conze; Philip W. Askenase; Mercedes Rincon; Fred S. Kantor; Erol Fikrig

Tick saliva has pleiotropic properties that facilitate persistence of the arthropod upon the host. We now describe a feeding-inducible protein in Ixodes scapularis saliva, Salp15, that inhibits CD4(+) T cell activation. The mechanism involves the repression of calcium fluxes triggered by TCR ligation and results in lower production of interleukin-2. Salp15 also inhibits the development of CD4(+) T cell-mediated immune responses in vivo, demonstrating the functional importance of this protein. Salp15 provides a molecular basis for understanding the immunosuppressive activity of I. scapularis saliva and vector-host interactions.


Molecular and Cellular Biology | 2005

Posttranscriptional downregulation of c-IAP2 by the ubiquitin protein ligase c-IAP1 in vivo.

Dietrich Conze; Lori Albert; David A. Ferrick; David V. Goeddel; Wen-Chen Yeh; Tak W. Mak; Jonathan D. Ashwell

ABSTRACT Inhibitor of apoptosis proteins (IAPs) c-IAP1 and c-IAP2 were identified as part of the tumor necrosis factor receptor 2 (TNFR2) signaling complex and have been implicated as intermediaries in tumor necrosis factor alpha signaling. Like all RING domain-containing IAPs, c-IAP1 and c-IAP2 have ubiquitin protein ligase (E3) activity. To explore the function of c-IAP1 in a physiologic setting, c-IAP1-deficient mice were generated by homologous gene recombination. These animals are viable and have no obvious sensitization to proapoptotic stimuli. Cells from c-IAP1−/− mice do, however, express markedly elevated levels of c-IAP2 protein in the absence of increased c-IAP2 mRNA. In contrast to reports implicating c-IAPs in the activation of NF-κB, resting and cytokine-induced NF-κB activation was not impaired in c-IAP1-deficient cells. Transient transfection studies with wild-type and E3-defective c-IAP1 revealed that c-IAP2 is a direct target for c-IAP1-mediated ubiquitination and subsequent degradation, which are potentiated by the adaptor function of TRAF2. Thus, the c-IAPs represent a pair of TNFR-associated ubiquitin protein ligases in which one regulates the expression of the other by a posttranscriptional and E3-dependent mechanism.


Molecular and Cellular Biology | 2008

Lys63-Linked Polyubiquitination of IRAK-1 Is Required for Interleukin-1 Receptor- and Toll-Like Receptor-Mediated NF-κB Activation

Dietrich Conze; Chuan-Jin Wu; James A. Thomas; Allison Landstrom; Jonathan D. Ashwell

ABSTRACT Stimulation through the interleukin-1 receptor (IL-1R) and some Toll-like receptors (TLRs) induces ubiquitination of TRAF6 and IRAK-1, signaling components required for NF-κB and mitogen-activated protein kinase activation. Here we show that although TRAF6 and IRAK-1 acquired Lys63 (K63)-linked polyubiquitin chains upon IL-1 stimulation, only ubiquitinated IRAK-1 bound NEMO, the regulatory subunit of IκB kinase (IKK). The sites of IRAK-1 ubiquitination were mapped to Lys134 and Lys180, and arginine substitution of these residues impaired IL-1R/TLR-mediated IRAK-1 ubiquitination, NEMO binding, and NF-κB activation. K63-linked ubiquitination of IRAK-1 required enzymatically active TRAF6, indicating that it is the physiologically relevant E3. Thus, K63-linked polyubiquitination of proximal signaling proteins is a common mechanism used by diverse innate immune receptors for recruiting IKK and activating NF-κB.


Journal of Experimental Medicine | 2002

c-Jun NH2-Terminal Kinase (JNK)1 and JNK2 Have Distinct Roles in CD8+ T Cell Activation

Dietrich Conze; Troy Krahl; Norman J. Kennedy; Linda K. Weiss; Joanne M. Lumsden; Patricia M. Hess; Richard A. Flavell; Graham Le Gros; Roger J. Davis; Mercedes Rincon

The c-Jun NH2-terminal kinase (JNK) signaling pathway is induced by cytokines and stress stimuli and is implicated in cell death and differentiation, but the specific function of this pathway depends on the cell type. Here we examined the role of JNK1 and JNK2 in CD8+ T cells. Unlike CD4+ T cells, the absence of JNK2 causes increased interleukin (IL)-2 production and proliferation of CD8+ T cells. In contrast, JNK1-deficient CD8+ T cells are unable to undergo antigen-stimulated expansion in vitro, even in the presence of exogenous IL-2. The hypoproliferation of these cells is associated with impaired IL-2 receptor α chain (CD25) gene and cell surface expression. The reduced level of nuclear activating protein 1 (AP-1) complexes in activated JNK1-deficient CD8+ T cells can account for the impaired IL-2 receptor α chain gene expression. Thus, JNK1 and JNK2 play different roles during CD8+ T cell activation and these roles differ from those in CD4+ T cells.


Molecular and Cellular Biology | 2000

Activation of p38 Mitogen-Activated Protein Kinase In Vivo Selectively Induces Apoptosis of CD8+ but Not CD4+ T Cells

Chris Merritt; Hervé Enslen; Nicole L. Diehl; Dietrich Conze; Roger J. Davis; Mercedes Rincon

ABSTRACT CD4+ and CD8+ T cells play specific roles during an immune response. Different molecular mechanisms could regulate the proliferation, death, and effector functions of these two subsets of T cells. The p38 mitogen-activated protein (MAP) kinase pathway is induced by cytokines and environmental stress and has been associated with cell death and cytokine expression. Here we report that activation of the p38 MAP kinase pathway in vivo causes a selective loss of CD8+ T cells due to the induction of apoptosis. In contrast, activation of p38 MAP kinase does not induce CD4+T-cell death. The apoptosis of CD8+ T cells is associated with decreased expression of the antiapoptotic protein Bcl-2. Regulation of the p38 MAP kinase pathway in T cells is therefore essential for the maintenance of CD4/CD8 homeostasis in the peripheral immune system. Unlike cell death, gamma interferon production is regulated by the p38 MAP kinase pathway in both CD4+ and CD8+ T cells. Thus, specific aspects of CD4+and CD8+ T-cell function are differentially controlled by the p38 MAP kinase signaling pathway.


Journal of Biological Chemistry | 2007

Tumor Necrosis Factor Receptor 2 Signaling Induces Selective c-IAP1-dependent ASK1 Ubiquitination and Terminates Mitogen-activated Protein Kinase Signaling

Yongge Zhao; Dietrich Conze; John A. Hanover; Jonathan D. Ashwell

TRAF2 and ASK1 play essential roles in tumor necrosis factor α (TNF-α)-induced mitogen-activated protein kinase signaling. Stimulation through TNF receptor 2 (TNFR2) leads to TRAF2 ubiquitination and subsequent proteasomal degradation. Here we show that TNFR2 signaling also leads to selective ASK1 ubiquitination and degradation in proteasomes. c-IAP1 was identified as the ubiquitin protein ligase for ASK1 ubiquitination, and studies with primary B cells from c-IAP1 knock-out animals revealed that c-IAP1 is required for TNFR2-induced TRAF2 and ASK1 degradation. Moreover, in the absence of c-IAP1 TNFR2-mediated p38 and JNK activation was prolonged. Thus, the ubiquitin protein ligase activity of c-IAP1 is responsible for regulating the duration of TNF signaling in primary cells expressing TNFR2.


Immunology and Cell Biology | 2000

Do T cells care about the mitogen-activated protein kinase signalling pathways?

Mercedes Rincon; Dietrich Conze; Linda Weiss; Nicole L. Diehl; Karen A. Fortner; Derek D. Yang; Richard A. Flavell; Hervé Enslen; Alan J. Whitmarsh; Roger J. Davis

Mitogen‐activated protein (MAP) kinases, which include the extracellular response kinases, p38 and c‐Jun amino terminal kinases (JNK), play a significant role in mediating signals triggered by cytokines, growth factors and environmental stress. The JNK and p38 MAP kinases have been involved in growth, differentiation and cell death in different cell types. In the present paper, we describe how the JNK and p38 MAP kinase signalling pathways are regulated and their role during thymocyte development and the activation and differentiation of T cells in the peripheral immune system. The results from these studies demonstrate that the JNK and p38 MAP kinase signalling pathways regulate different aspects of T‐cell mediated immune responses.


Molecular Immunology | 2000

Activation of p38 MAP kinase in T cells facilitates the immune response to the influenza virus

Dietrich Conze; Joanne M. Lumsden; Hervé Enslen; Roger J. Davis; Graham Le Gros; Mercedes Rincon

Activation of p38 MAP kinase in T cells leads to increased interferon-gamma production in CD4+ and CD8+ T cells, and the selective cell death of CD8+ T cells. To address the role of p38 MAP kinase activation in T cells during an in vivo immune response, we examined the response against the influenza virus in transgenic mice expressing a constitutively activated MKK6 (MKK6(Glu)), an upstream activator of p38 MAP kinase. Activated CD4+ T cells accumulate in the lung and mediastinal lymph node of both wild-type and MKK6(Glu) transgenic mice upon intranasal inoculation with the influenza virus. MKK6(Glu) CD8+ T cells, however, disappear rapidly from the mediastinal lymph node but accumulate in the lung tissue. We demonstrate that interleukin-6, a cytokine produced by lung epithelial cells, partially protects CD8+ T cells from the cell death induced by p38 MAP kinase activation. During the influenza infection in MKK6(Glu) transgenic mice, reduced virus titers were also observed despite a normal B-cell antibody response. These results indicate that the activation of p38 MAP kinase in T cells affects the in vivo antiviral immune response.


Infection and Immunity | 2006

Control of Borrelia burgdorferi-Specific CD4^+-T-Cell Effector Function by Interleukin-12- and T-Cell Receptor-Induced p38 Mitogen-Activated Protein Kinase Activity

Michael N. Hedrick; Chris M. Olson; Dietrich Conze; Tonya C. Bates; Mercedes Rincon; Juan Anguita

ABSTRACT Infection with Borrelia burgdorferi, the causative agent of Lyme disease, results in a Th1 response and proinflammatory cytokine production. Mice deficient for MKK3, an upstream activator of p38 mitogen-activated protein (MAP) kinase, develop a lower Th1 response and exhibit an impaired ability to produce proinflammatory cytokines upon infection with the spirochete. We investigated the contribution of p38 MAP kinase activity in gamma interferon (IFN-γ) production in CD4+ T cells in response to specific antigen through T-cell receptor (TCR)- and interleukin-12 (IL-12)-mediated signals. The specific inhibition of p38 MAP kinase in T cells and the administration of a pharmacological inhibitor of the kinase during the course of infection with the spirochete resulted in reduced levels of IFN-γ in the sera of infected mice. Our results also demonstrate that although p38 MAP kinase activity is not required for the differentiation of B. burgdorferi-specific CD4+ T cells, the production of IFN-γ by Th1 effector cells is regulated by the kinase. Both TCR engagement and IL-12 induced the production of the Th1 cytokine through the activation of the p38 MAP kinase pathway. Thus, the inhibition of this pathway in vitro resulted in decreased levels of IFN-γ during restimulation of B. burgdorferi-specific T cells in response to anti-CD3 and IL-12 stimulation. These results clarify the specific contribution of the p38 MAP kinase in the overall immune response to the spirochete and its role in the effector function of B. burgdorferi-specific T cells.

Collaboration


Dive into the Dietrich Conze's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Roger J. Davis

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Hervé Enslen

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Juan Anguita

University of Massachusetts Amherst

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jonathan D. Ashwell

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Tonya C. Bates

University of Massachusetts Amherst

View shared research outputs
Top Co-Authors

Avatar

Alan J. Whitmarsh

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Chris M. Olson

University of Massachusetts Amherst

View shared research outputs
Researchain Logo
Decentralizing Knowledge