Network


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

Hotspot


Dive into the research topics where Ninghai Wang is active.

Publication


Featured researches published by Ninghai Wang.


Nature Immunology | 2001

SAP controls T cell responses to virus and terminal differentiation of TH2 cells.

Chengbin Wu; Khuong B. Nguyen; Gary C. Pien; Ninghai Wang; Charles Gullo; Duncan Howie; Miriam Rodriguez Sosa; Matthew J. Edwards; Persephone Borrow; Abhay R. Satoskar; Arlene H. Sharpe; Christine A. Biron; Cox Terhorst

SH2D1A, which encodes signaling lymphocyte activation molecule (SLAM)–associated protein (SAP), is altered in patients with X-linked lymphoproliferative disease (XLP), a primary immunodeficiency. SAP-deficient mice infected with lymphocytic choriomeningitis virus had greatly increased numbers of CD8+ and CD4+ interferon-γ–producing spleen and liver cells compared to wild-type mice. The immune responses of SAP-deficient mice to infection with Leishmania major together with in vitro studies showed that activated SAP-deficient T cells had an impaired ability to differentiate into T helper 2 cells. The aberrant immune responses in SAP-deficient mice show that SAP controls several distinct key T cell signal transduction pathways, which explains in part the complexity of the XLP phenotypes.


Journal of Experimental Medicine | 2004

The Cell Surface Receptor SLAM Controls T Cell and Macrophage Functions

Ninghai Wang; Abhay R. Satoskar; William A. Faubion; Duncan Howie; Susumu Okamoto; Stefan Feske; Charles Gullo; Kareem Clarke; Miriam Rodriguez Sosa; Arlene H. Sharpe; Cox Terhorst

Signaling lymphocyte activation molecule (SLAM), a glycoprotein expressed on activated lymphocytes and antigen-presenting cells, has been shown to be a coregulator of antigen-driven T cell responses and is one of the two receptors for measles virus. Here we show that T cell receptor–induced interleukin (IL)-4 secretion by SLAM−/− CD4+ cells is down-regulated, whereas interferon γ production by CD4+ T cells is only slightly up-regulated. Although SLAM controls production of IL-12, tumor necrosis factor, and nitric oxide in response to lipopolysaccharide (LPS) by macrophages, SLAM does not regulate phagocytosis and responses to peptidoglycan or CpG. Thus, SLAM acts as a coreceptor that regulates signals transduced by the major LPS receptor Toll-like receptor 4 on the surface of mouse macrophages. A defective macrophage function resulted in an inability of SLAM−/− C57Bl/6 mice to remove the parasite Leishmania major. We conclude that the coreceptor SLAM plays a central role at the interface of acquired and innate immune responses.


Nature Immunology | 2010

SLAM is a microbial sensor that regulates bacterial phagosome functions in macrophages

Scott B. Berger; Xavier Romero; Chunyan Ma; Guoxing Wang; William A. Faubion; Gongxian Liao; Ewoud B. Compeer; Marton Keszei; Lucia E. Rameh; Ninghai Wang; Marianne Boes; Jose R. Regueiro; Hans-Christian Reinecker; Cox Terhorst

Phagocytosis is a pivotal process by which macrophages eliminate microorganisms after recognition by pathogen sensors. Here we unexpectedly found that the self ligand and cell surface receptor SLAM functioned not only as a costimulatory molecule but also as a microbial sensor that controlled the killing of Gram-negative bacteria by macrophages. SLAM regulated activity of the NADPH oxidase NOX2 complex and phagolysosomal maturation after entering the phagosome, following interaction with the bacterial outer membrane proteins OmpC and OmpF. SLAM recruited a complex containing the intracellular class III phosphatidylinositol kinase Vps34, its regulatory protein kinase Vps15 and the autophagy-associated molecule beclin-1 to the phagosome, which was responsible for inducing the accumulation of phosphatidylinositol-3-phosphate, a regulator of both NOX2 function and phagosomal or endosomal fusion. Thus, SLAM connects the Gram-negative bacterial phagosome to ubiquitous cellular machinery responsible for the control of bacterial killing.


Advances in Immunology | 2008

The SLAM and SAP gene families control innate and adaptive immune responses.

Silvia Calpe; Ninghai Wang; Xavier Romero; Scott B. Berger; Arpad Lanyi; Pablo Engel; Cox Terhorst

The nine SLAM-family genes, SLAMF1-9, a subfamily of the immunoglobulin superfamily, encode differentially expressed cell-surface receptors of hematopoietic cells. Engagement with their ligands, which are predominantly homotypic, leads to distinct signal transduction events, for instance those that occur in the T or NK cell immune synapse. Upon phosphorylation of one or more copies of a unique tyrosine-based signaling motif in their cytoplasmic tails, six of the SLAM receptors recruit the highly specific single SH2-domain adapters SLAM-associated protein (SAP), EAT-2A, and/or EAT-2B. These adapters in turn bind to the tyrosine kinase Fyn and/or other protein tyrosine kinases connecting the receptors to signal transduction networks. Individuals deficient in the SAP gene, SH2D1A, develop an immunodeficiency syndrome: X-linked lympho-proliferative disease. In addition to operating in the immune synapse, SLAM receptors initiate or partake in multiple effector functions of hematopoietic cells, for example, neutrophil and macrophage killing and platelet aggregation. Here we discuss the current understanding of the structure and function of these recently discovered receptors and adapter molecules in the regulation of adaptive and innate immune responses.


Journal of Experimental Medicine | 2011

A novel isoform of the Ly108 gene ameliorates murine lupus

Marton Keszei; Cynthia Detre; Svend T. Rietdijk; Pilar Muñoz; Xavier Romero; Scott B. Berger; Silvia Calpe; Gongxian Liao; Wilson Castro; Aimee Julien; Ying-Yu Wu; Dong-Mi Shin; Jaime Sancho; Mercedes Zubiaur; Herbert C. Morse; Laurence Morel; Pablo Engel; Ninghai Wang; Cox Terhorst

The expression of the new Ly108 isoform H1 weakens lupus-like disease of C57BL/6.Sle1b mice.


Immunogenetics | 2006

Identification and characterization of two related murine genes, Eat2a and Eat2b, encoding single SH2-domain adapters

Silvia Calpe; Erika Erdős; Gongxian Liao; Ninghai Wang; Svend T. Rietdijk; Maria Simarro; Beáta Scholtz; Jill M. Mooney; Chang Hoon Lee; Min Sun Shin; Éva Rajnavölgyi; John D. Schatzle; Herbert C. Morse; Cox Terhorst; Arpad Lanyi

Human EAT-2 (SH2D1B) and SLAM-associated protein (SAP) (SH2D1A) are single SH2-domain adapters, which bind to specific tyrosine residues in the cytoplasmic tail of six signaling lymphocytic activation molecule (SLAM) (SLAMF1)-related receptors. Here we report that, unlike in humans, the mouse and rat Eat2 genes are duplicated with an identical genomic organization. The coding regions of the mouse Eat2a and Eat2b genes share 91% identity at the nucleotide level and 84% at the protein level; similarly, segments of introns are highly conserved. Whereas expression of mouse Eat2a mRNA was detected in multiple tissues, Eat2b was only detectable in mouse natural killer cells, CD8+ T cells, and ovaries, suggesting a very restricted tissue expression of the latter. Both the EAT-2A and EAT-2B coimmunoprecipitated with mouse SLAM in transfected cells and augmented tyrosine phosphorylation of the cytoplasmic tail of SLAM. Both EAT-2A and EAT-2B bind to the Src-like kinases Fyn, Hck, Lyn, Lck, and Fgr, as determined by a yeast two-hybrid assay. However, unlike SAP, the EAT-2 proteins bind to their kinase domains and not to the SH3 domain of these kinases. Taken together, the data suggest that both EAT-2A and EAT-2B are adapters that recruit Src kinases to SLAM family receptors using a mechanism that is distinct from that of SAP.


Journal of Immunology | 2012

Cutting edge: protein phosphatase 2A confers susceptibility to autoimmune disease through an IL-17-dependent mechanism.

José C. Crispín; Sokratis A. Apostolidis; Florencia Rosetti; Marton Keszei; Ninghai Wang; Cox Terhorst; Tanya N. Mayadas; George C. Tsokos

The contribution of individual molecular aberrations to the pathogenesis of systemic lupus erythematosus (SLE), an autoimmune disease that affects multiple organs, is often difficult to evaluate because of the presence of abundant confounding factors. To assess the effect of increased expression of the phosphatase protein phosphatase 2A (PP2A) in T cells, as recorded in SLE patients, we generated a transgenic mouse that overexpresses the PP2Ac subunit in T cells. The transgenic mouse displays a heightened susceptibility to immune-mediated glomerulonephritis in the absence of other immune defects. CD4+ T cells produce increased amounts of IL-17 while the number of neutrophils in the peripheral blood is increased. IL-17 neutralization abrogated the development of glomerulonephritis. We conclude that increased PP2Ac expression participates in SLE pathogenesis by promoting inflammation through unchecked IL-17 production and facilitating the development of end-organ damage.


Journal of Immunology | 2011

Cutting Edge: An NK Cell-Independent Role for Slamf4 in Controlling Humoral Autoimmunity

Daniel R. Brown; Silvia Calpe; Marton Keszei; Ninghai Wang; Shannon McArdel; Cox Terhorst; Arlene H. Sharpe

Several genes within a syntenic region of human and mouse chromosome 1 are associated with predisposition to systemic lupus erythematosus. Analyses of lupus-prone congenic mice have pointed to an important role for the signaling lymphocyte activation molecule family (slamf)6 surface receptor in lupus pathogenesis. In this article, we demonstrate that a second member of the Slamf gene family, Slamf4 (Cd244), contributes to lupus-related autoimmunity. B6.Slamf4−/− mice spontaneously develop activated CD4 T cells and B cells and increased numbers of T follicular helper cells and a proportion develop autoantibodies to nuclear Ags. B6.Slamf4−/− mice also exhibit markedly increased autoantibody production in the B6.C-H-2bm12/KhEg → B6 transfer model of lupus. Although slamf4 function is best characterized in NK cells, the enhanced humoral autoimmunity of B6.Slamf4−/− mice is NK cell independent, as judged by depletion studies. Taken together, our findings reveal that slamf4 has an NK cell-independent negative regulatory role in the pathogenesis of lupus a normally non-autoimmune prone genetic background.


Blood | 2012

SAP expression in invariant NKT cells is required for cognate help to support B-cell responses

Cynthia Detre; Marton Keszei; Natividad Garrido-Mesa; Katalin Kis-Toth; Wilson Castro; Amma F. Agyemang; Natacha Veerapen; Gurdyal S. Besra; Michael C. Carroll; George C. Tsokos; Ninghai Wang; Elizabeth A. Leadbetter; Cox Terhorst

One of the manifestations of X-linked lymphoproliferative disease (XLP) is progressive agammaglobulinemia, caused by the absence of a functional signaling lymphocyte activation molecule (SLAM)-associated protein (SAP) in T, invariant natural killer T (NKT) cells and NK cells. Here we report that α-galactosylceramide (αGalCer) activated NKT cells positively regulate antibody responses to haptenated protein antigens at multiple checkpoints, including germinal center formation and affinity maturation. Whereas NKT cell-dependent B cell responses were absent in SAP(-/-).B6 mice that completely lack NKT cells, the small number of SAP-deficient NKT cells in SAP(-/-).BALB/c mice adjuvated antibody production, but not the germinal center reaction. To test the hypothesis that SAP-deficient NKT cells can facilitate humoral immunity, SAP was deleted after development in SAP(fl/fl).tgCreERT2.B6 mice. We find that NKT cell intrinsic expression of SAP is dispensable for noncognate helper functions, but is critical for providing cognate help to antigen-specific B cells. These results demonstrate that SLAM-family receptor-regulated cell-cell interactions are not limited to T-B cell conjugates. We conclude that in the absence of SAP, several routes of NKT cell-mediated antibody production are still accessible. The latter suggests that residual NKT cells in XLP patients might contribute to variations in dysgammaglobulinemia.


Journal of Biological Chemistry | 2012

Receptor Signaling Lymphocyte-activation Molecule Family 1 (Slamf1) Regulates Membrane Fusion and NADPH Oxidase 2 (NOX2) Activity by Recruiting a Beclin-1/Vps34/Ultraviolet Radiation Resistance-associated Gene (UVRAG) Complex

Chunyan Ma; Ninghai Wang; Cynthia Detre; Guoxing Wang; Michael O'Keeffe; Cox Terhorst

Background: The receptor Slamf1 functions as a microbial sensor regulating phagocytosis. Results: Slamf1 interacts with Vps34/Beclin-1/UVRAG complex in a wide range of cells. Conclusion: Slamf1 recruits a subset of ubiquitous autophagy-associated proteins, which is involved in membrane fusion and NOX2 regulation. Significance: These studies provide insight into the regulation of microbicidal function allowing for potential therapeutic discoveries. Phagocytosis is a pivotal process by which macrophages eliminate microorganisms upon recognition by pathogen sensors. Surprisingly, the self-ligand cell surface receptor Slamf1 functions not only as a co-stimulatory molecule but also as a microbial sensor of several Gram-negative bacteria. Upon entering the phagosome of macrophages Slamf1 induces production of phosphatidylinositol 3-phosphate, which positively regulates the activity of the NOX2 enzyme and phagolysosomal maturation. Here, we report that in Escherichia coli-containing phagosomes of mouse macrophages, Slamf1 interacts with the class III PI3K Vps34 in a complex with Beclin-1 and UVRAG. Upon phagocytosis of bacteria the NOX2 activity was reduced in macrophages isolated from Beclin-1+/− mice compared with wild-type mice. This Slamf1/Beclin-1/Vps34/UVRAG protein complex is formed in intracellular membrane compartments as it is found without inducing phagocytosis in macrophages, human chronic lymphocytic leukemia cells, and transfectant HEK293 cells. Elimination of its cytoplasmic tail abolished the interaction of Slamf1 with the complex, but deletion or mutation of the two ITAM motifs did not. Both the BD and CCD domains of Beclin-1 were required for efficient binding to Slamf1. Because Slamf1 did not interact with Atg14L or Rubicon, which can also form a complex with Vps34 and Beclin-1, we conclude that Slamf1 recruits a subset of Vps34-associated proteins, which is involved in membrane fusion and NOX2 regulation.

Collaboration


Dive into the Ninghai Wang's collaboration.

Top Co-Authors

Avatar

Cox Terhorst

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Gongxian Liao

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Marton Keszei

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Guoxing Wang

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Cynthia Detre

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michael O'Keeffe

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Silvia Calpe

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Wilson Castro

Beth Israel Deaconess Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge