Network


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

Hotspot


Dive into the research topics where Beinan Wang is active.

Publication


Featured researches published by Beinan Wang.


Cellular Microbiology | 2006

Integrin‐linked kinase is an essential link between integrins and uptake of bacterial pathogens by epithelial cells

Beinan Wang; Ryan S. Yurecko; Shoukat Dedhar; P. Patrick Cleary

Entry of Streptococcus pyogenes or group A streptococcus (GAS) into host cells is mediated by fibronectin bound to surface proteins, M1 or PrtF1, forming a bridge to α5β1 integrins. This interaction leads to cytoskeletal rearrangement and uptake of streptococci. We postulated that integrin‐linked kinase (ILK), which directly associates with integrins, is the universal link between integrins and several bacterial pathogens. We showed that inhibition of ILK expression by siRNA silencing, or ILK kinase activity by chemical inhibitors or expression of a dominant negative form of ILK reduced M1‐mediated invasion of epithelial cells up to 80%. To evaluate the ILK requirement for PrtF1‐mediated GAS invasion, a M1–PrtF1+ recombinant strain within the M1 background was constructed. Inhibition of ILK kinase activity also significantly reduced invasion of epithelial cells by this recombinant and wild‐type strain JRS4 that expresses PrtF1. In addition, impaired ILK kinase activity results in significant reduction of integrin‐dependent invasion mediated by invasins of two other important pathogens, Staphylococcus aureus and Yersinia spp. This study suggests that bacterial pathogens evolved different molecules and strategies to exploit the host integrin signalling pathway for their survival.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Induction of TGF-β1 and TGF-β1–dependent predominant Th17 differentiation by group A streptococcal infection

Beinan Wang; Thamotharampillai Dileepan; Sarah Briscoe; Kendra A. Hyland; Johnthomas Kang; Alexander Khoruts; P. Patrick Cleary

Recurrent group A Streptococcus (GAS) tonsillitis and associated autoimmune diseases indicate that the immune response to this organism can be ineffective and pathological. TGF-β1 is recognized as an essential signal for generation of regulatory T cells (Tregs) and T helper (Th) 17 cells. Here, the impact of TGF-β1 induction on the T-cell response in mouse nasal-associated lymphoid tissue (NALT) following intranasal (i.n.) infections is investigated. ELISA and TGF-β1-luciferase reporter assays indicated that persistent infection of mouse NALT with GAS sets the stage for TGF-β1 and IL-6 production, signals required for promotion of a Th17 immune response. As predicted, IL-17, the Th17 signature cytokine, was induced in a TGF-β1 signaling-dependent manner in single-cell suspensions of both human tonsils and NALT. Intracellular cytokine staining and flow cytometry demonstrated that CD4+ IL-17+ T cells are the dominant T cells induced in NALT by i.n. infections. Moreover, naive mice acquired the potential to clear GAS by adoptive transfer of CD4+ T cells from immunized IL-17A+/+ mice but not cells from IL-17A−/− mice. These experiments link specific induction of TGF-β1 by a bacterial infection to an in vivo Th17 immune response and show that this cellular response is sufficient for protection against GAS. The association of a Th17 response with GAS infection reveals a potential mechanism for destructive autoimmune responses in humans.


Proceedings of the National Academy of Sciences of the United States of America | 2015

Influenza viral neuraminidase primes bacterial coinfection through TGF-β–mediated expression of host cell receptors

Ning Li; Aihui Ren; Xiaoshuang Wang; Xin Fan; Yong Zhao; George F. Gao; P. Patrick Cleary; Beinan Wang

Significance Pneumonia caused by bacterial coinfection with influenza virus is the leading cause of mortality in influenza pandemics. TGF-β is known to be activated by influenza virus. In this study we demonstrated that cellular adhesins for bacteria, such as fibronectin and α5 integrin, are up-regulated in influenza viral infection. Inhibition of TGF-β impeded the up-regulation of these cellular adhesins and also influenza viral-enhanced bacterial adherence. In addition, we found that influenza viral-promoted bacterial adherence was dependent on bacterial fibronectin-binding protein. The results indicate that up-regulated expression of cellular adhesins by TGF-β, which is activated in influenza viral infection, increases host susceptibility to bacterial coinfection and suggest that TGF-β and host adhesion molecules are potential pharmaceutical targets for prevention of coinfection. Influenza infection predisposes the host to secondary bacterial pneumonia, which is a major cause of mortality during influenza epidemics. The molecular mechanisms underlying the bacterial coinfection remain elusive. Neuraminidase (NA) of influenza A virus (IAV) enhances bacterial adherence and also activates TGF-β. Because TGF-β can up-regulate host adhesion molecules such as fibronectin and integrins for bacterial binding, we hypothesized that activated TGF-β during IAV infection contributes to secondary bacterial infection by up-regulating these host adhesion molecules. Flow cytometric analyses of a human lung epithelial cell line indicated that the expression of fibronectin and α5 integrin was up-regulated after IAV infection or treatment with recombinant NA and was reversed through the inhibition of TGF-β signaling. IAV-promoted adherence of group A Streptococcus (GAS) and other coinfective pathogens that require fibronectin for binding was prevented significantly by the inhibition of TGF-β. However, IAV did not promote the adherence of Lactococcus lactis unless this bacterium expressed the fibronectin-binding protein of GAS. Mouse experiments showed that IAV infection enhanced GAS colonization in the lungs of wild-type animals but not in the lungs of mice deficient in TGF-β signaling. Taken together, these results reveal a previously unrecognized mechanism: IAV NA enhances the expression of cellular adhesins through the activation of TGF-β, leading to increased bacterial loading in the lungs. Our results suggest that TGF-β and cellular adhesins may be potential pharmaceutical targets for the prevention of coinfection.


Cellular Microbiology | 2005

Engagement of CD46 and α5β1 integrin by group A streptococci is required for efficient invasion of epithelial cells

Myrna S. Rezcallah; Kimberly Hodges; Darcy B. Gill; John P. Atkinson; Beinan Wang; P. Patrick Cleary

Membrane cofactor protein (MCP or CD46), a widely distributed complement regulatory human protein, is a cell surface receptor for many pathogens including group A streptococci (GAS). The surface M protein of GAS binds CD46 and mediates GAS adherence to keratinocytes. In the present study, we studied the role of CD46 in GAS invasion of human lung epithelial cells, A549. Anti‐CD46 antibody which specifically blocks the domain to which M protein binds inhibited adherence to and invasion of A549 cells by GAS. Moreover, downregulation of CD46 expression on A549 by RNA interference resulted in reduced invasion of these cells by GAS. A mutant form of CD46 with a deletion in the cytoplasmic domain was overexpressed in A549 cells, which resulted in partial inhibition of invasion. This indicates that the cytoplasmic tail is required for CD46 to promote invasion by GAS. Invasion assays with Lactococcus lactis that express M protein demonstrated the dependence of CD46‐promoted invasion on interaction with M protein. In addition, CD46‐mediated invasion was also found to be dependent on the extracellular matrix protein fibronectin.


Infection and Immunity | 2003

Up-regulation of interleukin-8 by novel small cytoplasmic molecules of nontypeable Haemophilus influenzae via p38 and extracellular signal-regulated kinase pathways.

Beinan Wang; P. Patrick Cleary; Haidong Xu; Jian Dong Li

ABSTRACT Nontypeable Haemophilus influenzae (NTHI) is an important etiological agent of otitis media (OM) and of exacerbated chronic obstructive pulmonary diseases (COPD). Inflammation is a hallmark of both diseases. Interleukin-8 (IL-8), one of the important inflammatory mediators, is induced by NTHI and may play a significant role in the pathogenesis of these diseases. Our studies demonstrated that a soluble cytoplasmic fraction (SCF) from NTHI induced much greater IL-8 expression by human epithelial cells than did NTHI lipooligosaccharides and envelope proteins. The IL-8-inducing activity was associated with molecules of ≤3 kDa from SCF and was peptidase and lipase sensitive, suggesting that small lipopeptides are responsible for the strong IL-8 induction. Moreover, multiple intracellular signaling pathways were activated in response to cytoplasmic molecules. The results indicated that the p38 mitogen-activated protein kinase (MAPK) and Src-dependent Raf-1-Mek1/2-extracellular signal-regulated kinase mitogen-activated protein kinase (ERK MAPK) pathways are required for NTHI-induced IL-8 production. In contrast, the phosphatidylinositol 3-kinase (PI3K)-Akt pathway did not affect IL-8 expression, although this pathway was concomitantly activated upon exposure to NTHI SCF. The PI3K-Akt pathway was also directly activated by IL-8 and significantly inhibited by an antagonist of IL-8 receptors during NTHI stimulation. These results indicated that the PI3K-Akt pathway is activated in response to IL-8 that is induced by NTHI and may lead to other important epithelial cell responses. This work provides insight into essential molecular and cellular events that may impact on the pathogenesis of OM and COPD and identifies rational targets for anti-inflammatory intervention.


Cellular Microbiology | 2007

Paxillin phosphorylation: bifurcation point downstream of integrin‐linked kinase (ILK) in streptococcal invasion

Beinan Wang; Shaoying Li; Shoukat Dedhar; P. Patrick Cleary

Efficient group A streptococcus (GAS) invasion of mammalian cells requires fibronectin (Fn) binding proteins, such as M1 and PrtF1/SfbI, that bridge bacteria to integrins and activate cellular signalling for ingestion. Previous studies of GAS invasion, mediated by both proteins, suggest a common signalling pathway. However, distinct cellular morphological changes at the port of bacterial entry suggest that different signals are also induced. Here we report that paxillin is phosphorylated in response to Fn‐bound GAS that express either M1 or PrtF1/SfbI protein, but is not phosphorylated in response to a mutant deficient in both proteins. Inhibition of paxillin phosphorylation by a tyrosine kinase inhibitor, PP2, or by expression of a dominant negative form of paxillin significantly reduced invasion by M1+ but did not affect ingestion of PrtF1/SfbI+ strains. In contrast, another tyrosine inhibitor, genistein, did not significantly prevent paxillin phosphorylation and had no effect on ingestion of the M1+ strain, but reduced PrtF1/SfbI‐mediated entry. This suggests that paxillin phosphorylation is induced by both proteins but only required for M1‐mediated invasion. A bifurcation point, downstream of integrin‐linked kinase (ILK) and phosphoinositide 3‐kinase, likely accounts for the distinct morphological changes. Furthermore, ILK activity is indispensable for M1‐induced paxillin recruitment and phosphorylation.


Infection and Immunity | 2007

Protein F1 and Streptococcus pyogenes Resistance to Phagocytosis

Kendra A. Hyland; Beinan Wang; P. Patrick Cleary

ABSTRACT Streptococcus pyogenes is a major cause of pharyngitis in humans and encodes several fibronectin-binding proteins. M protein and protein F1 (PrtF1/SfbI) are differentially regulated by CO2 and O2, respectively, and both mediate the invasion of epithelial cells. This study examined whether PrtF1/SfbI shares other properties with M protein. Expression of the PrtF1/SfbI protein by an M-negative mutant conferred resistance to phagocytosis and partial inhibition of C3 deposition on the S. pyogenes surface.


Fems Immunology and Medical Microbiology | 2009

The early interferon response of nasal-associated lymphoid tissue to Streptococcus pyogenes infection

Kendra A. Hyland; Robert Brennan; Stephen B. Olmsted; Eduardo Rojas; Ellen Murphy; Beinan Wang; P. Patrick Cleary

Streptococcus pyogenes is a major causative agent of tonsillitis or pharyngitis in children. Streptococcus pyogenes can persist in tonsils, and one-third of children treated with antibiotics continue to shed streptococci and have recurrent infections. Mouse nasal-associated lymphoid tissue (NALT) is functionally analogous to human oropharyngeal lymphoid tissues, and serves as a model for characterization of the mucosal innate immune response to S. pyogenes. Wild-type S. pyogenes induces transcription of both type I and interferon-gamma (IFN-gamma)-responsive genes, proinflammatory genes and acute-phase response proteins 24 h after intranasal infection. Invasion of NALT and the induction of the interferon response were not dependent on expression of antiphagocytic M protein. Intranasal infection induces a substantial influx of neutrophils into NALT at 24 h, which declines by 48 h after infection. Infection of IFN-gamma(-/-) [IFN-gamma knock-out mouse (GKO)] C57BL/6 mice with wild-type S. pyogenes resulted in local dissemination of bacteria to draining lymph nodes (LN), but did not lead to systemic infection by 48 h after infection. Infected GKO mice had an increased influx of neutrophils into NALT compared with immunocompetent mice. Thus, IFN-gamma-induced responses are required to prevent local dissemination of streptococci to the draining LN.


PLOS ONE | 2014

Sortase A Induces Th17-Mediated and Antibody-Independent Immunity to Heterologous Serotypes of Group A Streptococci

Xin Fan; Xiaoshuang Wang; Ning Li; Honglian Cui; Bin Gao; Paul Patrick Cleary; Beinan Wang

Group A streptococci (GAS) are associated with a variety of mucosal and invasive human infections. Recurrent infections by highly heterologous serotypes indicate that cross-serotype immunity is critical for prevention of GAS infections; however, mechanisms underlying serotype-independent protection are poorly understood. Here we report that intranasal vaccination of mice with Sortase A (SrtA), a conserved cell wall bound protein, reduced colonization of nasal-associated lymphoid tissue (NALT) by heterologous serotypes of GAS. Vaccination significantly increased CD4+ IL-17A+ cells in NALT and depletion of IL-17A by neutralizing antibody prevented GAS clearance from NALT which was dependent on immunization with SrtA. Vaccination also induced high levels of SrtA-specific antibodies; however, immunized, B cell-deficient mice cleared streptococcal challenges as efficiently as wild type mice, indicating that the cross-serotype protection is Th17-biased and antibody-independent. Furthermore, efficient GAS clearance from NALT was associated with a rapid neutrophil influx into NALT of immunized mice. These results suggest that serotype independent immune protection against GAS mucosal infection can be achieved by intranasal vaccination with SrtA and enhanced neutrophil function is critical for anti-GAS defense and might be a target for prevention of GAS infections.


Proceedings of the National Academy of Sciences of the United States of America | 2006

Streptococcal modulation of cellular invasion via TGF-β1 signaling

Beinan Wang; Shaoying Li; Peter J. Southern; P. Patrick Cleary

Collaboration


Dive into the Beinan Wang's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ning Li

China Agricultural University

View shared research outputs
Top Co-Authors

Avatar

Xiaoshuang Wang

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Xin Fan

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Shaoying Li

University of Minnesota

View shared research outputs
Top Co-Authors

Avatar

Shoukat Dedhar

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Bin Gao

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

George F. Gao

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Yong Zhao

Chinese Academy of Sciences

View shared research outputs
Researchain Logo
Decentralizing Knowledge