Network


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

Hotspot


Dive into the research topics where Zhi Q. Yao is active.

Publication


Featured researches published by Zhi Q. Yao.


Journal of Immunology | 2012

Tim-3 Pathway Controls Regulatory and Effector T Cell Balance during Hepatitis C Virus Infection

Jonathan P. Moorman; Jia M. Wang; Ying Zhang; Xiao J. Ji; Cheng J. Ma; Xiao Y. Wu; Zhan S. Jia; Ke S. Wang; Zhi Q. Yao

Hepatitis C virus (HCV) is remarkable at disrupting human immunity to establish chronic infection. Upregulation of inhibitory signaling pathways (such as T cell Ig and mucin domain protein-3 [Tim-3]) and accumulation of regulatory T cells (Tregs) play pivotal roles in suppressing antiviral effector T cell (Teff) responses that are essential for viral clearance. Although the Tim-3 pathway has been shown to negatively regulate Teffs, its role in regulating Foxp3+ Tregs is poorly explored. In this study, we investigated whether and how the Tim-3 pathway alters Foxp3+ Treg development and function in patients with chronic HCV infection. We found that Tim-3 was upregulated, not only on IL-2–producing CD4+CD25+Foxp3− Teffs, but also on CD4+CD25+Foxp3+ Tregs, which accumulate in the peripheral blood of chronically HCV-infected individuals when compared with healthy subjects. Tim-3 expression on Foxp3+ Tregs positively correlated with expression of the proliferation marker Ki67 on Tregs, but it was inversely associated with proliferation of IL-2–producing Teffs. Moreover, Foxp3+ Tregs were found to be more resistant to, and Foxp3− Teffs more sensitive to, TCR activation-induced cell apoptosis, which was reversible by blocking Tim-3 signaling. Consistent with its role in T cell proliferation and apoptosis, blockade of Tim-3 on CD4+CD25+ T cells promoted expansion of Teffs more substantially than Tregs through improving STAT-5 signaling, thus correcting the imbalance of Foxp3+ Tregs/Foxp3− Teffs that was induced by HCV infection. Taken together, the Tim-3 pathway appears to control Treg and Teff balance through altering cell proliferation and apoptosis during HCV infection.


PLOS ONE | 2011

Tim-3 Negatively Regulates IL-12 Expression by Monocytes in HCV Infection

Ying Zhang; Cheng J. Ma; Jia M. Wang; Xiao J. Ji; Xiao Y. Wu; Zhan S. Jia; Jonathan P. Moorman; Zhi Q. Yao

T cell immunoglobulin and mucin domain-containing protein 3 (Tim-3) is a newly identified negative immunomodulator that is up-regulated on dysfunctional T cells during viral infections. The expression and function of Tim-3 on human innate immune responses during HCV infection, however, remains poorly characterized. In this study, we report that Tim-3 is constitutively expressed on human resting CD14+ monocyte/macrophages (M/MØ) and functions as a cap to block IL-12, a key pro-inflammatory cytokine linking innate and adaptive immune responses. Tim-3 expression is significantly reduced and IL-12 expression increased upon stimulation with Toll-like receptor 4 (TLR4) ligand - lipopolysaccharide (LPS) and TLR7/8 ligand - R848. Notably, Tim-3 is over-expressed on un-stimulated as well as TLR-stimulated M/MØ, which is inversely associated with the diminished IL-12 expression in chronically HCV-infected individuals when compared to healthy subjects. Up-regulation of Tim-3 and inhibition of IL-12 are also observed in M/MØ incubated with HCV-expressing hepatocytes, as well as in primary M/MØ or monocytic THP-1 cells incubated with HCV core protein, an effect that mimics the function of complement C1q and is reversible by blocking the HCV core/gC1qR interaction. Importantly, blockade of Tim-3 signaling significantly rescues HCV-mediated inhibition of IL-12, which is primarily expressed by Tim-3 negative M/MØ. Tim-3 blockade reduces HCV core-mediated expression of the negative immunoregulators PD-1 and SOCS-1 and increases STAT-1 phosphorylation. Conversely, blocking PD-1 or silencing SOCS-1 gene expression also decreases Tim-3 expression and enhances IL-12 secretion and STAT-1 phosphorylation. These findings suggest that Tim-3 plays a crucial role in negative regulation of innate immune responses, through crosstalk with PD-1 and SOCS-1 and limiting STAT-1 phosphorylation, and may be a novel target for immunotherapy to HCV infection.


Journal of Immunology | 2011

Cross-Talk between Programmed Death-1 and Suppressor of Cytokine Signaling-1 in Inhibition of IL-12 Production by Monocytes/Macrophages in Hepatitis C Virus Infection

Ying Zhang; Cheng J. Ma; Lei Ni; Chun L. Zhang; Xiao Y. Wu; Uday Kumaraguru; Chuan F. Li; Jonathan P. Moorman; Zhi Q. Yao

Hepatitis C virus (HCV) dysregulates innate immune responses and induces persistent viral infection. We previously demonstrated that HCV core protein impairs IL-12 expression by monocytes/macrophages (M/MΦs) through interaction with a complement receptor gC1qR. Because HCV core-mediated lymphocyte dysregulation occurs through the negative immunomodulators programmed death-1 (PD-1) and suppressor of cytokine signaling-1 (SOCS-1), the aim of this study was to examine their role in HCV core-mediated IL-12 suppression in M/MΦs. We analyzed TLR-stimulated, primary CD14+ M/MΦs from chronically HCV-infected and healthy subjects or the THP-1 cell line for PD-1, SOCS-1, and IL-12 expression following HCV core treatment. M/MΦs from HCV-infected subjects at baseline exhibited comparatively increased PD-1 expression that significantly correlated with the degree of IL-12 inhibition. M/MΦs isolated from healthy and HCV-infected individuals and treated with HCV core protein displayed increased PD-1 and SOCS-1 expression and decreased IL-12 expression, an effect that was also observed in cells treated with gC1qR’s ligand, C1q. Blocking gC1qR rescued HCV core-induced PD-1 upregulation and IL-12 suppression, whereas blocking PD-1 signaling enhanced IL-12 production and decreased the expression of SOCS-1 induced by HCV core. Conversely, silencing SOCS-1 expression using small interfering RNAs increased IL-12 expression and inhibited PD-1 upregulation. PD-1 and SOCS-1 were found to associate by coimmunoprecipitation studies, and blocking PD-1 or silencing SOCS-1 in M/MΦ led to activation of STAT-1 during TLR-stimulated IL-12 production. These data suggested that HCV core/gC1qR engagement on M/MΦs triggers the expression of PD-1 and SOCS-1, which can associate to deliver negative signaling to TLR-mediated pathways controlling expression of IL-12, a key cytokine linking innate and adaptive immunity.


Immunology | 2011

PD-1 negatively regulates interleukin-12 expression by limiting STAT-1 phosphorylation in monocytes/macrophages duringchronic hepatitis C virus infection

Cheng J. Ma; Lei Ni; Ying Zhang; Chun L. Zhang; Xiao Y. Wu; Antwan N. Atia; Penny Thayer; Jonathan P. Moorman; Zhi Q. Yao

Hepatitis C virus (HCV) is remarkably efficient at evading host immunity to establish chronic infection. During chronic HCV infection, interleukin‐12 (IL‐12) produced by monocytes/macrophages (M/Mφ) is significantly suppressed. Programmed death‐1 (PD‐1), an inhibitory receptor on immune cells, plays a pivotal role in suppressing T‐cell responses during chronic viral infection. To determine whether PD‐1 regulates IL‐12 production by M/Mφ during chronic HCV infection, we examined the expressions of PD‐1, its ligand PDL‐1, and their relationship with IL‐12 production in M/Mφ from HCV‐infected, HCV‐resolved, and healthy subjects by flow cytometry. Toll‐like receptor (TLR) ‐mediated IL‐12 production by M/Mφ was selectively suppressed, while PD‐1/PDL‐1 expressions were up‐regulated, in HCV‐infected subjects compared with HCV‐resolved or healthy subjects. Up‐regulation of PD‐1 was inversely associated with the degree of IL‐12 inhibition in HCV infection. Interestingly, the reduced response of M/Mφ from HCV‐infected individuals to TLR ligands appeared not to be the result of a lack of the ability to sense pathogen, but to an impaired activation of intracellular janus kinase/signal transducer and activator of transfection (STAT) pathway as represented by inhibited STAT‐1 phosphorylation in M/Mφ from HCV‐infected individuals compared with HCV‐negative subjects. Successful HCV treatment with pegylated interferon/ribavirin or blocking PD‐1/PDL‐1 engagement ex vivo led to reduced PD‐1 expression and improved IL‐12 production as well as STAT‐1 activation in M/Mφ from HCV‐infected individuals. These results suggest that the PD‐1 inhibitory pathway may negatively regulate IL‐12 expression by limiting STAT‐1 phosphorylation in M/Mφ during chronic HCV infection.


Vaccine | 2011

Impaired hepatitis B vaccine responses during chronic hepatitis C infection: involvement of the PD-1 pathway in regulating CD4+ T cell responses

Jonathan P. Moorman; Chun L. Zhang; Lei Ni; Cheng J. Ma; Ying Zhang; Xiao Y. Wu; Penny Thayer; Tareq M. Islam; Thomas Borthwick; Zhi Q. Yao

Vaccination for hepatitis B virus (HBV) in the setting of hepatitis C virus (HCV) infection is recommended, but responses to vaccination are blunted when compared to uninfected populations. The mechanism for this failure of immune response in HCV-infected subjects remains unknown but is thought to be a result of lymphocyte dysfunction during chronic viral infection. We have recently demonstrated that PD-1, a novel negative immunomodulator for T cell receptor (TCR) signaling, is involved in T and B lymphocyte dysregulation during chronic HCV infection. In this report, we further investigated the role of the PD-1 pathway in regulation of CD4(+) T cell responses to HBV vaccination in HCV-infected individuals. In a prospective HCV infected cohort, a poor response rate to HBV vaccination as assayed by seroconversion was observed in HCV-infected subjects (53%), while a high response rate was observed in healthy or spontaneously HCV-resolved individuals (94%). CD4(+) T cell responses to ex vivo stimulations of anti-CD3/CD28 antibodies or hepatitis B surface antigen (HBsAg) were found to be lower in HBV vaccine non-responders compared to those responders in HCV-infected individuals who had received a series of HBV immunizations. PD-1 expression on CD4(+) T cells was detected at relatively higher levels in these HBV vaccine non-responders than those who responded, and this was inversely associated with the cell activation status. Importantly, blocking the PD-1 pathway improved T cell activation and proliferation in response to ex vivo HBsAg or anti-CD3/CD28 stimulation in HBV vaccine non-responders. These results suggest that PD-1 signaling may be involved in impairing CD4(+) T cell responses to HBV vaccination in subjects with HCV infection, and raise the possibility that blocking this negative signaling pathway might improve success rates of immunization in the setting of chronic viral infection.


Immunology and Cell Biology | 2011

PD-1 modulates regulatory T cells and suppresses T-cell responses in HCV-associated lymphoma

Lei Ni; Cheng J. Ma; Ying Zhang; Subhadra Nandakumar; Chun L. Zhang; Xiao Y. Wu; Thomas Borthwick; Agnes Hamati; Xin Y. Chen; Uday Kumaraguru; Jonathan P. Moorman; Zhi Q. Yao

T regulatory (TR) cells suppress T‐cell responses that are critical in the development of chronic viral infection and associated malignancies. Programmed death‐1 (PD‐1) also has a pivotal role in regulation of T‐cell functions during chronic viral infection. To examine the role of PD‐1 pathway in regulating TR‐cell functions that inhibit T‐cell responses during virus‐associated malignancy, TR cells were investigated in the setting of hepatitis C virus‐associated lymphoma (HCV‐L), non‐HCV‐associated lymphoma (non‐HCV‐L), HCV infection alone and healthy subjects (HS). Relatively high numbers of CD4+CD25+ and CD8+CD25+ TR cells, as well as high levels of PD‐1 expressions on these TR cells were found in the peripheral blood of subjects with HCV‐L compared with those from non‐HCV‐L or HCV alone or HS. TR cells from the HCV‐L subjects were capable of suppressing the autogeneic lymphocyte response, and depletion of TR cells in peripheral blood mononuclear cells from HCV‐L improved T‐cell proliferation. Additionally, the suppressed T‐cell activation and proliferation in HCV‐L was partially restored by blocking the PD‐1 pathway ex vivo, resulting in both a reduction in TR‐cell number and the ability of TR to suppress the activity of effector T cells. This study suggests that the PD‐1 pathway is involved in regulating TR cells that suppress T‐cell functions in the setting of HCV‐associated B‐cell lymphoma.


Journal of Innate Immunity | 2014

Myeloid-derived Suppressor Cells: Paradoxical Roles in Infection and Immunity

Jun Dai; Mohamed El Gazzar; Guang Y. Li; Jonathan P. Moorman; Zhi Q. Yao

Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of immature suppressor cells that are generated due to aberrant myelopoiesis under pathological conditions. Although MDSCs have been recognized for more than 20 years under the guise of different monikers, these particular populations of myeloid cells gained more attention recently due to their immunosuppressive properties, which halt host immune responses to growing cancers or overwhelming infections. While MDSCs may contribute to immune homeostasis after infection or tissue injury by limiting excessive inflammatory processes, their expansion may be at the expense of pathogen elimination and thus may lead to disease persistence. Therefore, MDSCs may be either damaging or obliging to the host by attenuating, for example, antitumor or anti-infectious immune responses. In this review, we recapitulate the biological and immunological aspects of MDSCs, including their generation, distribution, trafficking and the factors involved in their activation, expansion, suppressive functions, and interplay between MDSCs and regulatory T cells, with a focus on the perspectives of infection and inflammation.


European Journal of Immunology | 2013

HCV-infected hepatocytes drive CD4+CD25+Foxp3+ regulatory T-cell development through the Tim-3/Gal-9 pathway

Xiao J. Ji; Cheng J. Ma; Jia M. Wang; Xiao Y. Wu; Toshiro Niki; Mitsumi Hirashima; Jonathan P. Moorman; Zhi Q. Yao

HCV is remarkable at disrupting human immunity to establish chronic infection. The accumulation of Treg cells at the site of infection and upregulation of inhibitory signaling pathways (such as T‐cell Ig and mucin domain protein‐3 (Tim‐3) and galectin‐9 (Gal‐9)) play pivotal roles in suppressing antiviral effector T (Teff) cells that are essential for viral clearance. While Tim‐3/Gal‐9 interactions have been shown to negatively regulate Teff cells, their role in regulating Treg cells is poorly understood. To explore how Tim‐3/Gal‐9 interactions regulate HCV‐mediated Treg‐cell development, here we provide pilot data showing that HCV‐infected human hepatocytes express higher levels of Gal‐9 and TGF‐β, and upregulate Tim‐3 expression and regulatory cytokines TGF‐β/IL‐10 in co‐cultured human CD4+ T cells, driving conventional CD4+ T cells into CD25+Foxp3+ Treg cells. Additionally, recombinant Gal‐9 protein can transform TCR‐activated CD4+ T cells into Foxp3+ Treg cells in a dose‐dependent manner. Importantly, blocking Tim‐3/Gal‐9 ligations abrogates HCV‐mediated Treg‐cell induction by HCV‐infected hepatocytes, suggesting that Tim‐3/Gal‐9 interactions may regulate human Foxp3+ Treg‐cell development and function during HCV infection.


Viral Immunology | 2010

Programmed Death-1 Affects Suppressor of Cytokine Signaling-1 Expression in T Cells During Hepatitis C Infection

Ashley D. Frazier; Chun L. Zhang; Lei Ni; Cheng J. Ma; Ying Zhang; Xiao Y. Wu; Antwan N. Atia; Zhi Q. Yao; Jonathan P. Moorman

Chronic hepatitis C virus (HCV) infection is associated with T-cell exhaustion that is mediated through upregulation of the PD-1 negative regulatory pathway. PD-1 expression is induced by HCV core protein, which also induces upregulation of SOCS-1, a key modulator that controls the Jak/STAT pathway regulating cytokine expression. To determine whether these two negative regulatory pathways are linked during T-cell signaling, SOCS-1 expression was examined by blocking the PD-1 pathway in T cells stimulated with anti-CD3/CD28 in the presence of HCV core protein. T cells isolated from healthy subjects or HCV-infected individuals were treated with anti-PD-1 or anti-PDL-1 antibodies in the presence or absence of HCV core protein, and SOCS-1 gene expression was detected by RT-PCR or immunoblotting, while T-cell functions were assayed by flow cytometric analyses. Both PD-1 and SOCS-1 gene expression were upregulated in healthy T cells exposed to HCV core protein, and blocking the PD-1 pathway downregulated SOCS-1 gene expression in these cells. Additionally, T cells isolated from chronically HCV-infected subjects exhibited increased PD-1 and SOCS-1 expression compared to healthy subjects, and SOCS-1 expression in T cells isolated from HCV-infected subjects was also inhibited by blocking PD-1 signaling; this in turn enhanced the phosphorylation of STAT-1, and improved the impaired T-cell proliferation observed in the setting of HCV infection. These data demonstrate that PD-1 and SOCS-1 are linked in dysregulating T-cell signaling during HCV infection, and their cross-talk may coordinately inhibit T-cell signaling pathways that lead to T-cell exhaustion during chronic viral infection.


Journal of Virology | 2013

Differential Regulation of Interleukin-12 (IL-12)/IL-23 by Tim-3 Drives TH17 Cell Development during Hepatitis C Virus Infection

Jia M. Wang; Lei Shi; Cheng J. Ma; Xiao J. Ji; Ruo S. Ying; Xiao Y. Wu; Ke S. Wang; Guangyu Li; Jonathan P. Moorman; Zhi Q. Yao

ABSTRACT Cytokine production by innate immunity is critical for shaping the adaptive immunity through regulation of T cell differentiation. In this report, we studied T cell immunoglobulin mucin domain protein 3 (Tim-3) expression on monocytes and its regulatory effect on interleukin-12 (IL-12)/IL-23 production by CD14+ monocytes, as well as IL-17 production by CD4+ T cells in individuals with chronic hepatitis C virus (HCV) infection. We found that Tim-3 and IL-23p19 are highly expressed and that IL-12p35 is inhibited in human CD14+ monocytes, while IL-17 expression is upregulated in CD4+ T cells, in chronically HCV-infected individuals compared to healthy subjects. Interestingly, Tim-3 expression is closely associated with the differential regulation of IL-12/IL-23 expression in CD14+ monocytes and correlated to IL-17 production by CD4+ T cells. These Tim-3-associated IL-12/IL-23/IL-17 dysregulations in HCV-infected individuals are also recapitulated in vitro by incubating healthy monocytes or peripheral blood mononuclear cells with Huh-7 hepatoma cells transfected with HCV RNA. Importantly, blocking Tim-3 signaling on monocytes restores the balance of IL-12/IL-23 through the intracellular STAT3 signaling, which in turn reverses the upregulated IL-17 expression both ex vivo and in vitro. Our findings suggest that Tim-3-mediated differential regulation of IL-12/IL-23 drives TH17 cell development, a milieu favoring viral persistence and autoimmune phenomenon during HCV infection.

Collaboration


Dive into the Zhi Q. Yao's collaboration.

Top Co-Authors

Avatar

Ying Zhang

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Yun Zhou

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Zhansheng Jia

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Zhan S. Jia

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Mohamed El Gazzar

East Tennessee State University James H. Quillen College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Chao Fan

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Jian-Qi Lian

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Xiao J. Ji

East Tennessee State University James H. Quillen College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge