Network


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

Hotspot


Dive into the research topics where Chunhe Wang is active.

Publication


Featured researches published by Chunhe Wang.


Journal of Immunology | 2006

Splenic atrophy in experimental stroke is accompanied by increased regulatory T cells and circulating macrophages

Halina Offner; Sandhya Subramanian; Susan M. Parker; Chunhe Wang; Michael Afentoulis; Anne D. Lewis; Arthur A. Vandenbark; Patricia D. Hurn

Induction of stroke not only produces local ischemia and brain damage, but also has profound effects on peripheral immune responses. In the current study, we evaluated effects on spleen and blood cells 4 days after stroke induction. Surprisingly, there was a less inflammatory cytokine profile in the middle cerebral artery occlusion-affected right brain hemisphere at 96 h compared with earlier time points. Moreover, our results demonstrate that stroke leads to splenic atrophy characterized by a reduction in organ size, a drastic loss of splenocyte numbers, and induction of annexin V+ and TUNEL+ cells within the spleen that are in the late stages of apoptosis. The consequence of this process was to reduce T cell proliferation responses and secretion of inflammatory cytokines, resulting in a state of profound immunosuppression. These changes produced a drastic reduction in B cell numbers in spleen and blood, and a novel increase in CD4+FoxP3+ regulatory T cells. Moreover, we detected a striking increase in the percentage of nonapoptotic CD11b+ VLA-4-negative macrophages/monocytes in blood. Immunosuppression in response to brain injury may account for the reduction of inflammatory factors in the stroke-affected brain, but also potentially could curtail protective immune responses in the periphery. These findings provide new evidence to support the contention that damage to the brain caused by cerebral ischemia provides a powerful negative signal to the peripheral immune system that ultimately induces a drastic state of immunosuppression caused by cell death as well as an increased presence of CD4+FoxP3+ regulatory T cells.


Journal of Immunology | 2009

Membrane estrogen receptor regulates experimental autoimmune encephalomyelitis through up-regulation of programmed death 1

Chunhe Wang; Babak Dehghani; Yuexin Li; Laurie J. Kaler; Thomas M. Proctor; Arthur A. Vandenbark; Halina Offner

Although estrogens exert a pronounced protective effect on multiple sclerosis and its animal model, experimental autoimmune encephalomyelitis (EAE), their therapeutic application has been limited by undesirable side effects thought to be mediated primarily through estradiol binding to intracellular estrogen receptor α. In this study, we found that signaling through the putative membrane estrogen receptor, G protein-coupled receptor 30 (GPR30), was sufficient to mediate protection against EAE, which was significantly impaired in GPR30 gene-deficient mice. Treatment with G-1, an agonist that selectively activates GPR30 without engagement of the intracellular estrogen receptors, retained the ability of estradiol to protect against clinical and histological EAE without estradiol-associated side effects, deviated cytokine profiles, and enhanced suppressive activity of CD4+Foxp3+ T regulatory cells through a GPR30- and programmed death 1-dependent mechanism. This study is the first to evaluate the protective effect of GPR30 activation on EAE, and provides a strong foundation for the clinical application of GPR30 agonists such as G-1 in multiple sclerosis.


Journal of Immunology | 2003

Recombinant TCR Ligand Induces Tolerance to Myelin Oligodendrocyte Glycoprotein 35-55 Peptide and Reverses Clinical and Histological Signs of Chronic Experimental Autoimmune Encephalomyelitis in HLA-DR2 Transgenic Mice

Arthur A. Vandenbark; Cathleen Rich; Jeff Mooney; Alex Zamora; Chunhe Wang; Jianya Huan; Lars Fugger; Halina Offner; Richard E. Jones; Gregory G. Burrows

In a previous study, we demonstrated that myelin oligodendrocyte glycoprotein (MOG)-35-55 peptide could induce severe chronic experimental autoimmune encephalomyelitis (EAE) in HLA-DR2+ transgenic mice lacking all mouse MHC class II genes. We used this model to evaluate clinical efficacy and mechanism of action of a novel recombinant TCR ligand (RTL) comprised of the α1 and β1 domains of DR2 (DRB1*1501) covalently linked to the encephalitogenic MOG-35-55 peptide (VG312). We found that the MOG/DR2 VG312 RTL could induce long-term tolerance to MOG-35-55 peptide and reverse clinical and histological signs of EAE in a dose- and peptide-dependent manner. Some mice treated with lower doses of VG312 relapsed after cessation of daily treatment, but the mice could be successfully re-treated with a higher dose of VG312. Treatment with VG312 strongly reduced secretion of Th1 cytokines (TNF-α and IFN-γ) produced in response to MOG-35-55 peptide, and to a lesser degree purified protein derivative and Con A, but had no inhibitory effect on serum Ab levels to MOG-35-55 peptide. Abs specific for both the peptide and MHC moieties of the RTLs were also present after treatment with EAE, but these Abs had only a minor enhancing effect on T cell activation in vitro. These data demonstrate the powerful tolerance-inducing therapeutic effects of VG312 on MOG peptide-induced EAE in transgenic DR2 mice and support the potential of this approach to inhibit myelin Ag-specific responses in multiple sclerosis patients.


Immunology | 2009

Oestrogen modulates experimental autoimmune encephalomyelitis and interleukin‐17 production via programmed death 1

Chunhe Wang; Babak Dehghani; Yuexin Li; Laurie J. Kaler; Arthur A. Vandenbark; Halina Offner

The mechanism by which oestrogens suppress experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis, is only partially understood. We here demonstrate that treatment with 17β‐oestradiol (E2) in C57BL/6 mice boosted the expression of programmed death 1 (PD‐1), a negative regulator of immune responses, in the CD4+ FoxP3+ regulatory T (Treg) cell compartment in a dose‐dependent manner that correlated with the efficiency of EAE protection. Administration of E2 at pregnancy levels but not lower concentrations also enhanced the frequency of Treg cells. Additionally, E2 treatment drastically reduced the production of interleukin‐17 (IL‐17) in the periphery of immunized mice. However, E2 treatment did not protect against EAE or suppress IL‐17 production in PD‐1 gene‐deficient mice. Finally, E2 failed to prevent Treg‐deficient mice from developing spontaneous EAE. Taken together, our results suggest that E2‐induced protection against EAE is mediated by upregulation of PD‐1 expression within the Treg‐cell compartment.


Journal of Neurochemistry | 2005

Dopamine D2 receptor stimulation of mitogen‐activated protein kinases mediated by cell type‐dependent transactivation of receptor tyrosine kinases

Chunhe Wang; David C. Buck; Rui Yang; Tara A. Macey; Kim A. Neve

Dopamine D2 receptor activation of extracellular signal‐regulated kinases (ERKs) in non‐neuronal human embryonic kidney 293 cells was dependent on transactivation of the platelet‐derived growth factor (PDGF) receptor, as demonstrated by the effect of the PDGF receptor inhibitors tyrphostin A9 and AG 370 on quinpirole‐induced phosphorylation of ERKs and by quinpirole‐induced tyrosine phosphorylation of the PDGF receptor. In contrast, ectopically expressed D2 receptor or endogenous D2‐like receptor activation of ERKs in NS20Y neuroblastoma cells, which express little or no PDGF receptor, or in rat neostriatal neurons was largely dependent on transactivation of the epidermal growth factor (EGF) receptor, as demonstrated using the EGF receptor inhibitor AG 1478 and by quinpirole‐induced phosphorylation of the EGF receptor. The D2 receptor agonist quinpirole enhanced the coprecipitation of D2 and EGF receptors in NS20Y cells, suggesting that D2 receptor activation induced the formation of a macromolecular signaling complex that includes both receptors. Transactivation of the EGF receptor also involved the activity of a matrix metalloproteinase. Thus, although D2 receptor stimulation of ERKs in both cell lines was decreased by inhibitors of ERK kinase, Src‐family protein tyrosine kinases, and serine/threonine protein kinases, D2‐like receptors activated ERKs via transactivation of the EGF receptor in NS20Y neuroblastoma cells and rat embryonic neostriatal neurons, but via transactivation of the PDGF receptor in 293 cells.


European Journal of Immunology | 2011

Estrogen-induced protection against experimental autoimmune encephalomyelitis is abrogated in the absence of B cells.

Sheetal Bodhankar; Chunhe Wang; Arthur A. Vandenbark; Halina Offner

Increased remissions in multiple sclerosis (MS) during pregnancy suggest that elevated levels of sex steroids exert immunoregulatory activity. Estrogen (E2=17β‐estradiol) protects against experimental autoimmune encephalomyelitis (EAE), but the cellular basis for E2‐induced protection remains unclear. Studies demonstrate that depletion of B cells prior to induction of EAE exacerbates disease severity, implicating regulatory B cells. We thus evaluated pathogenic and E2‐induced protective mechanisms in B‐cell‐deficient (μMT−/−) mice. EAE‐protective effects of E2 were abrogated in μMT−/− mice, with no reduction in disease severity, cellular infiltration or pro‐inflammatory factors in the central nervous system compared to untreated controls. E2 treatment of WT mice selectively upregulated expression of PD‐L1 on B cells and increased the percentage of IL‐10‐producing CD1dhighCD5+ regulatory B cells. Upregulation of PD‐L1 was critical for E2‐mediated protection since E2 did not inhibit EAE in PD‐L1−/− mice. Direct treatment of B cells with E2 significantly reduced proliferation of MOG35–55‐specific T cells that required estrogen receptor‐α (ERα). These results demonstrate, for the first time, a requirement for B cells in E2‐mediated protection against EAE involving direct E2 effects on regulatory B cells mediated through ERα and the PD‐1/PD‐L1 negative co‐stimulatory pathway. E2‐primed B cells may represent an important regulatory mechanism in MS and have strong implications for women receiving current MS therapies that cause B‐cell depletion.


Journal of Immunology | 2001

Rudimentary TCR Signaling Triggers Default IL-10 Secretion by Human Th1 Cells

Gregory G. Burrows; Yuan K. Chou; Chunhe Wang; Justin W. Chang; Thomas P. Finn; Nicole Culbertson; Joseph Kim; Dennis Bourdette; Deborah A. Lewinsohn; David M. Lewinsohn; Masayuki Ikeda; Tohru Yoshioka; Charles N. Allen; Halina Offner; Arthur A. Vandenbark

Understanding the process of inducing T cell activation has been hampered by the complex interactions between APC and inflammatory Th1 cells. To dissociate Ag-specific signaling through the TCR from costimulatory signaling, rTCR ligands (RTL) containing the α1 and β1 domains of HLA-DR2b (DRA*0101:DRB1*1501) covalently linked with either the myelin basic protein peptide 85–99 (RTL303) or CABL-b3a2 (RTL311) peptides were constructed to provide a minimal ligand for peptide-specific TCRs. When incubated with peptide-specific Th1 cell clones in the absence of APC or costimulatory molecules, only the cognate RTL induced partial activation through the TCR. This partial activation included rapid TCR ζ-chain phosphorylation, calcium mobilization, and reduced extracellular signal-related kinase activity, as well as IL-10 production, but not proliferation or other obvious phenotypic changes. On restimulation with APC/peptide, the RTL-pretreated Th1 clones had reduced proliferation and secreted less IFN-γ; IL-10 production persisted. These findings reveal for the first time the rudimentary signaling pattern delivered by initial engagement of the external TCR interface, which is further supplemented by coactivation molecules. Activation with RTLs provides a novel strategy for generating autoantigen-specific bystander suppression useful for treatment of complex autoimmune diseases.


Journal of Immunology | 2003

Recombinant TCR ligand induces early TCR signaling and a unique pattern of downstream activation

Chunhe Wang; Jeffery L. Mooney; Roberto Meza-Romero; Yuan K. Chou; Jianya Huan; Arthur A. Vandenbark; Halina Offner; Gregory G. Burrows

Recombinant TCR ligands (RTLs) consisting of covalently linked α1 and β1 domains of MHC class II molecules tethered to specific antigenic peptides represent minimal TCR ligands. In a previous study we reported that the rat RTL201 construct, containing RT1.B MHC class II domains covalently coupled to the encephalitogenic guinea pig myelin basic protein (Gp-MBP72–89) peptide, could prevent and treat actively and passively induced experimental autoimmune encephalomyelitis in vivo by selectively inhibiting Gp-MBP72–89 peptide-specific CD4+ T cells. To evaluate the inhibitory signaling pathway, we tested the effects of immobilized RTL201 on T cell activation of the Gp-MBP72–89-specific A1 T cell hybridoma. Activation was exquisitely Ag-specific and could not be induced by RTL200 containing the rat MBP72–89 peptide that differed by a threonine for serine substitution at position 80. Partial activation by RTL201 included a CD3ζ p23/p21 ratio shift, ZAP-70 phosphorylation, calcium mobilization, NFAT activation, and transient IL-2 production. In comparison, anti-CD3ε treatment produced stronger activation of these cellular events with additional activation of NF-κB and extracellular signal-regulated kinases as well as long term increased IL-2 production. These results demonstrate that RTLs can bind directly to the TCR and modify T cell behavior through a partial activation mechanism, triggering specific downstream signaling events that deplete intracellular calcium stores without fully activating T cells. The resulting Ag-specific activation of the transcription factor NFAT uncoupled from the activation of NF-κB or extracellular signal-regulated kinases constitutes a unique downstream activation pattern that accounts for the inhibitory effects of RTL on encephalitogenic CD4+ T cells.


Journal of Neurochemistry | 2006

Antigen‐Specific Therapy Promotes Repair of Myelin and Axonal Damage in Established EAE

Chunhe Wang; Bruce G. Gold; Laurie J. Kaler; Xiaolin Yu; Michael Afentoulis; Gregory G. Burrows; Arthur A. Vandenbark; Dennis Bourdette; Halina Offner

Inflammation results in CNS damage in multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE), an animal model of MS. It is uncertain how much repair of injured myelin and axons can occur following highly selective anti‐inflammatory therapy in EAE and MS. In this study, SJL/J mice with established EAE were treated successfully with an antigen‐specific recombinant T cell receptor ligand (RTL), RTL401, a mouse I‐As/PLP‐139–151 construct, after the peak of EAE. To define the mechanisms by which late application of RTL401 inhibits EAE, we evaluated mice at different time points to assess the levels of neuroinflammation and myelin and axon damage in their spinal cords. Our results showed that RTL401 administered after the peak of acute EAE induced a marked reduction in inflammation in the CNS, associated with a significant reduction of demyelination, axonal loss and ongoing damage. Electron microscopy showed that RTL‐treated mice had reduced pathology compared with mice treated with vehicle and mice at the peak of disease, as demonstrated by a decrease in continued degeneration, increase in remyelinating axons and the presence of an increased number of small, presumably regenerative axonal sprouts. These findings indicate that RTL therapy targeting encephalitogenic T cells may promote CNS neuroregenerative processes.


Journal of Immunology | 2005

Treatment of Passive Experimental Autoimmune Encephalomyelitis in SJL Mice with a Recombinant TCR Ligand Induces IL-13 and Prevents Axonal Injury

Halina Offner; Sandhya Subramanian; Chunhe Wang; Michael Afentoulis; Arthur A. Vandenbark; Jianya Huan; Gregory G. Burrows

The major goal of this study was to evaluate the efficacy and mechanism of a rTCR ligand (RTL) construct (I-As/proteolipid protein (PLP)-139–151 peptide = RTL401) for treatment of SJL/J mice developing passive experimental autoimmune encephalomyelitis (EAE) that did not involve coimmunization with the highly inflammatory CFA. Our results demonstrated clearly that RTL401 was highly effective in treating passive EAE, with kinetics of recovery from disease very similar to treatment of actively induced EAE. The potent RTL401 treatment effect was reflected by a partial reduction of infiltrating mononuclear cells into CNS, minimal inflammatory lesions in spinal cord, and preservation of axons injured in vehicle-treated mice during the progression of EAE. Interestingly, in the absence of CFA, RTL401 treatment strongly enhanced production of the Th2 cytokine, IL-13, in spleen, blood, and spinal cord tissue, with variable effects on other Th1 and Th2 cytokines, and no significant effect on the Th3 cytokine, TGF-β1, or on FoxP3 that is expressed by regulatory T cells. Moreover, pretreatment of PLP-139–151-specific T cells with RTL401 in vitro induced high levels of secreted IL-13, with lesser induction of other pro- and anti-inflammatory cytokines. Given the importance of IL-13 for protection against EAE, these data strongly implicate IL-13 as a dominant regulatory cytokine induced by RTL therapy. Pronounced IL-13 levels coupled with marked reduction in IL-6 levels secreted by PLP-specific T cells from blood after treatment of mice with RTL401 indicate that IL-13 and IL-6 may be useful markers for following effects of RTL therapy in future clinical trials in multiple sclerosis.

Collaboration


Dive into the Chunhe Wang's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge