Network


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

Hotspot


Dive into the research topics where Yi Chi M Kong is active.

Publication


Featured researches published by Yi Chi M Kong.


Immunogenetics | 1982

Regulation of experimental autoimmune thyroiditis: Mapping of susceptibility to the I-A subregion of the mouse H-2

K. W. Beisel; Chella S. David; Alvaro A. Giraldo; Yi Chi M Kong; Noel R. Rose

Experimental autoimmune thyroiditis (EAT) is induced in certain mouse strains by injection of mouse thyroglobulin (MTg) with a suitable adjuvant (Rose et al. 1971). The severity of thyroiditis can be ascertained by the extent of mononuclear cell infiltration into the thyroid. Mice of the H-2 k haplotype displayed the highest incidence of severe disease, compared with 10w incidence of mild disease in mice of the b haplotype (Vladutiu and Rose 1971, Rose et al. 1981). The titers of MTg antibodies were less discriminating than histopathologic findings. Lymphocyte proliferative responses as well as the production of antibody and thyroid lesions are under H-2 control (Christadoss et al. 1978). The principal gene controlling susceptibility to EAT, Ir-Tg, was mapped to the K and/or I-A region (Tomazic et al. 1974). At that time, suitable intra-H-2 recombinant strains were not available to permit more precise localization of the gene. Recently we have found that genes at the D end modify the severity of thyroid lesions (Kong et al. 1979). Other studies of a K-region mutant, B6.C-H-2 bin1 (H1), have suggested that a gene in this region regulates susceptibility to thyroiditis (Maron and Cohen 1979). To distinguish the relative contribution of the K and I -A regions to EAT, several new intra-H-2 recombinant strains and H-2K-region mutants have been used to pinpoint the Ir-Tg gene. In this brief communication, we report the definitive mapping of the major Ir-Tg gene to the I-A subregion. Our intra-H-2 recombinant study further suggests that the K region, like the D region, may modify the incidence of thyroiditis. In contrast to the finding of Maron and Cohen (1979), our use of 6 K b region mutants, including bml, did not show an increased incidence of EAT.


Cancer Research | 2005

Concurrent Induction of Antitumor Immunity and Autoimmune Thyroiditis in CD4+CD25+ Regulatory T Cell–Depleted Mice

Wei Zen Wei; Jennifer B. Jacob; John Zielinski; Jeffrey C. Flynn; K. David Shim; Ghazwan Alsharabi; Alvaro A. Giraldo; Yi Chi M Kong

When CD4+ CD25+ regulatory T cells are depleted or inactivated for the purpose of enhancing antitumor immunity, the risk of autoimmune disease may be significantly elevated because these regulatory T cells control both antitumor immunity and autoimmunity. To evaluate the relative benefit and risk of modulating CD4+ CD25+ regulatory T cells, we established a new test system to measure simultaneously the immune reactivity to a tumor-associated antigen, neu, and an unrelated self-antigen, thyroglobulin. BALB/c mice were inoculated with TUBO cells expressing an activated rat neu and treated with anti-CD25 monoclonal antibody to deplete CD25+ cells. The tumors grew, then regressed, and neu-specific antibodies and IFN-gamma-secreting T cells were induced. The same mice were also exposed to mouse thyroglobulin by chronic i.v. injections. These mice produced thyroglobulin-specific antibody and IFN-gamma-secreting T cells with inflammatory infiltration in the thyroids of some mice. The immune responses to neu or thyroglobulin were greater in mice undergoing TUBO tumor rejection and thyroglobulin injection than in those experiencing either alone. To the best of our knowledge, this is the first experimental system to assess the concurrent induction and possible synergy of immune reactivity to defined tumor and self-antigens following reduction of regulatory T cells. These results illustrate the importance of monitoring immune reactivity to self-antigens during cancer immunotherapy that involves immunomodulating agents, and the pressing need for novel strategies to induce antitumor immunity while minimizing autoimmunity.


Clinical Immunology and Immunopathology | 1981

Effect of castration and sex hormones on experimental autoimmune thyroiditis

Isao Okayasu; Yi Chi M Kong; Noel R. Rose

Abstract To determine the effect of sex hormones on experimental autoimmune thyroiditis, male and female good responder and poor responder mice were castrated at 2 or 4 weeks of age. Following castration some groups of good responder mice were further treated with β-estradiol or 5-α-dihydrotestosterone implants. At 9 to 11 weeks of age, autoimmune thyroiditis was induced by injection of mouse thyroglobulin plus adjuvant. Castration significantly increased antibody levels only in good responder male mice, an effect that was reversed by continuous administration of testosterone. Raising estrogen levels did not affect either antibody levels or the extent of cellular infiltration of the thyroid in female mice but significantly increased antibody levels in good responder males.


Frontiers in Immunology | 2014

Opportunistic Autoimmune Disorders Potentiated by Immune-Checkpoint Inhibitors Anti-CTLA-4 and Anti-PD-1

Yi Chi M Kong; Jeffrey C. Flynn

To improve the efficacy of immunotherapy for cancer and autoimmune diseases, recent ongoing and completed clinical trials have focused on specific targets to redirect the immune network toward eradicating a variety of tumors and ameliorating the self-destructive process. In a previous review, both systemic immunomodulators and monoclonal antibodies (mAbs), anti-CTLA-4, and anti-CD52, were discussed regarding therapeutics and autoimmune sequelae, as well as predisposing factors known to exacerbate immune-related adverse events (irAEs). This review will focus on immune-checkpoint inhibitors, and the data from most clinical trials involve blockade with anti-CTLA-4 such as ipilimumab. However, despite the mild to severe irAEs observed with ipilimumab in ~60% of patients, overall survival (OS) averaged ~22–25% at 3–5 years. To boost OS, other mAbs targeting programed death-1 and its ligand are undergoing clinical trials as monotherapy or dual therapy with anti-CTLA-4. Therapeutic combinations may generate different spectrum of opportunistic autoimmune disorders. To simulate clinical scenarios, we have applied regulatory T cell perturbation to murine models combined to examine the balance between thyroid autoimmunity and tumor-specific immunity.


Clinical Immunology and Immunopathology | 1989

Resistance to experimental autoimmune thyroiditis: L3T4+ cells as mediators of both thyroglobulin-activated and TSH-induced suppression.

Yi Chi M Kong; Alvaro A. Giraldo; Herman Waldmann; Steve Cobbold; Brian E. Fuller

Mechanisms suppressive to induction of murine experimental autoimmune thyroiditis (EAT) can be activated by pretreatment with tolerogenic doses of mouse thyroglobulin (MTg) or prior TSH infusion to raise circulatory MTg levels. MTg-activated suppressor T cells (Ts), shown earlier to be Thy-1+ and probably I-J+, were further characterized by in vivo administration of paired rat monoclonal antibodies to distinct epitopes on the L3T4 or Lyt-2 molecule, either on the day of, or subsequent to, initiation of the tolerogenic regimes. The cells required at the time of MTg pretreatment were L3T4+, Lyt-2- and low anti-L3T4 doses had no effect on their activation. The cells that mediated the strong MTg-induced resistance following pretreatment were also L3T4+; their suppressor function could only be abrogated by depletion of L3T4+, but not Lyt-2+, cells. Injection of cyclophosphamide (20-100 mg/kg) either prior to EAT induction or after Ts activation did not affect the severity of disease. Similarly, the suppressor state evoked by TSH infusion could only be abrogated by anti-L3T4 treatment. These findings indicate that both MTg-activated and TSH-induced suppression are mediated by L3T4+ cells. We hypothesize that MTg-specific Ts are present in normal, EAT-susceptible mice in low numbers to contribute to the maintenance of self-tolerance and that they are stimulated by increased levels of circulatory MTg to expand/differentiate and mediate the marked resistance to EAT induction.


Clinical Immunology and Immunopathology | 1986

Activation of cytotoxic T cells and effector cells in experimental autoimmune thyroiditis by shared determinants of mouse and human thyroglobulins.

Laurel L. Simon; James M. Justen; Alvaro A. Giraldo; Christopher J. Krco; Yi Chi M Kong

Previous studies have shown that T cells from genetically susceptible mice developing experimental autoimmune thyroiditis (EAT) proliferate in response to restimulation with mouse thyroglobulin (MTg) in vitro and differentiate into cells cytotoxic for syngeneic thyroid monolayers. To examine further the effector cells involved in pathogenesis and the determinants on MTg responsible for their activation, spleen cells (SC) and lymph node cells (LNC) from mice immunized with MTg or human (H) Tg, and adjuvant (complete Freunds adjuvant (CFA) or lipopolysaccharide (LPS] were cultured in vitro with MTg or HTg. Control cultures were incubated with concanavalin A (Con A) or purified protein derivative (PPD). The in vitro-activated cells which proliferated in response to MTg, HTg, or Con A adoptively transferred thyroiditis to normal recipients, whereas cells transferred directly without in vitro culture were very ineffective. The capacity to transfer EAT was abrogated by irradiation (1500 R), and SC from CFA-immunized control mice which responded in vitro to PPD stimulation did not transfer thyroiditis. The serum titers of MTg autoantibodies were uniformly low and were not correlated with severity of disease. The localization of EAT-effector (precursor) cells depended upon the site of immunization; they were found in the spleens after inguinal (subcutaneous) or systemic (intravenous) immunizations, but were present in the popliteal lymph nodes after hind footpad injections. Both homologous MTg and heterologous HTg functioned as in vivo sensitizing antigen and in vitro activating antigen for each other; such cultured cells transferred thyroiditis in vivo and became cytotoxic for thyroid monolayers in vitro. These findings show that shared determinants are autoantigenic and thyroiditogenic, and support the hypothesis that EAT-effector cells responsible for initiating thyroid damage include cytotoxic cells.


Cellular Immunology | 1981

In vitro T-lymphocyte proliferative response to mouse thyroglobulin in experimental autoimmune thyroiditis

Isao Okayasu; Yi Chi M Kong; Chella S. David; Noel R. Rose

Abstract The in vitro lymphocyte proliferative response to mouse thyroglobulin (MTg) was studied in good and poor responder mice in relationship to in vivo antibody formation and thyroid infiltration. CBA( H-2 k ) and BALB/c( H-2 d ) mice were immunized in the hind footpads with MTg incorporated into complete Freunds adjuvant (CFA). At weekly intervals up to 28 days, groups of mice were sacrificed. Their popliteal lymph nodes were cultured in vitro for proliferative response to MTg and their antibody levels and thyroid involvement determined. In good responder CBA mice, the proliferative responses to MTg were strongest on Days 8 to 14, where they were 9- to 14-fold over control levels, depending on the day of harvest. The response declined to 2- to 4-fold over background on Days 21 to 28, although high antibody levels were present throughout this period. The proliferative response was abrogated by anti-Thy-1 treatment, indicating its dependence on T cells. In poor responder BALB/c mice, no significant proliferative responses to MTg were observed at any time, although the animals displayed moderate levels of MTg antibody. The responses to PPD, in contrast, were similar in both strains, usually being 4- to 7-fold above background. Thyroid infiltration, like the proliferative response to MTg, was observed only in CBA mice. Thus lymphocyte proliferation at 8 to 14 days represents a reliable, early in vitro correlate of autoimmune thyroiditis induced with CFA as adjuvant.


Cellular Immunology | 1989

Depletion of L3T4+ and Lyt-2+ cells by rat monoclonal antibodies alters the development of adoptively transferred experimental autoimmune thyroiditis

Jeffrey C. Flynn; Dale H. Conaway; Steve Cobbold; Herman Waldmann; Yi Chi M Kong

To delineate the contribution of L3T4+ and Lyt-2+ cells in the pathogenesis of experimental autoimmune thyroiditis (EAT), synergistic pairs of monoclonal antibodies (mAb) to the T cell subsets were used in conjunction with the adoptive transfer of mouse thyroglobulin (MTg)-activated cells from immunized mice. Initial experiments verified the important role of L3T4+ cells in the transfer of EAT. Subsequent experiments pointed to the relative contribution of both L3T4+ and Lyt-2+ cells, depending on the stage and extent of disease development. Treatment during disease with L3T4, but not Lyt-2, mAb alone significantly reduced thyroiditis. However, in situ analysis of the cellular infiltrate in thyroid sections revealed that, after treatment with mAb, the appropriate subset was eliminated without altering the amount of the other subset in the remaining lesion. In addition, treatment during severe thyroiditis following the transfer of MTg-activated lymph node cells showed that Lyt-2 mAb alone also reduced thyroid infiltration. When the recipients were pretreated with either pair of mAb before transfer, disease development was only moderately affected. We conclude that (i) donor L3T4+ cells are the primary cells responsible for the initial transfer and development of thyroiditis; and (ii) previous in vitro cytotoxicity data, plus current monoclonal antibody treatment of disease and in situ analysis, further implicate a role for Lyt-2+ cells in EAT pathogenesis.


Immunogenetics | 1997

Role of mouse and human class II transgenes in susceptibility to and protection against mouse autoimmune nhyroiditis

Yi Chi M Kong; Chella S. David; Lesley C. Lomo; Brian E. Fuller; Reinhard W. Motte; Alvaro A. Giraldo

Abstract Mouse experimental autoimmune thyroiditis (EAT), a model for Hashimoto’s thyroiditis, is induced by immunizing with mouse thyroglobulin (MTg). To study the extent of H2A involvement in EAT, we introduced AaAb genes from susceptible k mice into resistant or intermediately susceptible strains which do not express H2E molecules. Thyroiditis was severe in resistant B10.M (H2f) mice carrying the double transgene AakAbk. Likewise, thyroid infiltration was significantly extended in intermediate B10.Q (H2q) mice with the same transgene. To examine the effect of H2E molecules in the presence of H2A-mediated susceptibility, we introduced an Eaktransgene into E– B10.S mice to express the Eβs molecule and observed significant reduction in EAT severity in B10.S(E+) mice. On the other hand, the presence of an Ebd transgene in B10.RQB3 (H2Aq) mice resulting in the expression of H2Eβd molecules did not alter EAT susceptibility, suggesting a role for Eb gene polymorphism in protection against EAT. We have shown recently that the HLA-DRB1*0301 (DR3) transgene conferred EAT susceptibility to B10.M as well as class II-negative B10.Ab0 mice. However, we report here that the HLA-DQB1*0601 (DQ6b) transgene in B10.M or HLA-DQA1*0301/DQB1*0302 (DQ8) transgene in class II-negative Ab0 mice did not. These studies show the differential effects of class II molecules on EAT induction. Susceptibility can be determined when class II molecules from a single locus, H2A or HLA-DQ, are examined in transgenic mice, but the overall effect may depend upon the presence of both class II molecules H2A and H2E in mice and HLA-DQ and HLA-DR in humans.


Cellular Immunology | 1985

Characterization of the in vitro murine T-cell proliferative responses to murine and human thyroglobulins in thyroiditis-susceptible and -resistant mice

Laurel L. Simon; Christopher J. Krco; Chella S. David; Yi Chi M Kong

The in vitro proliferative response to autoantigenic mouse thyroglobulin (MTg) of lymph node cells (LNC) from thyroiditis-susceptible (high-responder) CBA/J (H-2k) mice was further characterized. The relatively weak response was enhanced by adding irradiated spleen cells from normal syngeneic mice to cultures of responding LNC. Furthermore, the adjuvant used for immunization was found to influence the magnitude of the response. Results of experiments varying both the adjuvant and the route of immunization (footpad versus subcutaneous) demonstrated that marked proliferative response to MTg in vitro was not necessarily a predictor of the severity of disease. However, the capacity to proliferate in response to MTg correlated with disease susceptibility, as reported previously. The response to MTg was dependent on Thy-1+, Lyt-1+2- cells and was inhibited by monoclonal I-A antibodies. Thus, proliferation is mediated by T cells of the helper/amplifier phenotype recognizing the autoantigen in association with Ia molecules. The determinants on human thyroglobulin (HTg) and MTg stimulating the proliferative responses of LNC from thyroiditis-susceptible and thyroiditis-resistant (low-responder) BALB/c (H-2d) mice were found to differ. Cells from resistant mice proliferated only in response to foreign determinants on HTg and not to shared or mouse-specific epitopes of MTg, whereas susceptible mice had T cells reactive to shared determinants expressed on MTg and HTg as well as to foreign determinants on HTg.

Collaboration


Dive into the Yi Chi M Kong's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Qiang Wan

Wayne State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Noel R. Rose

Brigham and Women's Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge