Clinical and experimental data support a pathogenic role for MHC-directed alloantibodies (alloAb) in both acute and chronic rejection after solid organ and cell transplantation. The absence of monitoring tools to detect and specific immunotherapies to suppress humoral alloimmunity presents significant clinical challenges. Cellular transplants are likely at higher risk for rejection and graft loss due to humoral immunity. Despite the importance of humoral alloimmunity in clinical transplant, there is relatively little known about the regulation of isotypes, quantity, specificity and pathogenicity of alloAb after transplant. The overall focus of this research project is to understand fundamental cellular and molecular mechanisms regulating alloAb production in the presence or absence of conventional immunosuppression. Our manuscript in press provides first evidence of the pivotal role that IFN?+CD8+ T cells play in inhibiting the magnitude of post transplant alloAb production. Our studies to elucidate this novel CD8-dependent mechanism of post transplant alloAb regulation represents a paradigm shift necessary to identify new cellular and molecular therapeutic targets.
In Aim 1, we will investigate the role of innate and adaptive immune cell subsets and cytokines/cytokine receptors critical to CD8-mediated inhibition of post transplant alloAb production.
In Aim 2 we will identify cytotoxic effector and MHC molecules necessary for CD8-mediated killing of alloprimed B cells and identify gene and protein expression profiles associated with alloprimed B cells in high and low alloAb producers.
In Aim 3 we will determine how mTOR and CN inhibition differentially suppress posttransplant alloAb production in vivo.

Public Health Relevance

Although current immunosuppressive drugs that inhibit T cells protect transplant patients from early rejection, both early and delayed rejection can occur due to B cell production of donor-reactive antibody (alloantibody). Our group recently discovered that CD8+ T cells regulate alloantibody production. Proposed studies will investigate how this novel pathway suppresses alloantibody and how alloantibody is regulated in the presence or absence of commonly used immunosuppressive drugs. This research is expected to lead to innovative immunosuppressive strategies to prevent alloantibody production and improve long-term graft survival thereby enhancing patients'health and/or quality of life.

Agency
National Institute of Health (NIH)
Institute
National Institute of Allergy and Infectious Diseases (NIAID)
Type
Research Project (R01)
Project #
5R01AI083456-04
Application #
8699666
Study Section
Transplantation, Tolerance, and Tumor Immunology Study Section (TTT)
Program Officer
Rice, Jeffrey S
Project Start
2011-08-15
Project End
2015-07-31
Budget Start
2014-08-01
Budget End
2015-07-31
Support Year
4
Fiscal Year
2014
Total Cost
Indirect Cost
Name
Ohio State University
Department
Surgery
Type
Schools of Medicine
DUNS #
City
Columbus
State
OH
Country
United States
Zip Code
43210
Zimmerer, Jason M; Liu, Xin L; Blaszczak, Alecia et al. (2018) Critical Role of Macrophage Fc?R Signaling and Reactive Oxygen Species in Alloantibody-Mediated Hepatocyte Rejection. J Immunol 201:3731-3740
Zimmerer, Jason M; Horne, Phillip H; Fisher, Mason G et al. (2016) Unique CD8+ T Cell-Mediated Immune Responses Primed in the Liver. Transplantation 100:1907-15
Avila, Christina L; Zimmerer, Jason M; Elzein, Steven M et al. (2016) mTOR Inhibition Suppresses Posttransplant Alloantibody Production Through Direct Inhibition of Alloprimed B Cells and Sparing of CD8+ Antibody-Suppressing T cells. Transplantation 100:1898-906
Zimmerer, J M; Swamy, P; Sanghavi, P B et al. (2014) Critical role of NKT cells in posttransplant alloantibody production. Am J Transplant 14:2491-9
Zimmerer, J M; Pham, T A; Wright, C L et al. (2014) Alloprimed CD8(+) T cells regulate alloantibody and eliminate alloprimed B cells through perforin- and FasL-dependent mechanisms. Am J Transplant 14:295-304
Zimmerer, Jason M; Horne, Phillip H; Fiessinger, Lori A et al. (2013) Inhibition of recall responses through complementary therapies targeting CD8+ T-cell- and alloantibody-dependent allocytotoxicity in sensitized transplant recipients. Cell Transplant 22:1157-69
Zimmerer, Jason M; Horne, Phillip H; Fiessinger, Lori A et al. (2012) Cytotoxic effector function of CD4-independent, CD8(+) T cells is mediated by TNF-?/TNFR. Transplantation 94:1103-10