Many aspects of acute and chronic rejection can be affected by complement because split products of complement influence the localization, activation and effector functions of platelets, granulocytes, monocytes, and lymphocytes. Transplants biopsied during acute rejection episodes associated with antibody and complement deposition demonstrate endothelial injury, neutrophil margination, platelet accumulation, and fibrin deposition in capillaries. The complement cascade is a potent, multifaceted mediator of inflammation. Systemic production of complement occurs in the liver, but macrophages, platelets, T-lymphocytes and endothelial cells can produce complement components in local sites of inflammation. The complement cascade can be activated by antibodies through the classical pathway, while injured cells have the potential to initiate complement via the alternative pathway. Once activated, soluble C3a and C5a increase vascular permeability as well as chemotactically attract and activate granulocytes; membrane bound C3b and iC3b increase the attachment of granulocytes and monocyte/macrophages to target cells; and the C5b-C9 membrane attack complex (MAC) aggregates and activates platelets in addition to damaging target cell membranes. C3b and iC3b also modulate developing and ongoing immune responses through complement receptors types l and 2 (CR1 and CR2) on antigen presenting cells, T and B lymphocytes. Complement inhibitors have been more effective than antibody depletion in delaying hyperacute rejection where complement and antibody play a central role. In C6 deficient rats, not only is hyperacute rejection delayed, but survival of allografts in naive recipients is prolonged. These experimental findings are consistent with immunohistological studies of clinical biopsies from acutely rejecting allografts in which deposition of complement components are observed even when little or no antibody is detected. Our working hypothesis is that complement can contribute to multiple aspects of acute and chronic rejection.
Our specific aims are to I) Analyze differences in the rejection reaction in C6 deficient versus normal rats; 2) Analyze differences in rejection of allografts subjected to warm ischemia in C6 deficient versus normal rats; 3) Modify specific complement-triggered effector mechanisms in order to define those mechanisms that contribute to allograft injury; and 4) Analyze differences in complement activation and the rejection reaction in agammaglobulinemic versus normal rats. Because most immunosuppressive protocols have been directed at inhibiting cellular immunity, complement-mediated graft injury may be accentuated in the clinical setting. Recombinant human sCR1 and sCR2 have already been found to delay effectively hyperacute rejection in rats and in preclinical primate trails. If these agents also prove effective in delaying acute rejection, then they could have clinical application in rejection episodes that are associated with complement activation.

Agency
National Institute of Health (NIH)
Institute
National Institute of Allergy and Infectious Diseases (NIAID)
Type
Research Project (R01)
Project #
5R01AI042387-07
Application #
6170773
Study Section
Surgery, Anesthesiology and Trauma Study Section (SAT)
Program Officer
Rose, Stephen M
Project Start
1994-08-01
Project End
2002-07-31
Budget Start
2000-08-01
Budget End
2001-07-31
Support Year
7
Fiscal Year
2000
Total Cost
$303,019
Indirect Cost
Name
Johns Hopkins University
Department
Pathology
Type
Schools of Medicine
DUNS #
045911138
City
Baltimore
State
MD
Country
United States
Zip Code
21218
Wehner, Jennifer R; Fox-Talbot, Karen; Halushka, Marc K et al. (2010) B cells and plasma cells in coronaries of chronically rejected cardiac transplants. Transplantation 89:1141-8
Soong, Thing Rinda; Pathak, Arvind P; Asano, Hiroshi et al. (2010) Lymphatic injury and regeneration in cardiac allografts. Transplantation 89:500-8
Kirk, A D; Morrell, C N; Baldwin 3rd, W M (2009) Platelets influence vascularized organ transplants from start to finish. Am J Transplant 9:14-22
Murata, Kazunori; Baldwin 3rd, William M (2009) Mechanisms of complement activation, C4d deposition, and their contribution to the pathogenesis of antibody-mediated rejection. Transplant Rev (Orlando) 23:139-50
Morrell, Craig N; Murata, Kazunori; Swaim, Anne Marie et al. (2008) In vivo platelet-endothelial cell interactions in response to major histocompatibility complex alloantibody. Circ Res 102:777-85
Lee, Maria Teresa; Parwani, Anil; Humphrey, Richard et al. (2008) Gamma heavy chain disease in a patient with diabetes and chronic renal insufficiency: diagnostic assessment of the heavy chain fragment. J Clin Lab Anal 22:146-50
Murata, Kazunori; Iwata, Takekazu; Nakashima, Shinji et al. (2008) C4d deposition and cellular infiltrates as markers of acute rejection in rat models of orthotopic lung transplantation. Transplantation 86:123-9
Calvert, John W; Gundewar, Susheel; Yamakuchi, Munekazu et al. (2007) Inhibition of N-ethylmaleimide-sensitive factor protects against myocardial ischemia/reperfusion injury. Circ Res 101:1247-54
Morrell, C N; Sun, H; Swaim, A M et al. (2007) Platelets an inflammatory force in transplantation. Am J Transplant 7:2447-54
Asano, Hiroshi; Lee, Chih-Yuan; Fox-Talbot, Karen et al. (2007) Treatment with riboflavin and ultraviolet light prevents alloimmunization to platelet transfusions and cardiac transplants. Transplantation 84:1174-82

Showing the most recent 10 out of 31 publications