Sustained long-term antibody levels are the cornerstone of protective immunity, yet the mechanism(s) by which such titers are durably maintained remains unclear. The prevailing paradigm is this occurs via continuous antigen-independent antibody production by a subset of long-lived plasma cells (LLPC) that survive within a limited number of specific niches in the bone marrow (BM). The paradigm's central tenets however, that BM LLPC are a separate subset defined by intrinsically distinct biology and that they in fact contribute to long-term antibody titers, have not been directly demonstrated. We have recently reported (Rozanski et al, JEM July, 2011) that long-term humoral immunity is dependent on plasma cell-intrinsic function of CD28 (best characterized as the prototypic T cell costimulatory receptor), which selectively supports the survival of BM LLPC but not splenic short-lived PC (SLPC). Although LLPC and SLPC both expressed CD28, CD28 signaling and enhanced survival only occurred in the LLPC (and unlike T cells did not require an exogenous signal 1). In vivo, even with sufficient T cell help the loss of CD28 in the plasma cells (or the global loss of the CD28 ligands CD80 or CD86) caused significant loss of the LLPC population, reduction of the half-life of LLPC survival in the BM from 426 to 63 days, and inability to maintain antibody titers long-term - without having any effect on SLPC populations. On the microenvironment/niche side, we found that CD80+ dendritic cells (DC) were in direct contact with LLPC within the BM, that myeloid DC supported long-term LLPC survival in a CD28-dependent fashion in vitro, and that backsignaling via CD80 and CD86 ligation induced the DC to produce the pro-PC survival cytokine IL-6 that was unexpectedly essential for sustaining immunoglobulin (Ig) production in vitro. Altogether, these findings establish the existence of the distinct BM LLPC subset that is necessary to sustain antibody titers, and uncover a previously unrecognized but essential role for PC-intrinsic CD28 function in the longevity of plasma cells and humoral immunity. While previous studies have clearly defined a role for CD28 in both protective and pathogenic (e.g. autoimmune) antibody responses, this has been almost entirely attributed to costimulation of helper T cell activation. Reconsideration of this literature raises he important question of how much of the humoral response is due to CD28 function in T cells vs. plasma cells. Thus we believe that PC-intrinsic CD28 function plays a central role in plasma cell biology and the generation of durable humoral immunity, but has been almost entirely uncharacterized in this context. The overall goal of this proposal is to define the specific mechanisms by which CD28 activation regulates PC survival, function, and the development of humoral immune responses.
The Aims of this proposal are to: 1). Define the signal transduction pathways and molecular targets downstream of CD28 activation that modulate PC survival and function, 2). Characterize the CD80/CD86+ LLPC BM stromal niche, and 3). Characterize the role of PC vs. T cell intrinsic CD28 function in humoral immune responses in mice conditionally knocked out for CD28 in the T or B cell lineage.

Public Health Relevance

Sustained long-term antibody levels are the cornerstone of protective immunity, yet the mechanism(s) by which such titers are durably maintained remains unclear. The prevailing paradigm is this occurs via continuous antigen-independent antibody production by a subset of long-lived plasma cells (LLPC) that survive within a limited number of specific niches within bone marrow (BM). We have found that plasma cell (PC) intrinsic function of CD28 (the prototypic T cell costimulatory receptor) plays a previously unrecognized but essential role in maintaining the survival and function of LLPC, but not short lived PC in the spleen, as well as maintaining durable humoral immunity. We have also found that the CD28 ligands CD80 and CD86 play an essential and non-redundant role in the LLPC bone marrow niche. The Aims of this proposal are to: 1). Define the signal transduction pathways and molecular targets downstream of CD28 activation that modulate PC survival and function, 2). Characterize the CD80/CD86+ LLPC bone marrow stromal niche, and 3). Characterize the role of PC vs. T cell intrinsic CD28 function in humoral immune responses in mice conditionally knocked out for CD28 in the T cell or B cell lineage.

Agency
National Institute of Health (NIH)
Institute
National Institute of Allergy and Infectious Diseases (NIAID)
Type
Research Project (R01)
Project #
4R01AI100157-04
Application #
9115028
Study Section
Cellular and Molecular Immunology - B Study Section (CMIB)
Program Officer
Ferguson, Stacy E
Project Start
2013-08-15
Project End
2017-07-31
Budget Start
2016-08-01
Budget End
2017-07-31
Support Year
4
Fiscal Year
2016
Total Cost
Indirect Cost
Name
Roswell Park Cancer Institute Corp
Department
Type
DUNS #
824771034
City
Buffalo
State
NY
Country
United States
Zip Code
14263
Nair, J R; Caserta, J; Belko, K et al. (2017) Novel inhibition of PIM2 kinase has significant anti-tumor efficacy in multiple myeloma. Leukemia 31:1715-1726
Rozanski, Cheryl H; Utley, Adam; Carlson, Louise M et al. (2015) CD28 Promotes Plasma Cell Survival, Sustained Antibody Responses, and BLIMP-1 Upregulation through Its Distal PYAP Proline Motif. J Immunol 194:4717-28
Fu, Chunmei; Liang, Xinjun; Cui, Weiguo et al. (2015) ?-Catenin in dendritic cells exerts opposite functions in cross-priming and maintenance of CD8+ T cells through regulation of IL-10. Proc Natl Acad Sci U S A 112:2823-8
Murray, Megan E; Gavile, Catherine M; Nair, Jayakumar R et al. (2014) CD28-mediated pro-survival signaling induces chemotherapeutic resistance in multiple myeloma. Blood 123:3770-9
Boise, Lawrence H; Kaufman, Jonathan L; Bahlis, Nizar J et al. (2014) The Tao of myeloma. Blood 124:1873-9
Farren, Matthew R; Carlson, Louise M; Netherby, Colleen S et al. (2014) Tumor-induced STAT3 signaling in myeloid cells impairs dendritic cell generation by decreasing PKC?II abundance. Sci Signal 7:ra16
Koorella, Chandana; Nair, Jayakumar R; Murray, Megan E et al. (2014) Novel regulation of CD80/CD86-induced phosphatidylinositol 3-kinase signaling by NOTCH1 protein in interleukin-6 and indoleamine 2,3-dioxygenase production by dendritic cells. J Biol Chem 289:7747-62