Network


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

Hotspot


Dive into the research topics where Ronald F. Parsons is active.

Publication


Featured researches published by Ronald F. Parsons.


Journal of Immunology | 2008

In Vivo BLyS/BAFF Neutralization Ameliorates Islet-Directed Autoimmunity in Nonobese Diabetic Mice

Ghazal Zekavat; Susan Y. Rostami; Armen Badkerhanian; Ronald F. Parsons; Brigitte Koeberlein; Ming Yu; Christopher D. Ward; Thi-Sau Migone; Liping Yu; George S. Eisenbarth; Michael P. Cancro; Ali Naji; Hooman Noorchashm

B lymphocytes are required for the pathogenesis of autoimmune diabetes in NOD mice. Previous studies established that a lymphopenic transitional (TR) B cell compartment reduces the competitive constraint on the entry of newly emerging TR B cells into the splenic follicle (FO), thereby disrupting a peripheral negative selection checkpoint in NOD mice. Thus, development of clinically feasible immunotherapeutic approaches for restoration of appropriate negative selection is essential for the prevention of anti-islet autoimmunity. In this study we hypothesized that in vivo neutralization of the B lymphocyte stimulator (BLyS/BAFF) may enhance the stringency of TR→FO selection by increasing TR B cell competition for follicular entry in NOD mice. This study demonstrated that in vivo BLyS neutralization therapy leads to the depletion of follicular and marginal zone B lymphocytes. Long-term in vivo BLyS neutralization caused an increased TR:FO B cell ratio in the periphery indicating a relative resistance to follicular entry. Moreover, in vivo BLyS neutralization: 1) restored negative selection at the TR→FO checkpoint, 2) abrogated serum insulin autoantibodies, 3) reduced the severity of islet inflammation, 4) significantly reduced the incidence of spontaneous diabetes, 5) arrested the terminal stages of islet cell destruction, and 6) disrupted CD4 T cell activation in NOD mice. Overall, this study demonstrates the efficacy of B lymphocyte-directed therapy via in vivo BLyS neutralization for the prevention of autoimmune diabetes.


Current Opinion in Immunology | 2011

Essential role for B cells in transplantation tolerance

Robert R. Redfield; Eduardo Rodriguez; Ronald F. Parsons; Kumar Vivek; Moiz M. Mustafa; Hooman Noorchashm; Ali Naji

T lymphocytes are the primary targets of immunotherapy in clinical transplantation. However, B lymphocytes are detrimental to graft survival by virtue of their capacity to present antigen to T cells via the indirect pathway of allorecognition and the generation of donor specific alloantibody. Furthermore, the long-term survival of organ allografts remains challenged by chronic rejection, a process in which activated B cells have been found to play a significant role. Therefore, the achievement of transplantation tolerance will likely require induction of both T and B cell tolerance to alloantigens. Moreover, human and animal investigations have shown that subsets of B cells, Transitional and Regulatory, are inherently tolerogenic. Developing therapeutic strategies that exploit these populations may be key to achieving transplantation tolerance. In this review we describe the current evidence for the essential role of B cells in transplant tolerance and discuss emerging B cell directed strategies to achieve allograft tolerance.


Immunologic Research | 2008

Homeostatic control of B lymphocyte subsets

Jenni E. Crowley; Jean L. Scholz; William J. Quinn; Jason Stadanlick; John Treml; Laura S. Treml; Yi Hao; Radhika Goenka; Patrick J. O’Neill; Andrew H. Matthews; Ronald F. Parsons; Michael P. Cancro

Lymphocyte homeostasis poses a multi-faceted biological puzzle, because steady pre-immune populations must be maintained at an acceptable steady state to yield effective protection, despite stringent selective events during their generation. In addition, activated, memory and both short- and long-term effectors must be governed by independent homeostatic mechanisms. Finally, advancing age is accompanied by substantial changes that impact the dynamics and behavior of these pools, leading to cumulative homeostatic perturbations and compensation. Our laboratory has focused on the overarching role of BLyS family ligands and receptors in these processes. These studies have led to a conceptual framework within which distinct homeostatic niches are specified by BLyS receptor signatures, which define the BLyS family ligands that can afford survival. The cues for establishing these receptor signatures, as well as the downstream survival mechanisms involved, are integrated with cell extrinsic inputs via cross talk among downstream mediators. A refined understanding of these relationships should yield insight into the selection and maintenance of B cell subsets, as well as an appreciation of how homeostatic mechanisms may contribute to immunosenescence.


Journal of Immunology | 2011

Acquisition of Humoral Transplantation Tolerance upon De Novo Emergence of B Lymphocytes

Ronald F. Parsons; Kumar Vivek; Susan Y. Rostami; Ghazal Zekavat; Seyed Ziaie; Yanping Luo; Brigitte Koeberlein; Robert R. Redfield; Michael P. Cancro; Ali Naji; Hooman Noorchashm

A major obstacle to transplantation tolerance is humoral immunity. In this paper, we demonstrate that the intrinsic developmental propensity of the B lymphocyte compartment for acquisition of self-tolerance can be harnessed to induce humoral unresponsiveness to transplanted alloantigens. In the current study, when transitional B cells developed in the presence of donor lymphoid cells, the mature B lymphocyte compartment failed to mount a donor-specific alloantibody response to an organ transplant—despite unrestrained acute T cell-mediated allograft rejection. Specifically, we generated an experimental system wherein a B6 strain B cell compartment developed de novo in the presence of F1 (B6xBALB/c) lymphoid cells and in a T cell-deficient setting. Following establishment of a steady-state B cell compartment, these B6 mice were transplanted with heterotopic cardiac allografts from allogeneic BALB/c donors. The mice were then inoculated with purified syngeneic B6 T cells. As expected, all cardiac allografts were acutely rejected. However, the B lymphocyte compartment of these mice was completely inert in its capacity to form a BALB/c-specific alloantibody response. Using an alloantigen-specific Ig transgenic system, we demonstrated that this profound degree of humoral tolerance was caused by clonal deletion of alloreactive specificities from the primary B cell repertoire. Thus, de novo B cell compartment development at the time of transplantation is of critical importance in recipient repertoire “remodeling” to a humoral tolerant state.


Transplantation | 2012

Murine islet allograft tolerance upon blockade of the B-lymphocyte stimulator, BLyS/BAFF.

Ronald F. Parsons; Ming Yu; Kumar Vivek; Ghazal Zekavat; Susan Y. Rostami; Amin Sam Ziaie; Yanping Luo; Brigitte Koeberlein; Robert R. Redfield; Christopher D. Ward; Thi Sau Migone; Michael P. Cancro; Ali Naji; Hooman Noorchashm

Background. Immunologic rejection is a major barrier to successful long-term outcomes in clinical transplantation. The importance of B lymphocytes—and their secretory products, alloantibodies—in the pathogenesis of allograft rejection is accepted. Furthermore, it is now clear that the dominant regulator of peripheral B-cell homeostasis and tolerance is the B-Lymphocyte Stimulator (BLyS), also referred to as the B-cell activating factor (BAFF). Recently, a novel class of clinical immunotherapeutic agents specific for BLyS, and its family of cytokines, has emerged for the treatment of B-cell-mediated diseases. In this study, we demonstrate the potential utility of BLyS-directed immunotherapy in preventing allograft rejection using a murine islet transplantation model. Methods. A transient period of mature peripheral B-cell depletion was induced by means of in vivo BLyS neutralization using a murine analog of the monoclonal antibody, Benlysta. Subsequently, fully major histocompatibility complex-mismatched islets were transplanted into naïve diabetic mice followed by a short course of rapamycin. Results. After BLyS neutralization, indefinite islet allograft survival was achieved. Induction therapy with rapamycin was necessary, but not sufficient, for the achievement of this long-term graft survival. The tolerant state was associated with (1) abrogation of the donor-specific antibody response, (2) transient preponderance of immature/transitional B cells in all lymphoid organs, (3) impaired CD4 T-cell activation during the period of B-cell depletion, and (4) presence of a “regulatory” cytokine milieu. Conclusions. In vivo BLyS neutralization effectively induces humoral tolerance and promotes long-term islet allograft survival in mice. Therefore, B-lymphocyte-directed immunotherapy targeting the homeostatic regulator, BLyS, may be effective in promoting transplantation tolerance.


Expert Review of Clinical Immunology | 2009

B-cell tolerance in transplantation: is repertoire remodeling the answer?

Ronald F. Parsons; Kumar Vivek; Robert R. Redfield; Thi Sau Migone; Michael P. Cancro; Ali Naji; Hooman Noorchashm

T lymphocytes are the primary targets of immunotherapy in clinical transplantation; however, B-lymphocytes and their secreted alloantibodies are also highly detrimental to the allograft. Therefore, the achievement of sustained organ transplant survival will likely require the induction of B-lymphocyte tolerance. During development, acquisition of B-cell tolerance to self-antigens relies on clonal deletion in the early stages of B-cell compartment ontogeny. We contend that this mechanism should be recapitulated in the setting of alloantigens and organ transplantation to eliminate the alloreactive B-cell subset from the recipient. Clinically feasible targets of B-cell-directed immunotherapy, such as CD20 and B-lymphocyte stimulator (BLyS), should drive upcoming clinical trials aimed at remodeling the recipient B-cell repertoire.


Seminars in Immunology | 2012

Primary B cell repertoire remodeling to achieve humoral transplantation tolerance

Ronald F. Parsons; Robert R. Redfield; Eduardo Rodriguez; Moiz M. Mustafa; Kumar Vivek; Michael Murayama; Ali Naji; Hooman Noorchashm

The current mainstay of immunotherapy in clinical transplantation is T lymphocyte directed. However, it has long been appreciated that the emergence of an alloimmune response mounted by the B lymphocyte compartment and detectable as donor-specific antibodies is a critical challenge to long-term graft survival. Thus, achieving robust transplantation tolerance will require induction of tolerance in both the T- and B-cell compartments. Here we propose that the natural developmental propensity of the B-lymphocyte compartment acquisition of tolerance to self-antigens can be recapitulated to achieve humoral transplantation tolerance. It is our contention B-lymphocyte directed induction immunotherapy would be an important component of emerging strategies for induction of Transplantation tolerance.


Clinical Transplantation | 2012

Underutilization of A2 ABO incompatible kidney transplantation.

Robert R. Redfield; Ronald F. Parsons; Eduardo Rodriguez; Moiz M. Mustafa; James Cassuto; Kumar Vivek; Hooman Noorchashm; Ali Naji; Matthew H. Levine; Peter L. Abt

Redfield RR, Parsons RF, Rodriguez E, Mustafa M, Cassuto J, Vivek K, Noorchashm H, Naji A, Levine MH, Abt PL. Underutilization of A2 ABO incompatible kidney transplantation. 
Clin Transplant 2011 DOI: 10.1111/j.1399‐0012.2011.01543.x. 
© 2011 John Wiley & Sons A/S.


Transplant Immunology | 2013

Sustained reduction of alloantibody secreting plasma cells and donor specific antibody with proteasome inhibition in mice.

Robert R. Redfield; Y. Lou; Eduardo Rodriguez; Susan Y. Rostami; Ronald F. Parsons; Hooman Noorchashm; Ali Naji; Peter L. Abt

The long-lived plasma cells, which develop after alloantigen sensitization, produce donor specific alloantibodies (DSAs) that generate a positive serum cross-match and preclude transplantation. Bortezomib, a proteasome inhibitor, is being investigated in clinical desensitization protocols, however preclinical studies in a transplant model are nonexistent. We hypothesized that sustained treatment with only a proteasome inhibitor would eliminate plasma cells and reduce DSA over time. Cardiac allografts were transplanted into murine recipients. Eight weeks after allograft rejection the proteasome inhibitor, bortezomib, was injected intravenously twice weekly for 60 days. Serum alloantibody responses were assayed using flow cross-match. Total and alloreactive plasma cell numbers were enumerated using flow cytometry and ELISPOT. All recipients of cardiac allografts rejected their graft promptly within 16 days and demonstrated alloantibody by flow cross-match. DSA was sustained in the control mice while mice treated with bortezomib had sustained elimination of DSA and a marked reduction in plasma cell population. Also, bortezomib was associated with an increased level of BLyS. Within a murine model, proteasome inhibition can eliminate alloantibody secreting plasma cells, and reduce alloantibody. Cessation of bortezomib is not associated with return of DSA.


Immunologic Research | 2011

Strategies for B-lymphocyte repertoire remodeling in transplantation tolerance.

Kumar Vivek; Moiz M. Mustafa; Eduardo Rodriguez; Robert R. Redfield; Ronald F. Parsons; Susan Y. Rostami; Thi Sau Migone; Michael P. Cancro; Ali Naji; Hooman Noorchashm

Transplantation tolerance remains an elusive goal as B-cell-initiated chronic humoral rejection evades current immunosuppression. B-cell-directed therapy is thus emerging as a key component in achieving transplantation tolerance and long-term graft survival. Here, we propose strategies of B-cell repertoire remodeling to achieve humoral unresponsiveness to donor antigens with implementation of fundamental B-cell immunobiology and use of newly developed B-cell-directed agents.

Collaboration


Dive into the Ronald F. Parsons's collaboration.

Top Co-Authors

Avatar

Ali Naji

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Hooman Noorchashm

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kumar Vivek

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Michael P. Cancro

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Susan Y. Rostami

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Eduardo Rodriguez

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Peter L. Abt

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Moiz M. Mustafa

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge