Network


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

Hotspot


Dive into the research topics where Susan Y. Rostami is active.

Publication


Featured researches published by Susan Y. Rostami.


Diabetes | 1997

B-Cells Are Required for the Initiation of Insulitis and Sialitis in Nonobese Diabetic Mice

Hooman Noorchashm; Negin Noorchashm; Kern Jh; Susan Y. Rostami; Clyde F. Barker; Ali Naji

Nonobese diabetic (NOD) mice spontaneously develop an acute onset of hyperglycemia reminiscent of human type I diabetes. The disease is the end result of a mononuclear cell infiltration of pancreatic islets (insulitis), culminating in the selective destruction of islet β-Cells by autoreactive T-cells. NOD mice also exhibit defects in B-cell tolerance as manifested by the presence of autoantibodies against islet cell autoantigens. Based on the potential ability of B-cells to act as antigen presenting cells, we hypothesized that autoreactive B-cells of NOD mice may be necessary for the activation of islet reactive CD4+ T-cells. In the present study, we utilized an anti–μ antibody to induce in vivo depletion of B-cells and found that B-cell depletion completely abrogates the development of insulitis and sialitis in NOD mice. In contrast, control IgG-treated NOD mice developed insulitis and sialitis by 5 weeks of age. Additionally, the discontinuation of anti–μ chain antibody treatment led to the full restoration of the B-cellpool and the reappearance of insulitis and sialitis. Thus, we conclude that B-cells are a requisite cell population for the genesis of the inflammatory lesions of the islets of Langerhans. This finding suggests that autoreactive B-cells may act as the antigen presenting cells necessary for the initial activation of β-cell-reactive CD4+ T-cells implicated in the pathogenesis of autoimmune diabetes.


Nature Medicine | 2007

B lymphocyte–directed immunotherapy promotes long-term islet allograft survival in nonhuman primates

Chengyang Liu; Hooman Noorchashm; Jennifer A. Sutter; Mina Naji; Eline T. Luning Prak; Jean D. Boyer; Taryn Green; Michael R. Rickels; John E. Tomaszewski; Brigitte Koeberlein; Zhonglin Wang; Michelle Paessler; Ergun Velidedeoglu; Susan Y. Rostami; Ming Yu; Clyde F. Barker; Ali Naji

We found that an induction immunotherapy regimen consisting of rabbit anti-thymocyte globulin (Thymoglobulin) and the monoclonal antibody to CD20 rituximab (Rituxan) promoted long-term islet allograft survival in cynomolgus macaques maintained on rapamycin monotherapy. B lymphocyte reconstitution after rituximab-mediated depletion was characterized by a preponderance of immature and transitional cells, whose persistence was associated with long-term islet allograft survival. Development of donor-specific alloantibodies was abrogated only in the setting of continued rapamycin monotherapy.


Journal of Immunology | 2006

B Cell-Mediated Antigen Presentation Is Required for the Pathogenesis of Acute Cardiac Allograft Rejection

Hooman Noorchashm; Amy J. Reed; Susan Y. Rostami; Raha Mozaffari; Ghazal Zekavat; Brigitte Koeberlein; Andrew J. Caton; Ali Naji

Acute allograft rejection requires the activation of alloreactive CD4 T cells. Despite the capacity of B cells to act as potent APCs capable of activating CD4 T cells in vivo, their role in the progression of acute allograft rejection was unclear. To determine the contribution of B cell APC function in alloimmunity, we engineered mice with a targeted deficiency of MHC class II-mediated Ag presentation confined to the B cell compartment. Cardiac allograft survival was markedly prolonged in these mice as compared to control counterparts (median survival time, >70 vs 9.5 days). Mechanistically, deficient B cell-mediated Ag presentation disrupted both alloantibody production and the progression of CD4 T cell activation following heart transplantation. These findings demonstrate that indirect alloantigen presentation by recipients’ B cells plays an important role in the efficient progression of acute vascularized allograft rejection.


Journal of Immunology | 2001

Impaired Activation of Islet-Reactive CD4 T Cells in Pancreatic Lymph Nodes of B Cell-Deficient Nonobese Diabetic Mice

Siri Atma W. Greeley; Daniel J. Moore; Hooman Noorchashm; Lauren E. Noto; Susan Y. Rostami; Alexander Schlachterman; Howard K. Song; Brigitte Koeberlein; Clyde F. Barker; Ali Naji

Despite the impressive protection of B cell-deficient (μMT−/−) nonobese diabetic (NOD) mice from spontaneous diabetes, existence of mild pancreatic islet inflammation in these mice indicates that initial autoimmune targeting of β cells has occurred. Furthermore, μMT−/− NOD mice are shown to harbor a latent repertoire of diabetogenic T cells, as evidenced by their susceptibility to cyclophosphamide-induced diabetes. The quiescence of this pool of islet-reactive T cells may be a consequence of impaired activation of T lymphocytes in B cell-deficient NOD mice. In this regard, in vitro anti-CD3-mediated stimulation demonstrates impaired activation of lymph node CD4 T cells in μMT−/− NOD mice as compared with that of wild-type counterparts, a deficiency that is correlated with an exaggerated CD4 T cell:APC ratio in lymph nodes of μMT−/− NOD mice. This feature points to an insufficient availability of APC costimulation on a per T cell basis, resulting in impaired CD4 T cell activation in lymph nodes of μMT−/− NOD mice. In accordance with these findings, an islet-reactive CD4 T cell clonotype undergoes suboptimal activation in pancreatic lymph nodes of μMT−/− NOD recipients. Overall, the present study indicates that B cells in the pancreatic lymph node microenvironment are critical in overcoming a checkpoint involving the provision of optimal costimulation to islet-reactive NOD CD4 T cells.


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.


Journal of Immunology | 2003

Alloreactive CD4 T cell activation in vivo: an autonomous function of the indirect pathway of alloantigen presentation.

Amy J. Reed; Hooman Noorchashm; Susan Y. Rostami; Yasaman Zarrabi; Alison R. Perate; Arjun N. Jeganathan; Andrew J. Caton; Ali Naji

Activation of alloreactive CD4 T cells occurs via the direct and indirect pathways of alloantigen presentation. A novel TCR/alloantigen transgenic system was designed that permitted in vivo visualization of CD4 T cell priming through these pathways. When both pathways of alloantigen presentation were intact, CD4 T cell activation in response to cardiac allografts was rapid and systemic by day 4 after transplantation, in contrast to that seen in response to skin allografts, which was delayed until 10–12 days after transplantation. Despite this systemic CD4 T cell activation in response to cardiac allografts, there was a paucity of activated graft-infiltrating CD4 T cells at 4 days posttransplantation. This finding suggests that the initial priming of alloimmune CD4 T cell responses occurs within draining lymphoid organs. Furthermore, alloantigens derived from cardiac allografts failed to promote thymic negative selection of developing thymocytes expressing the alloreactive TCR clonotype. In the absence of a functional direct pathway, the kinetics of activation, anatomic localization, and effector function of alloreactive CD4 T cells remained unchanged. Overall, the present study defines the anatomic and temporal characteristics of CD4 T cell alloimmune responses and demonstrates that CD4 T cell priming via the indirect pathway proceeds optimally in the absence of the direct pathway of alloantigen presentation.


Transplantation | 1998

Utility of adenoviral-mediated Fas ligand gene transfer to modulate islet allograft survival

Thomas A. Judge; Niraj M. Desai; Z. Yang; Susan Y. Rostami; Laura C. Alonso; Haidi Zhang; Youhai H. Chen; J. F. Markman; R. P. DeMateo; Clyde F. Barker; Ali Naji; Laurence A. Turka

BACKGROUND One of the best-defined mechanisms for the induction of apoptosis involves signaling via the cell surface molecule Fas, after binding of Fas ligand. Expression of Fas ligand is tightly regulated, being expressed primarily by T cells after activation, where it serves as a self-regulatory mechanism for immune responses. Fas ligand has also been found to be expressed constitutively at sites of immune privilege such as the testes and the anterior chamber of the eye. Recently, co-transplantation of Fas ligand-transfected myoblasts in association with islet cell allografts was shown to prolong islet allograft survival but only rarely led to indefinite graft survival. Graft rejection was associated with loss of Fas ligand on the myoblasts, suggesting that direct expression of the transgene on the islets might be more effective. METHODS A replication-defective adenoviral construct containing murine Fas ligand (Ad/MFL) was prepared by homologous recombination. NIH 3T3 cells, rodent splenocytes, and murine islets were infected with Ad/MFL and examined in vitro for functional murine Fas ligand expression. Survival of Ad/MFL-infected islets was subsequently evaluated in vivo in both syngeneic and allogeneic islet transplantation models. RESULTS Cell lines and islet allografts transfected with Ad/MFL expressed a functional Fas ligand, capable of inducing apoptosis (confirmed by three distinct assays for DNA fragmentation) in Fas+ targets, but not in Fas- controls. Furthermore, Ad/MFL was able to modify allogeneic immune responses in vitro, as addition of this virus, but not a control adenovirus, significantly reduced proliferation in a mixed lymphocyte reaction. Surprisingly, however, transplantation of islet allografts transfected with Ad/MFL resulted in long-term allograft survival in only 1 of 30 recipients. Moreover, adenoviral-mediated Fas ligand gene transfer was complicated by transient, dose-dependent islet dysfunction, perhaps contributing to the lack of long-term engraftment. CONCLUSION These data suggest that adenoviral-mediated Fas ligand expression may impair normal islet function in vivo, and indicate that alternative strategies for Fas ligand transgene delivery may be required in this setting.


Transplantation | 1999

Cardiac allograft tolerance induced by intra-arterial infusion of recombinant adenoviral CTLA4Ig.

Zandong Yang; Susan Y. Rostami; Brigitte Koeberlein; Clyde F. Barker; Ali Naji

BACKGROUND Systemic administration of soluble recombinant fusion protein of cytotoxic T lymphocyte antigen 4 (CTLA4Ig) induces blockade of the CD28/B7 costimulatory pathway and promotes survival of allogeneic and xenogeneic grafts. We tested the efficacy of local expression of CTLA4Ig gene in the myocardium, induced by transduction with a recombinant adenovirus encoding the CTLA4Ig gene, on the survival of rat cardiac allografts. METHODS The donor hearts were perfused ex vivo with recombinant adenovirus encoding CTLA4Ig cDNA (AdCTLA4Ig) via intra-aorta coronary artery before transplantation. The distribution and duration of CTLA4Ig transgene expression in the myocardium was assessed by reverse transcriptase polymerase chain reaction (RT-PCR) or in situ RT-PCR after transplantation. RESULTS In situ RT-PCR demonstrated abundant expression of CTLA4Ig transgene in the endo-myocardium of AdCTLA4Ig-perfused cardiac grafts. Lewis and Brown Norway cardiac allografts transduced with AdCTLA4Ig survived indefinitely in nonimmunosuppressed Wistar Furth recipients. However, donor-strain skin grafts were rejected by long-term recipients of cardiac allografts, which also triggered the rejection of the primary heart grafts. CONCLUSIONS A single ex vivo intra-aortic infusion of recombinant adenovirus encoding the CTLA4Ig gene induced efficient transduction of the endo-myocardium and promoted the permanent survival of cardiac allografts in nonimmunosuppressed hosts. Despite the beneficial effect of local immunosuppression on cardiac allograft survival, the strategy failed to promote a state of donor-specific peripheral tolerance.


Transplantation | 1999

A direct method for the calculation of alloreactive CD4+ T cell precursor frequency

Hooman Noorchashm; Yen K. Lieu; Susan Y. Rostami; Howard K. Song; Siri Atma S. Greeley; Samaneh Bazel; Clyde F. Barker; Ali Naji

BACKGROUND Direct measurement of the precursor frequency of alloreactive CD4+ T cells has been impossible due to the lack of a specific means of determining the absolute number of daughter cells generated with each division in a repertoire of stimulated T cells. METHODS Responder lymphocytes were fluorescently labeled and adoptively transferred into irradiated allogeneic stimulator mice or incubated in vitro with irradiated stimulator splenocytes. After a 65- to 70-hr stimulation period, responder cells were analyzed by flow cytometry. RESULTS The precursor frequency of dividing CD4+ T cells was determined both in vivo and in vitro. The observed number of alloreactive daughter cells generated with each round of division was used to calculate the frequency of alloantigen-specific CD4+ T cells. CONCLUSIONS A novel method for the direct calculation of the frequency of alloreactive CD4+ T cells is described. This technique allows the determination of changes in the frequency of alloreactive T cells that might underlie tolerance to alloantigens.


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.

Collaboration


Dive into the Susan Y. Rostami'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

Clyde F. Barker

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Howard K. Song

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Ronald F. Parsons

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Yen K. Lieu

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Ghazal Zekavat

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kumar Vivek

University of Pennsylvania

View shared research outputs
Researchain Logo
Decentralizing Knowledge