Olaf Boenisch
Brigham and Women's Hospital
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Olaf Boenisch.
Journal of Clinical Investigation | 2008
Takuya Ueno; Antje Habicht; Michael R. Clarkson; Monica J. Albin; Kazuhiro Yamaura; Olaf Boenisch; Joyce Popoola; Ying Wang; Hideo Yagita; Hisaya Akiba; M. Javeed Ansari; Jaeseok Yang; Laurence A. Turka; David M. Rothstein; Robert F. Padera; Nader Najafian; Mohamed H. Sayegh
T cell Ig mucin 1 (TIM-1) plays an important role in regulating immune responses in autoimmune and asthma models, and it is expressed on both Th1 and Th2 cells. Using an antagonistic TIM-1-specific antibody, we studied the role of TIM-1 in alloimmunity. A short course of TIM-1-specific antibody monotherapy prolonged survival of fully MHC-mismatched vascularized mouse cardiac allografts. This prolongation was associated with inhibition of alloreactive Th1 responses and preservation of Th2 responses. TIM-1-specific antibody treatment was more effective in Th1-type cytokine-deficient Stat4(-/-) recipients as compared with Th2-type cytokine-deficient Stat6(-/-) recipients. Subtherapeutic doses of rapamycin plus TIM-1-specific antibody resulted in allograft acceptance and prevented the development of chronic allograft vasculopathy. Allograft survival via this treatment was accompanied by a Th1- to Th2-type cytokine switch. Depletion of natural Tregs abrogated the graft-protecting effect of the TIM-1-specific antibody. Importantly, CD4(+)CD25(+) Tregs obtained from long-term survivors had enhanced regulatory activity as compared with naive CD4(+)CD25(+) Tregs. Consistent with this, TIM-1-specific antibody treatment both preserved Tregs and prevented the expansion of alloreactive effector Th1 cells in an alloreactive TCR transgenic adoptive transfer model. These studies define previously unknown functions of TIM-1 in regulating alloimmune responses in vivo and may provide a novel approach to promoting transplantation tolerance.
Proceedings of the National Academy of Sciences of the United States of America | 2009
Xueli Yuan; M. Javeed Ansari; Francesca D'Addio; Jesus Paez-Cortez; Isabella Schmitt; Michela Donnarumma; Olaf Boenisch; Xiaozhi Zhao; Joyce Popoola; Michael R. Clarkson; Hideo Yagita; Hisaya Akiba; Gordon J. Freeman; John Iacomini; Laurence A. Turka; Laurie H. Glimcher; Mohamed H. Sayegh
The ability to induce durable transplantation tolerance predictably and consistently in the clinic is a highly desired but elusive goal. Progress is hampered by lack of appropriate experimental models in which to study resistance to transplantation tolerance. Here, we demonstrate that T helper 1-associated T box 21 transcription factor (Tbet) KO recipients exhibit allograft tolerance resistance specifically mediated by IL-17-producing CD8 T (T17) cells. Neutralization of IL-17 facilitates long-term cardiac allograft survival with combined T cell co-stimulation (CD28-CD80/86 and CD154-CD40) blockade in Tbet KO recipients. We have used this T17-biased Tbet KO model of allograft tolerance resistance to study the impact of targeting a T cell-co-stimulatory pathway, and demonstrate that targeting T cell Ig and mucin domain-1 (Tim-1) with anti-Tim-1 overcomes this resistance by specifically inhibiting the pathogenic IL-17-producing CD8 T17 cells. These data indicate that in the absence of Th1 immunity, CD8 T17 alloreactivity constitutes a barrier to transplantation tolerance. Targeting TIM-1 provides an approach to overcome resistance to tolerance in clinical transplantation.
American Journal of Transplantation | 2012
Yohei Yamada; S. Boskovic; A. Aoyama; Toru Murakami; Prabhakar Putheti; R. N. Smith; T. Ochiai; O. Nadazdin; Ichiro Koyama; Olaf Boenisch; Nader Najafian; Manoj Bhasin; Robert B. Colvin; Joren C. Madsen; Terry B. Strom; David H. Sachs; Gilles Benichou; Cosimi Ab; Tatsuo Kawai
The presence of alloreactive memory T cells is a major barrier for induction of tolerance in primates. In theory, delaying conditioning for tolerance induction until after organ transplantation could further decrease the efficacy of the regimen, since preexisting alloreactive memory T cells might be stimulated by the transplanted organ. Here, we show that such “delayed tolerance” can be induced in nonhuman primates through the mixed chimerism approach, if specific modifications to overcome/avoid donor‐specific memory T‐cell responses are provided. These modifications include adequate depletion of CD8+ memory T cells and timing of donor bone marrow administration to minimize levels of proinflammatory cytokines. Using this modified approach, mixed chimerism was induced successfully in 11 of 13 recipients of previously placed renal allografts and long‐term survival without immunosuppression could be achieved in at least 6 of these 11 animals.
Journal of Immunology | 2010
Olaf Boenisch; Francesca D'Addio; Toshihiko Watanabe; Wassim Elyaman; Ciara N. Magee; Melissa Y. Yeung; Robert F. Padera; Scott J. Rodig; Takaya Murayama; Katsunori Tanaka; Xueli Yuan; Takuya Ueno; Anke Jurisch; Bechara Mfarrej; Hisaya Akiba; Hideo Yagita; Nader Najafian
T cell Ig domain and mucin domain (TIM)-3 has previously been established as a central regulator of Th1 responses and immune tolerance. In this study, we examined its functions in allograft rejection in a murine model of vascularized cardiac transplantation. TIM-3 was constitutively expressed on dendritic cells and natural regulatory T cells (Tregs) but only detected on CD4+FoxP3− and CD8+ T cells in acutely rejecting graft recipients. A blocking anti–TIM-3 mAb accelerated allograft rejection only in the presence of host CD4+ T cells. Accelerated rejection was accompanied by increased frequencies of alloreactive IFN-γ–, IL-6–, and IL-17–producing splenocytes, enhanced CD8+ cytotoxicity against alloantigen, increased alloantibody production, and a decline in peripheral and intragraft Treg/effector T cell ratio. Enhanced IL-6 production by CD4+ T cells after TIM-3 blockade plays a central role in acceleration of rejection. Using an established alloreactivity TCR transgenic model, blockade of TIM-3 increased allospecific effector T cells, enhanced Th1 and Th17 polarization, and resulted in a decreased frequency of overall number of allospecific Tregs. The latter is due to inhibition in induction of adaptive Tregs rather than prevention of expansion of allospecific natural Tregs. In vitro, targeting TIM-3 did not inhibit nTreg-mediated suppression of Th1 alloreactive cells but increased IL-17 production by effector T cells. In summary, TIM-3 is a key regulatory molecule of alloimmunity through its ability to broadly modulate CD4+ T cell differentiation, thus recalibrating the effector and regulatory arms of the alloimmune response.
American Journal of Transplantation | 2012
Xiaozhi Zhao; Olaf Boenisch; Melissa Y. Yeung; Bechara Mfarrej; Sunmi Yang; Laurence A. Turka; Mohamed H. Sayegh; John Iacomini; Xueli Yuan
The proinflammatory cytokine IL‐6 plays an important role in controlling T‐cell differentiation, especially the development of Th17 and regulatory T cells. To determine the function of IL‐6 in regulating allograft rejection and tolerance, BALB/c cardiac grafts were transplanted into wild‐type or IL‐6‐deficient C57BL/6 mice. We observed that production of IL‐6 and IFN‐γ was upregulated during allograft rejection in untreated wild‐type mice. In IL‐6‐deficient mice, IFN‐γ production was greater than that observed in wild‐type controls, suggesting that IL‐6 production affects Th1/Th2 balance during allograft rejection. CD28‐B7 blockade by CTLA4‐Ig inhibited IFN‐γ production in C57BL/6 recipients, but had no effect on the production of IL‐6. Although wild‐type C57BL/6 recipients treated with CTLA4‐Ig rejected fully MHC‐mismatched BALB/c heart transplants, treatment of IL‐6‐deficient mice with CTLA4‐Ig resulted in graft acceptance. Allograft acceptance appeared to result from the combined effect of costimulatory molecule blockade and IL‐6‐deficiency, which limited the differentiation of effector cells and promoted the migration of regulatory T cells into the grafts. These data suggest that the blockade of IL‐6, or its signaling pathway, when combined with strategies that inhibit Th1 responses, has a synergistic effect on the promotion of allograft acceptance. Thus, targeting the effects of IL‐6 production may represent an important part of costimulation blockade‐based strategies to promote allograft acceptance and tolerance.
Journal of Immunology | 2013
Lola Chabtini; Bechara Mfarrej; Marwan Mounayar; Bing Zhu; Ibrahim Batal; Pranal J. Dakle; Brian D. Smith; Olaf Boenisch; Nader Najafian; Hisaya Akiba; Hideo Yagita; Indira Guleria
TIM-3 is constitutively expressed on subsets of macrophages and dendritic cells. Its expression on other cells of the innate immune system and its role in fetomaternal tolerance has not yet been explored. In this study, we investigate the role of TIM-3–expressing innate immune cells in the regulation of tolerance at the fetomaternal interface (FMI) using an allogeneic mouse model of pregnancy. Blockade of TIM-3 results in accumulation of inflammatory granulocytes and macrophages at the uteroplacental interface and upregulation of proinflammatory cytokines. Furthermore, TIM-3 blockade inhibits the phagocytic potential of uterine macrophages resulting in a build up of apoptotic bodies at the uteroplacental interface that elicits a local immune response. In response to inflammatory cytokines, Ly-6ChiGneg monocytic myeloid–derived suppressor cells expressing inducible NO synthase and arginase 1 are induced. However, these suppressive cells fail to downregulate the inflammatory cascade induced by inflammatory granulocytes (Ly-6CintGhi) and apoptotic cells; the increased production of IFN-γ and TNF-α by inflammatory granulocytes leads to abrogation of tolerance at the FMI and fetal rejection. These data highlight the interplay between cells of the innate immune system at the FMI and their influence on successful pregnancy in mice.
Journal of The American Society of Nephrology | 2011
Song Rong; Joon-Keun Park; Torsten Kirsch; Hideo Yagita; Hisaya Akiba; Olaf Boenisch; Hermann Haller; Nader Najafian; Antje Habicht
CD4+ T cells contribute to the pathogenesis of ischemia-reperfusion injury, which is the primary cause of delayed graft failure after kidney transplantation. The TIM-1:TIM-4 pathway participates in the activation/differentiation of CD4+ T cells, suggesting that it may modulate ischemia-reperfusion injury. Here, we studied the role of TIM-1 in a murine uninephrectomized renal ischemia-reperfusion injury model. Blocking the TIM-1:TIM-4 pathway with an antagonistic monoclonal antibody protected renal function and diminished reperfusion injury resulting from 30 minutes of ischemia. Histologic examination showed significantly less evidence of renal damage as evidenced by diminished tubular necrosis, preservation of the brush border, fewer cast formations, and less tubular dilation. Blocking TIM-1 also reduced the number of apoptotic cells and diminished local inflammation within ischemic kidneys, the latter shown by decreased recruitment of macrophages, neutrophils, and CD4+ T cells and by reduced local production of proinflammatory cytokines. Furthermore, TIM-1 blockade significantly improved survival after ischemia-reperfusion injury. Taken together, these data suggest that the TIM-1:TIM-4 pathway enhances injury after renal ischemia-reperfusion injury and may be a therapeutic target.
American Journal of Transplantation | 2009
Jun Yang; Leonardo V. Riella; Olaf Boenisch; Joyce Popoola; S. Robles; Toshihiko Watanabe; Vijay K. Vanguri; Xueli Yuan; Indira Guleria; Laurence A. Turka; Mohamed H. Sayegh; Anil Chandraker
Blockade of the B7: CD28 costimulatory pathway has emerged as a promising therapy to prevent allograft rejection. However, this pathway has also been demonstrated to be important for the generation and maintenance of regulatory T cells. In this study, we investigated the role of the B7: CD28 pathway in the ‘bm12 into B6’ MHC class II‐mismatched vascularized cardiac transplant model of chronic rejection. Allograft rejection was remarkably accelerated in B6 background B7DKO and CD28KO recipients compared with B6 wild‐type (WT) recipients. Allograft rejection was associated with a significantly enhanced Th1/Th2 alloreactivity and marked reduction in the ratio of regulatory T cells to CD4+ effector/memory cells. We noted that administration of anti‐B7‐1 and anti‐B7‐2 mAb prior to transplantation also accelerated allograft rejection. Furthermore, depleting CD25+ cells in B6 WT recipients of bm12 hearts prior to transplant also precipitated rejection at a similar rate. Neither B7/CD28 deficiency nor CD25 depletion affected graft survival in single MHC class I‐mismatched (bm1 into B6) recipients. This study highlights the paradoxical functions of B7: CD28 costimulation in a MHC class II‐mismatched model, in which the B7: CD28 pathway is demonstrated to be important in preventing rejection through the generation and maintenance of Tregs.
American Journal of Transplantation | 2012
Olaf Boenisch; M. Lopez; Wassim Elyaman; Ciara N. Magee; U. Ahmad; Nader Najafian
The addition of low, nondepleting doses of rabbit antithymocyte globulin (ATG) to human peripheral blood mononuclear cells has been shown to expand functional CD4+CD25+FoxP3+ regulatory T cells (Tregs) in vitro. This report is the first to elucidate the exact cellular mechanisms of ATG‐mediated Treg expansion. CD4+ T cells require monocytes, but not other antigen presenting cell subsets, to be present in coculture to expand Tregs. However, T cells do not require direct cell–cell contact with monocytes, suggesting the importance of soluble factors. Moreover, ATG initially “reprograms” CD4+ T cells, but not monocytes, and induces STAT3 and STAT5 signaling in CD4+ cells. These reprogrammed CD4+ T cells subsequently secrete GM‐CSF and IL‐10 only in case of intact STAT3 signaling, which in turn promote the generation of tolerogenic CD14+CD11c+ dendritic cells characterized by enhanced IL‐10 and decreased IL‐12 production. Treg expansion following ATG treatment is accompanied by enhanced gene expression of both GM‐CSF and Bcl‐2, but not TGF‐β, in peripheral blood mononuclear cells. These results demonstrate that ex vivo expansion of human Tregs by ATG is due to its ability to reprogram CD4+ T cells in a STAT3‐dependent but TGF‐β‐independent manner, leading to the generation of monocyte‐derived dendritic cells with a tolerogenic cytokine profile.
PLOS ONE | 2013
Francesca D’Addio; Olaf Boenisch; Ciara N. Magee; Melissa Y. Yeung; Xueli Yuan; Bechara Mfarrej; Andrea Vergani; Mohammed Javeed Ansari; Paolo Fiorina; Nader Najafian
Background Despite significant nephrotoxicity, calcineurin inhibitors (CNIs) remain the cornerstone of immunosuppression in solid organ transplantation. We, along with others, have reported tolerogenic properties of anti-thymocyte globulin (ATG, Thymoglobulin®), evinced by its ability both to spare Tregs from depletion in vivo and, when administered at low, non-depleting doses, to expand Tregs ex vivo. Clinical trials investigating B7/CD28 blockade (LEA29Y, Belatacept) in kidney transplant recipients have proven that the replacement of toxic CNI use is feasible in selected populations. Methods Rabbit polyclonal anti-murine thymocyte globulin (mATG) was administered as induction and/or prolonged, low-dose therapy, in combination with CTLA4-Ig, in a stringent, fully MHC-mismatched murine skin transplant model to assess graft survival and mechanisms of action. Results Prolonged, low-dose mATG, combined with CTLA4-Ig, effectively promotes engraftment in a stringent transplant model. Our data demonstrate that mATG achieves graft acceptance primarily by promoting Tregs, while CTLA4-Ig enhances mATG function by limiting activation of the effector T cell pool in the early stages of treatment, and by inhibiting production of anti-rabbit antibodies in the maintenance phase, thereby promoting regulation of alloreactivity. Conclusion These data provide the rationale for development of novel, CNI-free clinical protocols in human transplant recipients.