Network


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

Hotspot


Dive into the research topics where Christene A. Huang is active.

Publication


Featured researches published by Christene A. Huang.


Journal of Clinical Investigation | 2000

Stable mixed chimerism and tolerance using a nonmyeloablative preparative regimen in a large-animal model

Christene A. Huang; Yasushi Fuchimoto; Rachel Scheier-Dolberg; Michael C. Murphy; David M. Neville; David H. Sachs

Bone marrow transplantation (BMT) has considerable potential for the treatment of malignancies, hemoglobinopathies, and autoimmune diseases, as well as the induction of transplantation allograft tolerance. Toxicities associated with standard preparative regimens for bone marrow transplantation, however, make this approach unacceptable for all but the most severe of these clinical situations. Here, we demonstrate that stable mixed hematopoietic cell chimerism and donor-specific tolerance can be established in miniature swine, using a relatively mild, non-myeloablative preparative regimen. We conditioned recipient swine with whole-body and thymic irradiation, and we depleted their T-cells by CD3 immunotoxin-treatment. Infusion of either bone marrow cells or cytokine-mobilized peripheral blood stem cells from leukocyte antigen-matched animals resulted in stable mixed chimerism, as detected by flow cytometry in the peripheral blood, thymus, and bone marrow, without any clinical evidence of graft-versus-host disease (GvHD). Long-term acceptance of donor skin and consistent rejection of third-party skin indicated that the recipients had developed donor-specific tolerance.


Journal of Clinical Investigation | 2000

Mixed chimerism and tolerance without whole body irradiation in a large animal model

Yasushi Fuchimoto; Christene A. Huang; Kazuhiko Yamada; Akira Shimizu; Hiroshi Kitamura; Robert B. Colvin; Vincent R. Ferrara; Michael C. Murphy; Megan Sykes; M. E. White-Scharf; David M. Neville; David H. Sachs

Mixed hematopoietic chimerism may provide a treatment for patients with nonmalignant hematologic diseases, and may tolerize patients to organ allografts without requiring chronic immunosuppression. However, the toxicity of the usual conditioning regimens has limited the clinical applicability of this approach. These regimens generally include some level of whole body irradiation (WBI), which is thought to facilitate engraftment either by making room for donor hematopoietic stem cells or by providing sufficient host immunosuppression to enable donor cells to engraft. Here, we have established mixed chimerism across both minor and major histocompatibility barriers in swine, by using high doses of peripheral blood stem cells in the absence of WBI. After mixed chimerism was established, swine leukocyte antigen-matched (SLA-matched) donor skin grafts were tolerated and maintained for a prolonged period, whereas third-party SLA-matched skin was rejected promptly. Donor-matched kidney allografts were also accepted without additional immunosuppression. Because of its low toxicity, this approach has potential for a wide range of clinical applications. Our data may indicate that niches for engrafting stem cells are filled by mass action and that WBI, which serves to empty some of these niches, can be omitted if the donor inoculum is sufficiently large and if adequate host T-cell depletion is achieved before transplant.


Transplantation | 2004

Tolerance to composite tissue allografts across a major histocompatibility barrier in miniature swine

Shehan Hettiaratchy; Elizabeth Melendy; Mark A. Randolph; Rebecca C. Coburn; David M. Neville; David H. Sachs; Christene A. Huang; W. P. Andrew Lee

Background. Tolerance to composite tissue allografts might allow the widespread clinical use of reconstructive allotransplantation if protocols to achieve this could be rendered sufficiently nontoxic. The authors investigated whether tolerance could be generated in miniature swine to composite tissue allografts across a major histocompatibility (MHC) barrier. A clinically relevant tolerance protocol involving hematopoietic cell transplantation without the need for irradiation or myelosuppressive drugs was tested. Methods. Seven recipient animals were transiently T-cell depleted and a short course of cyclosporine was initiated. Twenty-four hours later, a donor hematopoietic cell transplant consisting of cytokine-mobilized peripheral blood mononuclear cells or bone marrow cells and a heterotopic limb transplant were performed. In vitro anti-donor responsiveness was assessed by mixed-lymphocyte reaction and cell-mediated lympholysis assays. Acceptance of the limb allografts was determined by gross and histologic appearance. Chimerism in the peripheral blood and lymphohematopoietic organs was assessed by flow cytometry. Results. All seven experimental animals accepted the musculoskeletal elements but rejected the skin of the allografts. All but one of the animals displayed donor-specific unresponsiveness in vitro. The animals that received cytokine mobilized-peripheral blood mononuclear cells showed chimerism but had clinical evidence of graft-versus-host disease (GVHD). None of the animals that received bone marrow cells showed stable chimerism and none developed GVHD. Conclusions. This protocol can achieve tolerance to the musculoskeletal elements of composite tissue allografts across an MHC barrier in miniature swine. Stable chimerism does not appear to be necessary for tolerance and may not be desirable because of the risk of GVHD.


Transplantation | 2002

Pig hematopoietic cell chimerism in baboons conditioned with a nonmyeloablative regimen and CD154 blockade.

L. Bühler; Michel Awwad; S. Treter; Q. Chang; M. Basker; Ian P. J. Alwayn; Katsuhito Teranishi; Thomas Ericsson; K. Moran; David Harper; Barbara Kurilla-Mahon; Christene A. Huang; Robert Sackstein; Megan Sykes; M. E. White-Scharf; David H. Sachs; Julian D. Down; David K. C. Cooper

BACKGROUND In an attempt to induce mixed hematopoietic chimerism and transplantation tolerance in the pig-to-primate model, we have infused high-dose porcine peripheral blood progenitor cells (PBPC) into baboons pretreated with a nonmyeloablative regimen and anti-CD154 monoclonal antibody (mAb). METHODS Group 1 baboons (n=2) received a nonmyeloablative regimen including whole body irradiation, pharmacological immunosuppression, porcine hematopoietic growth factors, and immunoadsorption of anti-Galalpha1,3Gal (Gal) antibody before infusion of high doses of PBPC (2.7-4.6x10(10) cells/kg). In group 2 (n=5), cyclosporine was replaced by anti-CD154 mAb. Group 3 (n=3) received the group 1 regimen plus anti-CD154 mAb. RESULTS In group 1, pig chimerism was detected in the blood by flow cytometry (FACS) for 5 days (with a maximum of 14%), and continuously up to 13 days by polymerase chain reaction (PCR). In group 2, pig chimerism was detectable for 5 days by FACS (maximum 33%) and continuously up to 28 days by PCR. In group 3, initial pig chimerism was detectable for 5 days by FACS (maximum 73%). Two of three baboons showed reappearance of pig cells on days 11 and 16, respectively. In one, in which no anti-Gal IgG could be detected for 30 days, pig cells were documented in the blood by FACS on days 16-22 (maximum 6% on day 19) and pig colony-forming cells were present in the blood on days 19-33, which we interpreted as evidence of engraftment. Microchimerism was continuous by PCR up to 33 days. CONCLUSIONS These results suggest that there is no absolute barrier to pig hematopoietic cell engraftment in primates, and that this may be facilitated if the return of anti-Gal IgG can be prevented.


American Journal of Transplantation | 2014

Vascularized Composite Allograft Tolerance Across MHC Barriers in a Large Animal Model

David A. Leonard; John M. Kurtz; Christopher Mallard; Alexander Albritton; Raimon Duran-Struuck; Evan A. Farkash; Rebecca L. Crepeau; Abraham J. Matar; Benjamin M. Horner; Mark A. Randolph; David H. Sachs; Christene A. Huang; Curtis L. Cetrulo

Vascularized composite allograft (VCA) transplantation can restore form and function following severe craniofacial injuries, extremity amputations or massive tissue loss. The induction of transplant tolerance would eliminate the need for long‐term immunosuppression, realigning the risk–benefit ratio for these life‐enhancing procedures. Skin, a critical component of VCA, has consistently presented the most stringent challenge to transplant tolerance. Here, we demonstrate, in a clinically relevant miniature swine model, induction of immunologic tolerance of VCAs across MHC barriers by induction of stable hematopoietic mixed chimerism. Recipient conditioning consisted of T cell depletion with CD3‐immunotoxin, and 100 cGy total body irradiation prior to hematopoietic cell transplantation (HCT) and a 45‐day course of cyclosporine A. VCA transplantation was performed either simultaneously to induction of mixed chimerism or into established mixed chimeras 85–150 days later. Following withdrawal of immunosuppression both VCAs transplanted into stable chimeras (n = 4), and those transplanted at the time of HCT (n = 2) accepted all components, including skin, without evidence of rejection to the experimental end point 115–504 days posttransplant. These data demonstrate that tolerance across MHC mismatches can be induced in a clinically relevant VCA model, providing proof of concept for long‐term immunosuppression‐free survival.


Transplantation | 1999

In vivo T cell depletion in miniature swine using the swine CD3 immunotoxin, pCD3-CRM9.

Christene A. Huang; Kazuhiko Yamada; Michael C. Murphy; Akira Shimizu; Robert B. Colvin; David M. Neville; David H. Sachs

BACKGROUND Partially inbred miniature swine developed in this laboratory provide a unique preclinical large animal model for studying transplant tolerance. The importance of in vivo T cell depletion for establishing stable mixed hematopoietic cell chimerism using a clinically relevant sublethal regimen has been well documented in murine studies (1). Until now, the lack of an effective in vivo T cell-depleting reagent in swine has limited the progress of studies involving hematopoietic cell transplants. METHODS The swine CD3 immunotoxin, pCD3-CRM9, was prepared by conjugating our porcine-specific CD3 monoclonal antibody 898H2-6-15 to the diphtheria toxin derivative, CRM9. The resultant immunotoxin was administered i.v. to several miniature swine at doses ranging from 0.15-0.2 mg/kg either in a single dose or two doses 2 days apart. T-cell depletion was monitored in the peripheral blood, mesenteric lymph node, and thymus by flow cytometric analysis and histological examination. RESULTS T cells were depleted to less than 1% of their pretreatment levels based on absolute numbers in the peripheral blood. Fluorescence activated cell sorter analysis and histological examination of serial lymph node biopsies confirmed depletion of the CD3+ T cells rather than down modulation or masking of the surface CD3 expression. Depletion of the CD3 bright medullary thymocytes could also be detected by flow cytometry and histological examination after immunotoxin treatment. CONCLUSIONS Administration of the immunotoxin i.v. drastically depletes mature T cells from the peripheral blood, lymph node, and thymus compartments of the pig. This first description of an effective in vivo T-cell depleting reagent for the pig provides a valuable tool for studies of transplant tolerance in this large animal model. It also makes possible preclinical studies of T cell depletion with anti-CD3 immunotoxin in this large animal model.


Transplantation | 2001

Skin-specific alloantigens in miniature swine

Yasushi Fuchimoto; Zachary L. Gleit; Christene A. Huang; Hiroshi Kitamura; Margaret L. Schwarze; Matthew T. Menard; Kwabena Mawulawde; Joren C. Madsen; David H. Sachs

BACKGROUND The acceptance of skin allografts has historically been among the most challenging problems in the field of transplantation, attributed, at least in part, to the existence of antigens expressed by skin but not by other tissues. Many studies have suggested the existence of skin-specific antigens in rodents, but data in large-animal models are more limited. METHODS We have recently developed protocols for attaining stable mixed hematopoietic chimerism in miniature swine, using MHC-matched donors and recipients. We have now assessed tolerance to donor-derived skin and cardiac allografts in these chimeric animals. RESULTS Skin-graft rejection was seen in four of six animals receiving skin grafts taken from the respective hematopoietic donors. In the other two animals, donor-derived skin grafts survived indefinitely. No cardiac-allograft rejection was observed in mixed-chimeric animals that received heart transplants from their hematopoietic donors, even in animals that had already rejected skin allografts from the same donors. In all animals assessed, in vitro hyporesponsiveness to donor hematopoietic cells persisted. CONCLUSION These findings support the concept that skin expresses immunogenic alloantigens that either are not expressed or are not immunogenic in cardiac or hematopoietic tissue.


The Annals of Thoracic Surgery | 2000

Mixed hematopoietic chimerism induces long-term tolerance to cardiac allografts in miniature swine

Margaret L. Schwarze; Matthew T. Menard; Yasushi Fuchimoto; Christene A. Huang; Stuart L. Houser; Kwabena Mawulawde; Kenneth S. Allison; David H. Sachs; Joren C. Madsen

BACKGROUND Tolerance to cardiac allografts has not been achieved in large animals using methods that are readily applicable to human recipients. We investigated the effects of mixed hematopoietic chimerism on cardiac allograft survival and chronic rejection in miniature swine METHODS Recipients were T-cell depleted using a porcine CD3 immunotoxin, and each received either of two nonmyeloablative preparative regimens previously demonstrated to permit the establishment of stable mixed hematopoietic chimerism across MHC-matched, minor antigen-mismatched histocompatibility barriers. Five to 12 months after the chimerism was induced, hearts from the original cell donors were heterotopically transplanted into the stable mixed chimeras. RESULTS Cardiac allografts transplanted into untreated recipients across similar minor antigen barriers were rejected within 44 days (within 21, 28, 35, 39, 44 days among individual study subjects). In contrast, hearts transplanted into the mixed chimeras were all accepted long term ( > 153, > 225, > 286, > 362 days) without immunosuppressive drugs and developed minimal vasculopathy. CONCLUSIONS Mixed hematopoietic chimerism, established in miniature swine using clinically relevant, non-myeloablative conditioning regimens, permits long-term cardiac allograft survival without chronic immunosuppressive therapy, significant vasculopathy, or graft-versus-host disease.


Transplantation Proceedings | 2009

Induction of Tolerance to an Allogeneic Skin Flap Transplant in a Preclinical Large Animal Model

Benjamin M. Horner; Mark A. Randolph; Raimon Duran-Struuck; Erica L. Hirsh; Kelly K. Ferguson; Alexander G.S. Teague; Peter E. Butler; Christene A. Huang

Clinical composite tissue allotransplantation can adequately reconstruct defects that are not possible by other means. However, immunosuppressant toxicity limits the use of these techniques. Clinical attempts to reduce the amount of immunosuppression required by induction of an immunologically permissive state have so far been unsuccessful. The aim of this study was to induce tolerance in a preclinical large animal model. Donor hematopoietic stem cell (HSC) engraftment was induced by T-cell depletion, irradiation, and a short course of cyclosporine administered to the recipient, along with a hematopoietic cell infusion from a single haplotype major histocompatibility complex (MHC) mismatched donor. Skin was then allotransplanted from the donor. Both primarily vascularized skin flaps and secondarily vascularized conventional skin grafts were allotransplanted to investigate if the mode of transplantation affected outcome. Control animals received the skin allotransplants without conditioning. Tolerance was defined as no evidence of rejection at 90 days following transplantation. Conventional skin grafts only achieved prolonged survival (<65 days) in HSC-engrafted animals (P < .01). In contrast, there was indefinite skin flap survival with the achievement of tolerance in HSC-engrafted animals; this was confirmed on histology with donor-specific unresponsiveness on MLR and CML. Furthermore, a conventional skin donor graft subsequently applied to an animal tolerant to a skin flap was not rejected and did not trigger skin flap rejection. To our knowledge, this is the first time skin tolerance has been achieved across a MHC barrier in a large animal model. This is a significant step toward the goal of clinical skin tolerance induction.


Xenotransplantation | 1999

Characterization of a monoclonal anti-porcine CD3 antibody.

Christene A. Huang; Thomas Lorf; J. Scott Arn; Gloria C. Koo; Tom Blake; David H. Sachs

Huang CA, Lorf T, Arn JS, Koo GC, Blake T, Sachs DH. Characterization of a monoclonal anti‐porcine CD3 antibody. Xenotransplantation 1999; 6: 201‐212. © Munksgaard, Copenhagen

Collaboration


Dive into the Christene A. Huang's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge