1
|
Humanization of Immunodeficient Animals for the Modeling of Transplantation, Graft Versus Host Disease, and Regenerative Medicine. Transplantation 2021; 104:2290-2306. [PMID: 32068660 PMCID: PMC7590965 DOI: 10.1097/tp.0000000000003177] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The humanization of animals is a powerful tool for the exploration of human disease pathogenesis in biomedical research, as well as for the development of therapeutic interventions with enhanced translational potential. Humanized models enable us to overcome biologic differences that exist between humans and other species, while giving us a platform to study human processes in vivo. To become humanized, an immune-deficient recipient is engrafted with cells, tissues, or organoids. The mouse is the most well studied of these hosts, with a variety of immunodeficient strains available for various specific uses. More recently, efforts have turned to the humanization of other animal species such as the rat, which offers some technical and immunologic advantages over mice. These advances, together with ongoing developments in the incorporation of human transgenes and additional mutations in humanized mouse models, have expanded our opportunities to replicate aspects of human allotransplantation and to assist in the development of immunotherapies. In this review, the immune and tissue humanization of various species is presented with an emphasis on their potential for use as models for allotransplantation, graft versus host disease, and regenerative medicine.
Collapse
|
2
|
Indirectly Activated Treg Allow Dominant Tolerance to Murine Skin-grafts Across an MHC Class I Mismatch After a Single Donor-specific Transfusion. Transplantation 2020; 104:1385-1395. [PMID: 32053573 DOI: 10.1097/tp.0000000000003173] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Tolerance induced in stringent animal transplant models using donor-specific transfusions (DST) has previously required additional immunological manipulation. Here, we demonstrate a dominant skin-allograft tolerance model induced by a single DST across an major histocompatibility class I mismatch in an unmanipulated B6 host. METHODS C57BL/6 (H-2) (B6) mice were injected intravenously with splenocytes from B6.C.H-2 (H-2k) (bm1) or F1 (B6 × bm1) mice before skin transplantation. Mice were transplanted 7 days postinjection with donor (bm1 or F1) and third-party B10.BR (H-2) skin grafts. RESULTS B6 hosts acutely rejected skin grafts from B6.C.H-2 (bm1) and F1 (B6 × bm1) mice. A single transfusion of F1 splenocytes into B6 mice without any additional immune modulation led to permanent acceptance of F1 skin grafts. This graft acceptance was associated with persistence of donor cells long-term in vivo. The more rapid removal of DST bm1 cells than F1 cells was reduced by natural killer-cell depletion. Tolerant grafts survived an in vivo challenge with naive splenocytes. Both CD4CD25 and CD4CD25 T cells from F1 DST treated B6 mice suppressed alloproliferation in vitro. Tolerance was associated with expansion of peripheral Foxp3CD4CD25 regulatory T cells (Treg) and increased forkhead box P3 (Foxp3) expression in tolerant grafts. In tolerant mice, Foxp3 Treg arises from the proliferation of indirectly activated natural Foxp3 Treg (nTreg) and depletion of Foxp3 Treg abrogates skin-graft tolerance. CONCLUSIONS This study demonstrates that the persistence of transfused semiallogeneic donor cells mismatched at major histocompatibility class I can enhance tolerance to subsequent skin allografts through indirectly expanded nTreg leading to dominant tolerance without additional immunological manipulation.
Collapse
|
3
|
|
4
|
Saetersmoen ML, Hammer Q, Valamehr B, Kaufman DS, Malmberg KJ. Off-the-shelf cell therapy with induced pluripotent stem cell-derived natural killer cells. Semin Immunopathol 2018; 41:59-68. [PMID: 30361801 DOI: 10.1007/s00281-018-0721-x] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 09/28/2018] [Indexed: 12/15/2022]
Abstract
Cell therapy is emerging as a very promising therapeutic modality against cancer, spearheaded by the clinical success of chimeric antigen receptor (CAR) modified T cells for B cell malignancies. Currently, FDA-approved CAR-T cell products are based on engineering of autologous T cells harvested from the patient, typically using a central manufacturing facility for gene editing before the product can be delivered to the clinic and infused to the patients. For a broader implementation of advanced cell therapy and to reduce costs, it would be advantageous to use allogeneic "universal" cell therapy products that can be stored in cell banks and provided upon request, in a manner analogous to biopharmaceutical drug products. In this review, we outline a roadmap for development of off-the-shelf cell therapy based on natural killer (NK) cells derived from induced pluripotent stem cells (iPSCs). We discuss strategies to engineer iPSC-derived NK (iPSC-NK) cells for enhanced functional potential, persistence, and homing.
Collapse
Affiliation(s)
| | - Quirin Hammer
- Department of Medicine, Huddinge, Karolinska Institute, Solna, Sweden
| | | | - Dan S Kaufman
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Karl-Johan Malmberg
- The KG Jebsen Center for Cancer Immunotherapy, University of Oslo, Oslo, Norway. .,Department of Medicine, Huddinge, Karolinska Institute, Solna, Sweden. .,Institute for Cancer research, Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
5
|
Rodriguez-Barbosa JI, Ferreras MC, Buhler L, Jones ND, Schneider P, Perez-Simon JA, Del Rio ML. Therapeutic implications of NK cell regulation of allogeneic CD8 T cell-mediated immune responses stimulated through the direct pathway of antigen presentation in transplantation. MAbs 2018; 10:1030-1044. [PMID: 30036156 PMCID: PMC6204794 DOI: 10.1080/19420862.2018.1502127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Natural killer (NK) cells are a population of innate type I lymphoid cells essential for early anti-viral responses and are known to modulate the course of humoral and cellular-mediated T cell responses. We assessed the role of NK cells in allogeneic CD8 T cell-mediated responses in an immunocompetent mouse model across an MHC class I histocompatibility barrier to determine its impact in therapeutic clinical interventions with polyclonal or monoclonal antibodies (mAbs) targeting lymphoid cells in transplantation. The administration of an NK cell depleting antibody to either CD8 T cell replete or CD8 T cell-depleted naïve C57BL/6 immunocompetent mice accelerated graft rejection. This accelerated rejection response was associated with an in vivo increased cytotoxic activity of CD8 T cells against bm1 allogeneic hematopoietic cells and bm1 skin allografts. These findings show that NK cells were implicated in the control host anti-donor cytotoxic responses, likely by competing for common cell growth factors in both CD8 T cell replete and CD8 T cell-depleted mice, the latter reconstituting in response to lymphopenia. Our data calls for precaution in solid organ transplantation under tolerogenic protocols involving extensive depletion of lymphocytes. These pharmacological biologics with depleting properties over NK cells may accelerate graft rejection and promote aggressive CD8 T cell cytotoxic alloresponses refractory to current immunosuppression.
Collapse
Affiliation(s)
- J I Rodriguez-Barbosa
- a Transplantation Immunobiology Section , University of León , Leon , Spain.,h Acción Estratégica en Salud , Consorcio CIBER-ONC, Seville, Spain
| | - M C Ferreras
- b Department of Animal Health, Mountain Livestock Institute (CSIC), School of Veterinary Sciences , University of Leon , Leon , Spain
| | - L Buhler
- c Visceral and Transplantation Surgery, Department of Surgery , University Hospitals of Geneva and Faculty of Medicine , Geneva , Switzerland
| | - N D Jones
- d MRC Centre of Immune Regulation, School of Immunity and Infection, Medical School , University of Birmingham , Birmingham , United Kingdom
| | - P Schneider
- e Department of Biochemistry , University of Lausanne , Epalinges , Switzerland
| | - J A Perez-Simon
- f Department of Hematology , University Hospital Virgen del Rocio/Institute of Biomedicine (IBIS/CSIC) , Sevilla , Spain.,h Acción Estratégica en Salud , Consorcio CIBER-ONC, Seville, Spain
| | - M L Del Rio
- a Transplantation Immunobiology Section , University of León , Leon , Spain.,g Leon Regional Transplantation Coordination Center , Leon University Hospital , Leon , Spain.,h Acción Estratégica en Salud , Consorcio CIBER-ONC, Seville, Spain
| |
Collapse
|
6
|
Norero B, Serrano CA, Sanchez-Fueyo A, Duarte I, Torres J, Ocquetau M, Barrera F, Arrese M, Soza A, Benítez C. Conversion to mycophenolate mofetil monotherapy in liver recipients: Calcineurin inhibitor levels are key. Ann Hepatol 2017; 16:94-106. [PMID: 28051798 DOI: 10.5604/16652681.1226820] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The use of calcineurin inhibitors (CNI) after liver transplantation is associated with post-transplant nephrotoxicity. Conversion to mycophenolate mofetil (MMF) monotherapy improves renal function, but is related to graft rejection in some recipients. Our aim was to identify variables associated with rejection after conversion to MMF monotherapy. Conversion was attempted in 40 liver transplant recipients. Clinical variables were determined and peripheral mononuclear blood cells were immunophenotyped during a 12-month follow-up. Conversion was classified as successful (SC) if rejection did not occur during the follow-up. MMF conversion was successful with 28 patients (70%) and was associated with higher glomerular filtration rates at the end of study. It also correlated with increased time elapsed since transplantation, low baseline CNI levels (Tacrolimus ≤ 6.5 ng/mL or Cyclosporine ≤ 635 ng/mL) and lower frequency of tacrolimus use. The only clinical variable independently related to SC in multivariate analysis was low baseline CNI levels (p = 0.02, OR: 6.93, 95%, CI: 1.3-29.7). Mean baseline fluorescent intensity of FOXP3+ T cells was significantly higher among recipients with SC. In conclusion, this study suggests that baseline CNI levels can be used to identify recipients with higher probability of SC to MMF monotherapy. Clinicaltrials.gov identification: NCT01321112.
Collapse
Affiliation(s)
- Blanca Norero
- Departamentos de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile
| | - Carolina A Serrano
- Departamentos de Gastroenterología y Nutrición Pediátrica, Escuela de Medicina, Pontificia Universidad Católica de Chile
| | - Alberto Sanchez-Fueyo
- Institute of Liver Studies, MRC Centre for Transplantation, King's College London, London SE5 9RS, United Kingdom
| | - Ignacio Duarte
- Patología y Escuela de Medicina, Pontificia Universidad Católica de Chile
| | - Javiera Torres
- Patología y Escuela de Medicina, Pontificia Universidad Católica de Chile
| | - Mauricio Ocquetau
- Hemato-Oncología, Escuela de Medicina, Pontificia Universidad Católica de Chile
| | | | - Marco Arrese
- Departamentos de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile
| | - Alejandro Soza
- Departamentos de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile
| | - Carlos Benítez
- Departamentos de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile
| |
Collapse
|
7
|
Liu Y, Kloc M, Li XC. Macrophages as Effectors of Acute and Chronic Allograft Injury. CURRENT TRANSPLANTATION REPORTS 2016; 3:303-312. [PMID: 28546901 PMCID: PMC5440082 DOI: 10.1007/s40472-016-0130-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Organ transplants give a second chance of life to patients with end-stage organ failure. However, the immuno-logical barriers prove to be very challenging to overcome and graft rejection remains a major hurdle to long-term transplant survival. For decades, adaptive immunity has been the focus of studies, primarily based on the belief that T cells are necessary and sufficient for rejection. With better-developed immunosuppressive drugs and protocols that effectively control adaptive cells, innate immune cells have emerged as key effector cells in triggering graft injury and have therefore attracted much recent attention. In this review, we discuss current understanding of macrophages and their role in transplant rejection, their dynamics, distinct phenotypes, locations, and functions. We also discuss novel therapeutic approaches under development to target macrophages in transplant recipients.
Collapse
Affiliation(s)
- Yianzhu Liu
- Immunobiology and Transplant Science Center, Houston Methodist Research Institute, Texas Medical Center, 6670 Bertner Avenue, Houston, TX 77030, USA
- Department of Neurology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Malgorzata Kloc
- Immunobiology and Transplant Science Center, Houston Methodist Research Institute, Texas Medical Center, 6670 Bertner Avenue, Houston, TX 77030, USA
| | - Xian C. Li
- Immunobiology and Transplant Science Center, Houston Methodist Research Institute, Texas Medical Center, 6670 Bertner Avenue, Houston, TX 77030, USA
| |
Collapse
|
8
|
Lin CM, Plenter RJ, Coulombe M, Gill RG. Interferon Gamma and Contact-dependent Cytotoxicity Are Each Rate Limiting for Natural Killer Cell-Mediated Antibody-dependent Chronic Rejection. Am J Transplant 2016; 16:3121-3130. [PMID: 27163757 PMCID: PMC5083186 DOI: 10.1111/ajt.13865] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Revised: 04/26/2016] [Accepted: 05/02/2016] [Indexed: 01/25/2023]
Abstract
Natural killer (NK) cells are key components of the innate immune system. In murine cardiac transplant models, donor-specific antibodies (DSA), in concert with NK cells, are sufficient to inflict chronic allograft vasculopathy independently of T and B cells. In this study, we aimed to determine the effector mechanism(s) required by NK cells to trigger chronic allograft vasculopathy during antibody-mediated rejection. Specifically, we tested the relative contribution of the proinflammatory cytokine interferon gamma (IFN-γ) versus the contact-dependent cytotoxic mediators of perforin and the CD95/CD95L (Fas/Fas ligand [FasL]) pathway for triggering these lesions. C3H/HeJ cardiac allografts were transplanted into immune-deficient C57BL/6 rag-/- γc-/- recipients, who also received monoclonal anti-major histocompatibility complex (MHC) class I DSA. The combination of DSA and wild-type NK cell transfer triggered aggressive chronic allograft vasculopathy. However, transfer of IFN-γ-deficient NK cells or host IFN-γ neutralization led to amelioration of these lesions. Use of either perforin-deficient NK cells or CD95 (Fas)-deficient donors alone did not alter development of vasculopathy, but simultaneous disruption of NK cell-derived perforin and allograft Fas expression resulted in prevention of these abnormalities. Therefore, both NK cell IFN-γ production and contact-dependent cytotoxic activity are rate-limiting effector pathways that contribute to this form of antibody-induced chronic allograft vasculopathy.
Collapse
Affiliation(s)
- C M Lin
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, CO.
| | - R J Plenter
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, CO
| | - M Coulombe
- Department of Surgery, University of Colorado, Aurora, CO
| | - R G Gill
- Department of Surgery, University of Colorado, Aurora, CO
| |
Collapse
|
9
|
Assadiasl S, Sepanjnia A, Aghili B, Nafar M, Ahmadpoor P, Pourrezagholi F, Parvin M, Shahlaee A, Nicknam MH, Amirzargar A. Natural Killer Cell Subsets and IL-2, IL-15, and IL-18 Genes Expressions in Chronic Kidney Allograft Dysfunction and Graft Function in Kidney Allograft Recipients. Int J Organ Transplant Med 2016; 7:212-217. [PMID: 28078060 PMCID: PMC5219582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND While acute rejection and early graft loss rates have decreased substantially over the past four decades, progressive chronic allograft dysfunction (CAD) still remains a common cause of late graft loss in kidney transplant recipients. OBJECTIVE This study was conducted to investigate the percentage of natural killer (NK) cell subsets and IL-2, 15 and 18 genes expression in two groups of CAD and well-function graft (WFG) recipients. METHODS 30 renal allograft recipients with biopsy-proven interstitial fibrosis/tubular atrophy (IF/TA) and impaired renal function, and 30 sex- and age-matched WFG patients were enrolled in this study. The percentage of NK cell subsets including NK CD56bright and NK CD56dim cells were determined by flowcytometry; IL-2, IL-15, and IL-18 genes expressions were assessed by real-time PCR. RESULTS Compared to WFG patients, there was a significant (p<0.05) increase in the percentage of NK CD56bright cells in CAD patients. However, the difference in percentage of NK CD56dim cells or CD56dim/CD56bright ratio between the studied groups was not significant. In addition, IL-2, 15 and 18 genes expressions were almost similar in CAD and WFG patients. CONCLUSION We found higher percentages of NK CD56bright subset in kidney transplant recipients with CAD without considerable changes in related cytokines' gene expression, suggesting a possible defect of NK cells maturation in these patients.
Collapse
Affiliation(s)
- S. Assadiasl
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - A. Sepanjnia
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - B. Aghili
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - M. Nafar
- Chronic Kidney Disease Research Center, Labbafinejad Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - P. Ahmadpoor
- Chronic Kidney Disease Research Center, Labbafinejad Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - F. Pourrezagholi
- Chronic Kidney Disease Research Center, Labbafinejad Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - M. Parvin
- Department of Pathology, Labbafinejad Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - A. Shahlaee
- Molecular Immunology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - M. H. Nicknam
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Molecular Immunology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - A. Amirzargar
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Molecular Immunology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Legris T, Picard C, Todorova D, Lyonnet L, Laporte C, Dumoulin C, Nicolino-Brunet C, Daniel L, Loundou A, Morange S, Bataille S, Vacher-Coponat H, Moal V, Berland Y, Dignat-George F, Burtey S, Paul P. Antibody-Dependent NK Cell Activation Is Associated with Late Kidney Allograft Dysfunction and the Complement-Independent Alloreactive Potential of Donor-Specific Antibodies. Front Immunol 2016; 7:288. [PMID: 27563301 PMCID: PMC4980873 DOI: 10.3389/fimmu.2016.00288] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 07/18/2016] [Indexed: 12/21/2022] Open
Abstract
Although kidney transplantation remains the best treatment for end-stage renal failure, it is limited by chronic humoral aggression of the graft vasculature by donor-specific antibodies (DSAs). The complement-independent mechanisms that lead to the antibody-mediated rejection (ABMR) of kidney allografts remain poorly understood. Increasing lines of evidence have revealed the relevance of natural killer (NK) cells as innate immune effectors of antibody-dependent cellular cytotoxicity (ADCC), but few studies have investigated their alloreactive potential in the context of solid organ transplantation. Our study aimed to investigate the potential contribution of the antibody-dependent alloreactive function of NK cells to kidney graft dysfunction. We first conducted an observational study to investigate whether the cytotoxic function of NK cells is associated with chronic allograft dysfunction. The NK-Cellular Humoral Activation Test (NK-CHAT) was designed to evaluate the recipient and antibody-dependent reactivity of NK cells against allogeneic target cells. The release of CD107a/Lamp1+ cytotoxic granules, resulting from the recognition of rituximab-coated B cells by NK cells, was analyzed in 148 kidney transplant recipients (KTRs, mean graft duration: 6.2 years). Enhanced ADCC responsiveness was associated with reduced graft function and identified as an independent risk factor predicting a decline in the estimated glomerular filtration rate over a 1-year period (hazard ratio: 2.83). In a second approach, we used the NK-CHAT to reveal the cytotoxic potential of circulating alloantibodies in vitro. The level of CD16 engagement resulting from the in vitro recognition of serum-coated allogeneic B cells or splenic cells was further identified as a specific marker of DSA-induced ADCC. The NK-CHAT scoring of sera obtained from 40 patients at the time of transplant biopsy was associated with ABMR diagnosis. Our findings indicate that despite the administration of immunosuppressive treatments, robust ADCC responsiveness can be maintained in some KTRs. Because it evaluates both the Fab recognition of alloantigens and Fc-driven NK cell activation, the NK-CHAT represents a potentially valuable tool for the non-invasive and individualized evaluation of humoral risk during transplantation.
Collapse
Affiliation(s)
- Tristan Legris
- Nephrology Dialysis Renal Transplantation Center, Assistance Publique des Hôpitaux de Marseille, Hospital de la Conception , Marseille , France
| | - Christophe Picard
- Établissement Français du Sang Alpes Méditerranée, Marseille, France; ADES UMR 7268, CNRS, EFS, Aix-Marseille Université, Marseille, France
| | - Dilyana Todorova
- UMR 1076, Vascular Research Center of Marseille, INSERM, Aix-Marseille University , Marseille , France
| | - Luc Lyonnet
- Hematology Unit, Assistance Publique des Hôpitaux de Marseille, Hopital de la Conception , Marseille , France
| | - Cathy Laporte
- Hematology Unit, Assistance Publique des Hôpitaux de Marseille, Hopital de la Conception , Marseille , France
| | - Chloé Dumoulin
- Hematology Unit, Assistance Publique des Hôpitaux de Marseille, Hopital de la Conception , Marseille , France
| | - Corinne Nicolino-Brunet
- Hematology Unit, Assistance Publique des Hôpitaux de Marseille, Hopital de la Conception , Marseille , France
| | - Laurent Daniel
- Laboratory for Anatomy, Pathology, Neuropathology, Hôpital de la Timone, Aix-Marseille University , Marseille , France
| | - Anderson Loundou
- Unité d'Aide méthodologique à la Recherche Clinique et Epidémiologique, DRRC, Assistance Publique Hôpitaux de Marseille , Marseille , France
| | - Sophie Morange
- Centre d'Investigation Clinique, Hôpital de la Conception , Marseille , France
| | - Stanislas Bataille
- Nephrology Dialysis Renal Transplantation Center, Assistance Publique des Hôpitaux de Marseille, Hospital de la Conception , Marseille , France
| | - Henri Vacher-Coponat
- Nephrology Dialysis Renal Transplantation Center, Assistance Publique des Hôpitaux de Marseille, Hospital de la Conception , Marseille , France
| | - Valérie Moal
- Nephrology Dialysis Renal Transplantation Center, Assistance Publique des Hôpitaux de Marseille, Hospital de la Conception , Marseille , France
| | - Yvon Berland
- Nephrology Dialysis Renal Transplantation Center, Assistance Publique des Hôpitaux de Marseille, Hospital de la Conception , Marseille , France
| | - Francoise Dignat-George
- UMR 1076, Vascular Research Center of Marseille, INSERM, Aix-Marseille University, Marseille, France; Hematology Unit, Assistance Publique des Hôpitaux de Marseille, Hopital de la Conception, Marseille, France
| | - Stéphane Burtey
- Nephrology Dialysis Renal Transplantation Center, Assistance Publique des Hôpitaux de Marseille, Hospital de la Conception, Marseille, France; UMR 1076, Vascular Research Center of Marseille, INSERM, Aix-Marseille University, Marseille, France
| | - Pascale Paul
- UMR 1076, Vascular Research Center of Marseille, INSERM, Aix-Marseille University, Marseille, France; Hematology Unit, Assistance Publique des Hôpitaux de Marseille, Hopital de la Conception, Marseille, France
| |
Collapse
|
11
|
Harper IG, Ali JM, Harper SJF, Wlodek E, Alsughayyir J, Negus MC, Qureshi MS, Motalleb-Zadeh R, Saeb-Parsy K, Bolton EM, Bradley JA, Clatworthy MR, Conlon TM, Pettigrew GJ. Augmentation of Recipient Adaptive Alloimmunity by Donor Passenger Lymphocytes within the Transplant. Cell Rep 2016; 15:1214-27. [PMID: 27134179 PMCID: PMC4870521 DOI: 10.1016/j.celrep.2016.04.009] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Revised: 01/29/2016] [Accepted: 03/28/2016] [Indexed: 01/18/2023] Open
Abstract
Chronic rejection of solid organ allografts remains the major cause of transplant failure. Donor-derived tissue-resident lymphocytes are transferred to the recipient during transplantation, but their impact on alloimmunity is unknown. Using mouse cardiac transplant models, we show that graft-versus-host recognition by passenger donor CD4 T cells markedly augments recipient cellular and humoral alloimmunity, resulting in more severe allograft vasculopathy and early graft failure. This augmentation is enhanced when donors were pre-sensitized to the recipient, is dependent upon avoidance of host NK cell recognition, and is partly due to provision of cognate help for allo-specific B cells from donor CD4 T cells recognizing B cell MHC class II in a peptide-degenerate manner. Passenger donor lymphocytes may therefore influence recipient alloimmune responses and represent a therapeutic target in solid organ transplantation. Donor CD4 T cells provide cognate, but peptide-degenerate, help to all host B cells Antibody specificity is determined by concurrent B cell receptor ligation Passenger donor CD4 T cells can therefore augment host alloantibody responses Host NK cell allorecognition is critical for preventing this augmentation
Collapse
Affiliation(s)
- Ines G Harper
- School of Clinical Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Jason M Ali
- School of Clinical Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Simon J F Harper
- School of Clinical Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Elizabeth Wlodek
- School of Clinical Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | | | - Margaret C Negus
- School of Clinical Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - M Saeed Qureshi
- School of Clinical Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | | | - Kourosh Saeb-Parsy
- School of Clinical Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Eleanor M Bolton
- School of Clinical Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - J Andrew Bradley
- School of Clinical Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Menna R Clatworthy
- School of Clinical Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Thomas M Conlon
- School of Clinical Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Gavin J Pettigrew
- School of Clinical Medicine, University of Cambridge, Cambridge CB2 0QQ, UK.
| |
Collapse
|
12
|
Overcoming immunological barriers in regenerative medicine. Nat Biotechnol 2015; 32:786-94. [PMID: 25093888 DOI: 10.1038/nbt.2960] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 06/14/2014] [Indexed: 02/06/2023]
Abstract
Regenerative therapies that use allogeneic cells are likely to encounter immunological barriers similar to those that occur with transplantation of solid organs and allogeneic hematopoietic stem cells (HSCs). Decades of experience in clinical transplantation hold valuable lessons for regenerative medicine, offering approaches for developing tolerance-induction treatments relevant to cell therapies. Outside the field of solid-organ and allogeneic HSC transplantation, new strategies are emerging for controlling the immune response, such as methods based on biomaterials or mimicry of antigen-specific peripheral tolerance. Novel biomaterials can alter the behavior of cells in tissue-engineered constructs and can blunt host immune responses to cells and biomaterial scaffolds. Approaches to suppress autoreactive immune cells may also be useful in regenerative medicine. The most innovative solutions will be developed through closer collaboration among stem cell biologists, transplantation immunologists and materials scientists.
Collapse
|
13
|
Ramirez AE, Cheng HY, Lao WW, Wang YL, Wen CJ, Wallace CG, Lin CF, Shih LY, Chuang SH, Wei FC. A novel rat full-thickness hemi-abdominal wall/hindlimb osteomyocutaneous combined flap: influence of allograft mass and vascularized bone marrow content on vascularized composite allograft survival. Transpl Int 2014; 27:977-86. [DOI: 10.1111/tri.12364] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Revised: 11/10/2013] [Accepted: 05/19/2014] [Indexed: 11/26/2022]
Affiliation(s)
- Alejandro E. Ramirez
- Department of Plastic and Reconstructive Surgery; Chang Gung Memorial Hospital at Linkou; Taoyuan Taiwan
- Pontifical Catholic University of Chile; Santiago Chile
| | - Hui-Yun Cheng
- Center for Vascularized Composite Allotransplantation; Chang Gung Memorial Hospital at Linkou; Taoyuan Taiwan
| | - William W. Lao
- Department of Plastic and Reconstructive Surgery; Chang Gung Memorial Hospital at Linkou; Taoyuan Taiwan
- Medical College of Wisconsin; Milwaukee WI USA
| | - Yen-Ling Wang
- Center for Vascularized Composite Allotransplantation; Chang Gung Memorial Hospital at Linkou; Taoyuan Taiwan
| | - Chih-Jen Wen
- School of Medicine; Chang Gung University; Taoyuan Taiwan
| | - Christopher G. Wallace
- Department of Plastic and Reconstructive Surgery; Chang Gung Memorial Hospital at Linkou; Taoyuan Taiwan
| | - Chih-Fan Lin
- Center for Vascularized Composite Allotransplantation; Chang Gung Memorial Hospital at Linkou; Taoyuan Taiwan
| | - Ling-Yi Shih
- Department of Plastic and Reconstructive Surgery; Chang Gung Memorial Hospital at Linkou; Taoyuan Taiwan
| | - Sheng-Hao Chuang
- Department of Plastic and Reconstructive Surgery; Chang Gung Memorial Hospital at Linkou; Taoyuan Taiwan
| | - Fu-Chan Wei
- Department of Plastic and Reconstructive Surgery; Chang Gung Memorial Hospital at Linkou; Taoyuan Taiwan
- Center for Vascularized Composite Allotransplantation; Chang Gung Memorial Hospital at Linkou; Taoyuan Taiwan
- School of Medicine; Chang Gung University; Taoyuan Taiwan
| |
Collapse
|
14
|
Caterson EJ, Lopez J, Medina M, Pomahac B, Tullius SG. Ischemia-reperfusion injury in vascularized composite allotransplantation. J Craniofac Surg 2014; 24:51-6. [PMID: 23321872 DOI: 10.1097/scs.0b013e31827104e1] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Vascularized composite allotransplantation may now be considered a viable treatment option in patients with complex craniofacial and limb defects. However, the field is still in its infancy, and challenges continue to exist. These challenges, most notably the adverse effects of lifelong immunosuppression, must be weighed against the benefits of the procedure. Improvements in this risk-benefit ratio can be achieved by achieving tolerance and preventing rejection. Five decades after Dr. Joseph E. Murray introduced the field of transplantation to the world, we now have a better understanding of the immunologic factors that may contribute to rejection and inhibit tolerance. In this article, we review emerging evidence that suggests that "danger signals" associated with ischemia-reperfusion injury contribute to innate immune activation, promoting rejection, and inhibiting tolerance. Based on this understanding, we also describe several strategies that may ameliorate the damaging effects of ischemia-reperfusion and the clinical implications of ischemia-reperfusion on the vascularized composite tissue allotransplantation outcome.
Collapse
Affiliation(s)
- Edward J Caterson
- Divisions of Plastic Surgery and Transplant Surgery, Brigham and Women's Hospital, Boston, MA 02115, USA.
| | | | | | | | | |
Collapse
|
15
|
Shilling RA. Harnessing natural killer cells to protect lung transplants from acute rejection. Am J Respir Crit Care Med 2013; 187:1284-6. [PMID: 23767901 DOI: 10.1164/rccm.201304-0634ed] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
16
|
Wood K, Shankar S, Mittal S. Concepts and challenges in organ transplantation. Clin Immunol 2013. [DOI: 10.1016/b978-0-7234-3691-1.00095-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
17
|
Khalifian S, Broyles JM, Tuffaha SH, Alrakan M, Ibrahim Z, Sarhane KA. Immune mechanisms of ischemia-reperfusion injury in transplantation. ACTA ACUST UNITED AC 2013. [DOI: 10.4236/oji.2013.33020] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
18
|
Hirohashi T, Chase CM, Pelle PD, Sebastian D, Alessandrini A, Madsen JC, Russell PS, Colvin RB. A novel pathway of chronic allograft rejection mediated by NK cells and alloantibody. Am J Transplant 2012; 12:313-21. [PMID: 22070565 PMCID: PMC3667648 DOI: 10.1111/j.1600-6143.2011.03836.x] [Citation(s) in RCA: 167] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Chronic allograft vasculopathy (CAV) in murine heart allografts can be elicited by adoptive transfer of donor specific antibody (DSA) to class I MHC antigens and is independent of complement. Here we address the mechanism by which DSA causes CAV. B6.RAG1(-/-) or B6.RAG1(-/-)C3(-/-) (H-2(b)) mice received B10.BR (H-2(k)) heart allografts and repeated doses of IgG2a, IgG1 or F(ab')(2) fragments of IgG2a DSA (anti-H-2(k)). Intact DSA regularly elicited markedly stenotic CAV in recipients over 28 days. In contrast, depletion of NK cells with anti-NK1.1 reduced significantly DSA-induced CAV, as judged morphometrically. Recipients genetically deficient in mature NK cells (γ-chain knock out) also showed decreased severity of DSA-induced CAV. Direct NK reactivity to the graft was not necessary. F(ab')(2) DSA fragments, even at doses twofold higher than intact DSA, were inactive. Graft microvascular endothelial cells responded to DSA in vivo by increased expression of phospho-extracellular signal-regulated kinase (pERK), a response not elicited by F(ab')(2) DSA. We conclude that antibody mediates CAV through NK cells, by an Fc dependent manner. This new pathway adds to the possible mechanisms of chronic rejection and may relate to the recently described C4d-negative chronic antibody-mediated rejection in humans.
Collapse
Affiliation(s)
- T. Hirohashi
- Transplantation and Cardiac Surgical Divisions, Department of Surgery of the Harvard Medical School, Massachusetts General Hospital, Boston, MA
| | - C. M. Chase
- Transplantation and Cardiac Surgical Divisions, Department of Surgery of the Harvard Medical School, Massachusetts General Hospital, Boston, MA
| | - P. Della Pelle
- Department of Pathology, Massachusetts General Hospital, Boston, MA
| | - D. Sebastian
- Department of Pathology, Massachusetts General Hospital, Boston, MA
| | - A. Alessandrini
- Transplantation and Cardiac Surgical Divisions, Department of Surgery of the Harvard Medical School, Massachusetts General Hospital, Boston, MA
| | - J. C. Madsen
- Transplantation and Cardiac Surgical Divisions, Department of Surgery of the Harvard Medical School, Massachusetts General Hospital, Boston, MA
| | - P. S. Russell
- Transplantation and Cardiac Surgical Divisions, Department of Surgery of the Harvard Medical School, Massachusetts General Hospital, Boston, MA
| | - R. B. Colvin
- Department of Pathology, Massachusetts General Hospital, Boston, MA,Corresponding author: Robert B. Colvin,
| |
Collapse
|
19
|
Karp SJ, Mannon RB. What's New, What's Hot in Solid Organ Transplantation? Summary of the American Transplant Congress 2011. Am J Transplant 2011; 11:2308-16. [PMID: 21967066 DOI: 10.1111/j.1600-6143.2011.03774.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Breakthroughs in basic and clinical science in solid organ transplantation were presented at the American Transplant Congress 2011. Key areas of presentation included the pathogenesis of late allograft failure, immune regulation and tolerance, pathways in allograft injury, electing appropriate patients for transplantation, determining the best allocation schemes to maximize effective utilization, organ preservation, monitoring the alloimmune response and immunosuppressive management. In this review, we present highlights of the meeting. These presentations demonstrate the exciting promise in translating from the bench to affect patient care.
Collapse
Affiliation(s)
- S J Karp
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN, USA
| | | |
Collapse
|
20
|
Moroso V, van der Meer A, Tilanus HW, Kazemier G, van der Laan LJW, Metselaar HJ, Joosten I, Kwekkeboom J. Donor and recipient HLA/KIR genotypes do not predict liver transplantation outcome. Transpl Int 2011; 24:932-42. [PMID: 21672051 DOI: 10.1111/j.1432-2277.2011.01286.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Whether or not Natural Killer (NK) cells affect the immune response to solid organ allografts is still controversial. Main determinants of NK-cell activation are specific HLA/killer-cell immunoglobulin-like receptors (KIR) interactions that, in transplantation, may induce NK-cell alloreactivity. So far, in liver transplantation (LTX) donor-versus-recipient alloreactivity has not been investigated; in addition, studies of predicted recipient-versus-donor NK-cell alloreactivity have led to contradicting results. We typed a cohort of LTX donors and recipients for HLA-C/Bw4 and KIRs. We estimated the effect of NK-cell alloreactivity, as predicted by classically used models, in the donor-versus-recipient direction. The results indicate that HLA/KIR mismatches in the donor-versus-recipient direction do not predict graft rejection nor graft or patient survival, suggesting that donor-derived NK cells do not play a major role in LTX outcome. In addition, when considering predicted NK-cell alloreactivity in the reverse direction (recipient-versus-donor), we first confirmed that donor HLA-C genotype was not associated with acute rejection, graft or patient survival and secondly we found that none of the models describing NK-cell alloreactivity could predict LTX outcome. Overall our observations suggest that, in contrast to what is shown in haematopoietic stem cell transplantation, donor-derived NK cells may not contribute in preventing liver graft rejection, and that recipient-versus-donor NK-cell alloreactivity does not predict LTX outcome.
Collapse
Affiliation(s)
- Viviana Moroso
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
21
|
|