1
|
Laumont CM, Nelson BH. B cells in the tumor microenvironment: Multi-faceted organizers, regulators, and effectors of anti-tumor immunity. Cancer Cell 2023; 41:466-489. [PMID: 36917951 DOI: 10.1016/j.ccell.2023.02.017] [Citation(s) in RCA: 76] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/11/2023] [Accepted: 02/12/2023] [Indexed: 03/14/2023]
Abstract
Our understanding of tumor-infiltrating lymphocytes (TILs) is rapidly expanding beyond T cell-centric perspectives to include B cells and plasma cells, collectively referred to as TIL-Bs. In many cancers, TIL-Bs carry strong prognostic significance and are emerging as key predictors of response to immune checkpoint inhibitors. TIL-Bs can perform multiple functions, including antigen presentation and antibody production, which allow them to focus immune responses on cognate antigen to support both T cell responses and innate mechanisms involving complement, macrophages, and natural killer cells. In the stroma of the most immunologically "hot" tumors, TIL-Bs are prominent components of tertiary lymphoid structures, which resemble lymph nodes structurally and functionally. Additionally, TIL-Bs participate in a variety of other lympho-myeloid aggregates and engage in dynamic interactions with the tumor stroma. Here, we summarize our current understanding of TIL-Bs in human cancer, highlighting the compelling therapeutic opportunities offered by their unique tumor recognition and effector mechanisms.
Collapse
Affiliation(s)
- Céline M Laumont
- Deeley Research Centre, BC Cancer, Victoria, BC V8R 6V5, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Brad H Nelson
- Deeley Research Centre, BC Cancer, Victoria, BC V8R 6V5, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC V8P 3E6, Canada.
| |
Collapse
|
2
|
Mature naive B cells regulate the outcome of murine acute graft-versus-host disease in an IL-10 independent manner. Transplant Cell Ther 2022; 28:181.e1-181.e9. [PMID: 35032717 DOI: 10.1016/j.jtct.2022.01.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 01/04/2022] [Accepted: 01/06/2022] [Indexed: 12/30/2022]
Abstract
Graft-versus-host disease (GVHD) is the main complication of bone marrow transplantation (BMT). T CD4+ lymphocytes are the main effector cells for disease development but other cell types can determine disease outcome through cytokine production and antigen presentation. B cells are abundant in BMT products and are involved in chronic GVHD immunopathogenesis. However, their role in acute GVHD is still unclear. Here, we studied the role of donor resting B cells in a model of acute GVHD. Animals receiving transplants depleted of B cells presented a more severe disease, indicating a protective role for B cells. Mice transplanted with IL-10 KO B cells developed GVHD as severe as those receiving WT B cells. Besides that, mice transplanted with MHC II deficient B cells and as so, unable to present antigen to CD4+ T cells, developed as severe GVHD as animals transplanted without B cells. This result suggests that protection provided by mature naive B cells depends on antigen presentation and not IL-10 production by B cells. In the absence of donor B cells, transplanted mice exhibited disorganized lymphoid splenic tissue. Additionally, donor B cell depletion diminished the follicular T (Tfh)/T effector (Teff) ratio suggesting that protection was correlated with a shift to Tfh differentiation, reducing the number of effector T cells. Importantly, the Tfh/Teff shift impacts disease outcome since observed proinflammatory cytokine levels and tissue damage in target organs were consistent with disease protection. The role of transplanted B cells in the outcome of BMT and the development of acute GVHD should be carefully studied, since these cells are abundant in BMT products and are potent modulator and effector cells in allogeneic response. Extended Abstract Background: B cells are widely known for their ability to produce antibodies. In addition, B cells can act efficiently as antigen-presenting cells, implying the mutual regulation of both T and B lymphocyte subsets. T cell help for B cells has been known for more than 50 years; however, B cell help for T cells, especially regarding the modulation of follicular and regulatory phenotypes, had only lately been explored. Here, we studied the role of resting B cells in a model of systemic inflammatory disease mediated by T cells, graft-versus-host disease (GVHD), which is the main complication of allogeneic bone marrow transplantation. Objetive: The objective of this paper is to investigate the role of donor B cells in acute Graft-versus-Host Disease. STUDY DESIGN To investigate the role of donor B cells in aGVHD, we used a full MHC-mismatched bone marrow transplantation model. We infused C57BL/6 BM cells along with splenocytes depleted or not of B220+ cells into lethally irradiated BALB/c mice. We also used B cells from IL-10 KO mice to investigate the role of IL-10 produced by donor B cells and B cells from mice which cannot express MHC-II (CIITA KO) to investigate the role of cognate interaction between donor B and T cells. RESULTS Animals receiving transplants depleted of B cells presented a more severe disease, showing the existence of B cell-dependent protection. This protection was dependent on the T cell-B cell cognate interaction but not on IL-10 or Treg induction. In the absence of donor B cells, transplanted mice exhibited fewer GCs and a lower follicular T (Tfh)/T effector (Teff) ratio than mice transplanted in the presence of B cells. Protection was correlated with a shift to Tfh differentiation, reducing the number of effector cells. Importantly, the Tfh/Teff shift impacts disease outcome with less T cell-mediated disease due to more B cell-dependent Tfh generation with fewer effector T cells and lower proinflammatory cytokine levels detected in target organs. CONCLUSION We show that B-cell depleted bone marrow transplantation leads to a more severe disease, with earlier mortality related to increased organ damage. Such differences depend on cognate interactions between T cells and B cells, are IL-10 independent and are related to a shift in the differentiation of lymphocytes from the follicular helper phenotype to the effector phenotype. Therefore, Teffs, which are circulating cells, become relatively more numerous and can reach and damage the target tissues. These results point to caution in the early posttransplantation elimination of donor B cells. It is not a matter of eliminating only antibody-forming cells or cells that mediate Tfh generation but of B cells, which interact and modulate T cell activity, impacting a disease that is not antibody mediated.
Collapse
|
3
|
Li J, Thomson AW, Rogers NM. Myeloid and Mesenchymal Stem Cell Therapies for Solid Organ Transplant Tolerance. Transplantation 2021; 105:e303-e321. [PMID: 33756544 PMCID: PMC8455706 DOI: 10.1097/tp.0000000000003765] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Transplantation is now performed globally as a routine procedure. However, the increased demand for donor organs and consequent expansion of donor criteria has created an imperative to maximize the quality of these gains. The goal is to balance preservation of allograft function against patient quality-of-life, despite exposure to long-term immunosuppression. Elimination of immunosuppressive therapy to avoid drug toxicity, with concurrent acceptance of the allograft-so-called operational tolerance-has proven elusive. The lack of recent advances in immunomodulatory drug development, together with advances in immunotherapy in oncology, has prompted interest in cell-based therapies to control the alloimmune response. Extensive experimental work in animals has characterized regulatory immune cell populations that can induce and maintain tolerance, demonstrating that their adoptive transfer can promote donor-specific tolerance. An extension of this large body of work has resulted in protocols for manufacture, as well as early-phase safety and feasibility trials for many regulatory cell types. Despite the excitement generated by early clinical trials in autoimmune diseases and organ transplantation, there is as yet no clinically validated, approved regulatory cell therapy for transplantation. In this review, we summarize recent advances in this field, with a focus on myeloid and mesenchymal cell therapies, including current understanding of the mechanisms of action of regulatory immune cells, and clinical trials in organ transplantation using these cells as therapeutics.
Collapse
Affiliation(s)
- Jennifer Li
- Center of Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, Australia
- Faculty of Medicine and Health, Sydney Medical School, University of Sydney, Sydney, Australia
| | - Angus W Thomson
- Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Natasha M Rogers
- Center of Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, Australia
- Faculty of Medicine and Health, Sydney Medical School, University of Sydney, Sydney, Australia
| |
Collapse
|
4
|
Wang Y, Li Z, Hu Q. Emerging self-regulated micro/nano drug delivery devices: A step forward towards intelligent diagnosis and therapy. NANO TODAY 2021; 38:101127. [DOI: 10.1016/j.nantod.2021.101127] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
5
|
Farooq SM, Ashour HM. Type II Collagen-Specific B Cells Induce Immune Tolerance in Th1-Skewed, Th2-Skewed, and Arthritis-Prone Strains of Mice. Cells 2021; 10:cells10040870. [PMID: 33921248 PMCID: PMC8068989 DOI: 10.3390/cells10040870] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 04/01/2021] [Accepted: 04/05/2021] [Indexed: 01/15/2023] Open
Abstract
Antigen-specific regulatory T cells play key immune suppressive roles in autoimmune disease models and regulate the peripheral tolerance achieved via anterior chamber-associated immune deviation (ACAID). Articular cartilage has type II collagen (CII), which is a potent autoantigen protein in arthritis. There has not been much research on the clinical importance of CII-associated diseases. Moreover, the capability of CII to induce immune tolerance has not been previously assessed. We reported that delivery of CII either directly into the eye or via intravenous injection of CII-specific ACAID antigen presenting cells (APCs) can induce ACAID. Here, we hypothesized that peripheral tolerance can be induced following adoptive transfer of in vitro generated CII-specific ACAID B cells to naive mice. Delayed hypersensitivity (DTH) assays were used to assess the suppressive ability of adoptively transferred B cells. Immune responses of ACAID B cell-injected mice were significantly suppressed following challenges with CII as compared to positive controls. This effect was replicated in three different strains of mice (C57BL/6, BALB/c, and DBA/1). Thus, CII-specific ACAID B cells were able to induce immune tolerance in Th1-skewed, Th2-skewed, and arthritis-prone mice. ACAID B cell-mediated tolerance induced by CII could have therapeutic implications for the treatment of CII-mediated autoimmune diseases.
Collapse
Affiliation(s)
- Shukkur M. Farooq
- Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA;
| | - Hossam M. Ashour
- Department of Integrative Biology, College of Arts and Sciences, University of South Florida, St. Petersburg, FL 33701, USA
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
- Correspondence:
| |
Collapse
|
6
|
Irure-Ventura J, San Segundo D, Rodrigo E, Merino D, Belmar-Vega L, Ruiz San Millán JC, Valero R, Benito A, López-Hoyos M. High Pretransplant BAFF Levels and B-cell Subset Polarized towards a Memory Phenotype as Predictive Biomarkers for Antibody-Mediated Rejection. Int J Mol Sci 2020; 21:ijms21030779. [PMID: 31991734 PMCID: PMC7037386 DOI: 10.3390/ijms21030779] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 01/20/2020] [Accepted: 01/22/2020] [Indexed: 01/05/2023] Open
Abstract
Antibody-mediated rejection (AbMR) is one of the leading causes of graft loss in kidney transplantation and B cells play an important role in the development of it. A B-cell activating factor (BAFF) is a cytokine involved in B cell ontogeny. Here, we analyzed whether B cell maturation and the effect of B cell soluble factors, such as BAFF could be involved in AbMR. Serum BAFF levels and B and T cell subpopulations were analyzed 109 kidney transplant patients before transplantation and at 6 and 12 months after kidney transplantation. Pretransplant serum BAFF levels as well as memory B cell subpopulations were significantly higher in those patients who suffered clinical AbMR during the first 12 months after kidney transplantation. Similar results were observed in the prospective analysis of patients with subclinical antibody-mediated rejection detected in the surveillance biopsy performed at 12 months after kidney transplantation. A multivariate analysis confirmed the independent role of BAFF in the development of AbMR, irrespective of other classical variables. Pretransplant serum BAFF levels could be an important non-invasive biomarker for the prediction of the development of AbMR and posttransplant increased serum BAFF levels contribute to AbMR.
Collapse
Affiliation(s)
- Juan Irure-Ventura
- Immunology Department. University Hospital Marqués de Valdecilla-IDIVAL, 39008 Santander, Spain; (J.I.-V.); (D.S.S.)
| | - David San Segundo
- Immunology Department. University Hospital Marqués de Valdecilla-IDIVAL, 39008 Santander, Spain; (J.I.-V.); (D.S.S.)
| | - Emilio Rodrigo
- Nephrology Department. University Hospital Marqués de Valdecilla-IDIVAL, 39008 Santander, Spain, (L.B.-V.); (J.C.R.S.M.); (R.V.)
| | - David Merino
- Health Research Institute-IDIVAL, 39011 Santander, Spain; (D.M.); (A.B.)
| | - Lara Belmar-Vega
- Nephrology Department. University Hospital Marqués de Valdecilla-IDIVAL, 39008 Santander, Spain, (L.B.-V.); (J.C.R.S.M.); (R.V.)
| | - Juan Carlos Ruiz San Millán
- Nephrology Department. University Hospital Marqués de Valdecilla-IDIVAL, 39008 Santander, Spain, (L.B.-V.); (J.C.R.S.M.); (R.V.)
| | - Rosalía Valero
- Nephrology Department. University Hospital Marqués de Valdecilla-IDIVAL, 39008 Santander, Spain, (L.B.-V.); (J.C.R.S.M.); (R.V.)
| | - Adalberto Benito
- Health Research Institute-IDIVAL, 39011 Santander, Spain; (D.M.); (A.B.)
| | - Marcos López-Hoyos
- Immunology Department. University Hospital Marqués de Valdecilla-IDIVAL, 39008 Santander, Spain; (J.I.-V.); (D.S.S.)
- Correspondence: (M.L.-H.); Tel.: +34-942-202520 (ext. 73225)
| |
Collapse
|
7
|
Sönmez RE, Ilhan M, Ertekin C. Post-Transplant Immune Tolerance in Rats Following Lymphocyte Injection Into the Anterior Chamber of the Eye. Transplant Proc 2019; 51:2132-2135. [PMID: 31399189 DOI: 10.1016/j.transproceed.2019.04.064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 01/24/2019] [Accepted: 04/22/2019] [Indexed: 10/26/2022]
Abstract
OBJECTIVE We aimed to compare the clinical and histopathological results of skin graft transplants between rats that had been injected with lymphocytes into the anterior chamber of the eye with those that had not. METHODS A total of 16 Wistar albino, male rats were included in the study. Subjects were divided into 2 groups, namely a test group and a control group. Lymphocyte suspensions derived from the subjects of the control group were injected into the anterior chamber of the eye of each opposing subject of the test group. Also, an identical volume of physiological saline was injected into the anterior chamber of each subject in the control group to prevent bias. One week after this procedure, circular skin grafts of 1 cm in diameter were transplanted within the opposing groups. After a period of 1 week, transplanted graft tissues were excised to compare tissue healing. RESULTS The occurence of granulation and reepithelialization was more evident in the test group (96% and 33%, respectively, vs 80% and 17% for the control group, respectively). On the other hand, it was determined that acute inflammation was more intense in the control group (77% vs 50% for the test group). CONCLUSION We had created immune tolerance in rats through anterior chamber lymphocyte injection, which slowed down the rejection process. If this can be successfully implemented in practice, survival for transplant patients without long-term rejection will move closer to becoming a reality.
Collapse
Affiliation(s)
- Recep Erçin Sönmez
- Department of General Surgery, Istanbul Medeniyet University, Istanbul, Turkey.
| | - Mehmet Ilhan
- Department of General Surgery, Istanbul University, Istanbul Medical Faculty, Istanbul, Turkey
| | - Cemalettin Ertekin
- Department of General Surgery, Istanbul University, Istanbul Medical Faculty, Istanbul, Turkey
| |
Collapse
|
8
|
Wang XZ, Wan Z, Xue WJ, Zheng J, Li Y, Ding CG. B-Cell Activating Factor Predicts Acute Rejection Risk in Kidney Transplant Recipients: A 6-Month Follow-Up Study. Front Immunol 2019; 10:1046. [PMID: 31156628 PMCID: PMC6529956 DOI: 10.3389/fimmu.2019.01046] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Accepted: 04/24/2019] [Indexed: 01/03/2023] Open
Abstract
B cell activating factor (BAFF) belonging to TNF family is a cytokine that enhances B-cell proliferation and differentiation. Recently, It has been suggested that BAFF might be a potential therapeutic target for treating autoimmune disease. However, the relationship between BAFF and allograft rejection is controversial, and the clinical significance of BAFF in predicting allograft rejection need to be further explored. We conducted 6-month follow-up study to confirm the hypothesis that BAFF might be a risk factor for predicting acute rejection in kidney transplant recipients. At the end of the study, a total of 155 kidney transplant recipients were recruited from October 2015 to October 2017, and classified into acute rejection group (n = 34) and stable renal function group (n = 121) according to their clinical course. We demonstrate that the serum BAFF levels when acute rejection occurred was significantly higher than that in the stable renal function group (2426.19 ± 892.19 vs. 988.17 ± 485.63 pg/mL, P < 0.05). BAFF expression was significantly enhanced in the membrane and cytoplasm of renal tubule epithelial cells in the transplant kidney tissue with acute rejection, and a positive correlation between BAFF and C4d expression was also observed (r = 0.880, P = 0.001). ROC analyses highlight the superiority of serum BAFF level before transplant over those on other post-transplant days in prediction of acute rejection episodes. The sensitivity, specificity and AUC (area under curve) were 83.3, 89.5, and 0.886%, respectively. Kaplan-Meier survival analysis showed that recipients with higher pretransplant BAFF levels had higher acute rejection incidence (P = 0.003). In conclusion, we have identified that BAFF levels are associated with the acute rejection and could be a promising biomarker to predict kidney transplant rejection risks.
Collapse
Affiliation(s)
- Xu-Zhen Wang
- Critical Care Medicine, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhen Wan
- Department of General Surgery, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Wu-Jun Xue
- Department of Kidney Transplantation, Hospital of Nephropathy, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jin Zheng
- Department of Kidney Transplantation, Hospital of Nephropathy, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yang Li
- Department of Kidney Transplantation, Hospital of Nephropathy, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Chen Guang Ding
- Department of Kidney Transplantation, Hospital of Nephropathy, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
9
|
Bath NM, Ding X, Wilson NA, Verhoven BM, Boldt BA, Sukhwal A, Reese SR, Panzer SE, Djamali A, Redfield RR. Desensitization and treatment with APRIL/BLyS blockade in rodent kidney transplant model. PLoS One 2019; 14:e0211865. [PMID: 30735519 PMCID: PMC6368307 DOI: 10.1371/journal.pone.0211865] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 01/23/2019] [Indexed: 11/19/2022] Open
Abstract
Alloantibody represents a significant barrier in kidney transplant through the sensitization of patients prior to transplant through antibody mediated rejection (ABMR). APRIL BLyS are critical survival factors for mature B lymphocytes plasma cells, the primary source of alloantibody. We examined the effect of APRIL/BLyS blockade via TACI-Ig (Transmembrane activator calcium modulator cyclophilin lig interactor-Immunoglobulin) in a preclinical rodent model as treatment for both desensitization ABMR. Lewis rats were sensitized with Brown Norway (BN) blood for 21 days. Following sensitization, animals were then sacrificed or romized into kidney transplant (G4, sensitized transplant control); desensitization with TACI-Ig followed by kidney transplant (G5, sensitized + pre-transplant TACI-Ig); kidney transplant with post-transplant TACI-Ig for 21 days (G6, sensitized + post-transplant TACI-Ig); desensitization with TACI-Ig followed by kidney transplant post-transplant TACI-Ig for 21 days (G7, sensitized + pre- post-transplant TACI-Ig). Animals were sacrificed on day 21 post-transplant tissues were analyzed using flow cytometry, IHC, ELISPOT, RT-PCR. Sensitized animals treated with APRIL/BLyS blockade demonstrated a significant decrease in marginal zone non-switched B lymphocyte populations (p<0.01). Antibody secreting cells were also significantly reduced in the sensitized APRIL/BLyS blockade treated group. Post-transplant APRIL/BLyS blockade treated animals were found to have significantly less C4d deposition less ABMR as defined by Banff classification when compared to groups receiving APRIL/BLyS blockade before transplant or both before after transplant (p<0.0001). The finding of worse ABMR in groups receiving APRIL/BLyS blockade before both before after transplant may indicate that B lymphocyte depletion in this setting also resulted in regulatory lymphocyte depletion resulting in a worse rejection. Data presented here demonstrates that the targeting of APRIL BLyS can significantly deplete mature B lymphocytes, antibody secreting cells, effectively decrease ABMR when given post-transplant in a sensitized animal model.
Collapse
Affiliation(s)
- Natalie M. Bath
- Department of Surgery, Division of Transplant, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Xiang Ding
- Department of Surgery, Division of Transplant, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Nancy A. Wilson
- Department of Medicine, Division of Nephrology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Bret M. Verhoven
- Department of Surgery, Division of Transplant, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Brittney A. Boldt
- Department of Medicine, Division of Nephrology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Adarsh Sukhwal
- Department of Medicine, Division of Nephrology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Shannon R. Reese
- Department of Medicine, Division of Nephrology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Sarah E. Panzer
- Department of Medicine, Division of Nephrology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Arjang Djamali
- Department of Medicine, Division of Nephrology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Robert R. Redfield
- Department of Surgery, Division of Transplant, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| |
Collapse
|
10
|
Manzia TM, Gazia C, Baiocchi L, Lenci I, Milana M, Santopaolo F, Angelico R, Tisone G. Clinical Operational Tolerance and Immunosuppression Minimization in Kidney Transplantation: Where Do We Stand? Rev Recent Clin Trials 2019; 14:189-202. [PMID: 30868959 DOI: 10.2174/1574887114666190313170205] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 02/27/2019] [Accepted: 03/05/2019] [Indexed: 06/09/2023]
Abstract
BACKGROUND The 20th century represents a breakthrough in the transplantation era, since the first kidney transplantation between identical twins was performed. This was the first case of tolerance, since the recipient did not need immunosuppression. However, as transplantation became possible, an immunosuppression-free status became the ultimate goal, since the first tolerance case was a clear exception from the hard reality nowadays represented by rejection. METHODS A plethora of studies was described over the past decades to understand the molecular mechanisms responsible for rejection. This review focuses on the most relevant studies found in the literature where renal tolerance cases are claimed. Contrasting, and at the same time, encouraging outcomes are herein discussed and a glimpse on the main renal biomarkers analyzed in this field is provided. RESULTS The activation of the immune system has been shown to play a central role in organ failure, but also it seems to induce a tolerance status when an allograft is performed, despite tolerance is still rare to register. Although there are still overwhelming challenges to overcome and various immune pathways remain arcane; the immunosuppression minimization might be more attainable than previously believed. CONCLUSION . Multiple biomarkers and tolerance mechanisms suspected to be involved in renal transplantation have been investigated to understand their real role, with still no clear answers on the topic. Thus, the actual knowledge provided necessarily leads to more in-depth investigations, although many questions in the past have been answered, there are still many issues on renal tolerance that need to be addressed.
Collapse
Affiliation(s)
- Tommaso Maria Manzia
- Transplant and Hepatobiliary Unit, Department of Surgery, University of Rome Tor Vergata, Rome, Italy
| | - Carlo Gazia
- Transplant and Hepatobiliary Unit, Department of Surgery, University of Rome Tor Vergata, Rome, Italy
- Department of Surgery, Abdominal Organ Transplant Program, Wake Forest Baptist Medical Center, Winston Salem, NC, United States
- Wake Forest Institute for Regenerative Medicine, Department of Surgery, Winston-Salem, NC, United States
| | - Leonardo Baiocchi
- Hepatology and Liver Transplant Unit, University of Tor Vergata, Rome, Italy
| | - Ilaria Lenci
- Hepatology and Liver Transplant Unit, University of Tor Vergata, Rome, Italy
| | - Martina Milana
- Hepatology and Liver Transplant Unit, University of Tor Vergata, Rome, Italy
| | | | - Roberta Angelico
- Division of Abdominal Transplantation and Hepatobiliopancreatic Surgery, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - Giuseppe Tisone
- Transplant and Hepatobiliary Unit, Department of Surgery, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
11
|
Contreras-Kallens P, Terraza C, Oyarce K, Gajardo T, Campos-Mora M, Barroilhet MT, Álvarez C, Fuentes R, Figueroa F, Khoury M, Pino-Lagos K. Mesenchymal stem cells and their immunosuppressive role in transplantation tolerance. Ann N Y Acad Sci 2017; 1417:35-56. [PMID: 28700815 DOI: 10.1111/nyas.13364] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 03/13/2017] [Accepted: 03/29/2017] [Indexed: 12/23/2022]
Abstract
Since they were first described, mesenchymal stem cells (MSCs) have been shown to have important effector mechanisms and the potential for use in cell therapy. A great deal of research has been focused on unveiling how MSCs contribute to anti-inflammatory responses, including describing several cell populations involved and identifying soluble and other effector molecules. In this review, we discuss some of the contemporary evidence for use of MSCs in the field of immune tolerance, with a special emphasis on transplantation. Although considerable effort has been devoted to understanding the biological function of MSCs, additional resources are required to clarify the mechanisms of their induction of immune tolerance, which will undoubtedly lead to improved clinical outcomes for MSC-based therapies.
Collapse
Affiliation(s)
- Pamina Contreras-Kallens
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Claudia Terraza
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Karina Oyarce
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Tania Gajardo
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Mauricio Campos-Mora
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - María Teresa Barroilhet
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Carla Álvarez
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Ricardo Fuentes
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Fernando Figueroa
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Maroun Khoury
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile.,Cells for Cells, Santiago, Chile.,Consorcio Regenero, Chilean Consortium for Regenerative Medicine, Santiago, Chile
| | - Karina Pino-Lagos
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| |
Collapse
|
12
|
Viklicky O, Hruba P, Tomiuk S, Schmitz S, Gerstmayer B, Sawitzki B, Miqueu P, Mrazova P, Tycova I, Svobodova E, Honsova E, Janssen U, Volk HD, Reinke P. Sequential Targeting of CD52 and TNF Allows Early Minimization Therapy in Kidney Transplantation: From a Biomarker to Targeting in a Proof-Of-Concept Trial. PLoS One 2017; 12:e0169624. [PMID: 28085915 PMCID: PMC5234822 DOI: 10.1371/journal.pone.0169624] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Accepted: 12/11/2016] [Indexed: 01/04/2023] Open
Abstract
Background There is high medical need for safe long-term immunosuppression monotherapy in kidney transplantation. Selective targeting of post-transplant alloantigen-(re)activated effector-T cells by anti-TNF antibodies after global T cell depletion may allow safe drug minimization, however, it is unsolved what might be the best maintenance monotherapy. Methods In this open, prospective observational single-centre trial, 20 primary deceased donor kidney transplant recipients received 2x20 mg Alemtuzumab (d0/d1) followed by 5 mg/kg Infliximab (d2). For 14 days all patients received only tacrolimus, then they were allocated to either receive tacrolimus (TAC, n = 13) or sirolimus (SIR, n = 7) monotherapy, respectively. Protocol biopsies and extensive immune monitoring were performed and patients were followed-up for 60 months. Results TAC-monotherapy resulted in excellent graft survival (5yr 92%, 95%CI: 56.6–98.9) and function, normal histology, and no proteinuria. Immune monitoring revealed low intragraft inflammation (urinary IP-10) and hints for the development of operational tolerance signature in the TAC- but not SIR-group. Remarkably, the TAC-monotherapy was successful in all five presensitized (ELISPOT+) patients. However, recruitment into SIR-arm was stopped (after n = 7) because of high incidence of proteinuria and acute/chronic rejection in biopsies. No opportunistic infections occurred during follow-up. Conclusions In conclusion, our novel fast-track TAC-monotherapy protocol is likely to be safe and preliminary results indicated an excellent 5-year outcome, however, a full–scale study will be needed to confirm our findings. Trial Registration EudraCT Number: 2006-003110-18
Collapse
Affiliation(s)
- Ondrej Viklicky
- Department of Nephrology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
- Transplant Laboratory, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
- * E-mail:
| | - Petra Hruba
- Transplant Laboratory, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | | | | | | | - Birgit Sawitzki
- Institute of Medical Immunology, Charité University Medicine Berlin, Germany
- Berlin-Brandenburg Center for Regenerative Medicine (BCRT), Charité University Medicine Berlin, Germany
| | - Patrick Miqueu
- Institut National de la Santé et de la Recherche Médicale INSERM U1064, France
- Institut de Transplantation Urologie Néphrologie du Centre Hospitalier Universitaire Hôtel Dieu, Nantes, France
| | - Petra Mrazova
- Transplant Laboratory, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Irena Tycova
- Transplant Laboratory, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Eva Svobodova
- Department of Immunogenetics, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Eva Honsova
- Department of Clinical and Transplant Pathology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Uwe Janssen
- Miltenyi Biotec GmbH, Bergisch Gladbach, Germany
| | - Hans-Dieter Volk
- Institute of Medical Immunology, Charité University Medicine Berlin, Germany
- Berlin-Brandenburg Center for Regenerative Medicine (BCRT), Charité University Medicine Berlin, Germany
| | - Petra Reinke
- Berlin-Brandenburg Center for Regenerative Medicine (BCRT), Charité University Medicine Berlin, Germany
- Department of Nephrology and Intensive Care Medicine, Charité University Medicine Berlin, Germany
| |
Collapse
|
13
|
Min JW, Kim KW, Kim BM, Doh KC, Choi MS, Choi BS, Park CW, Yang CW, Kim YS, Oh EJ, Chung BH. Clinical Significance of Pre- and Post-Transplant BAFF Levels in Kidney Transplant Recipients. PLoS One 2016; 11:e0162964. [PMID: 27631619 PMCID: PMC5025051 DOI: 10.1371/journal.pone.0162964] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 08/31/2016] [Indexed: 11/18/2022] Open
Abstract
It is well known that pre-transplant B cell activating factor (BAFF) levels are associated with the development of de novo anti-HLA antibodies and antibody mediated rejection post-transplant. However, the clinical significance of BAFF values at allograft rejection has not been determined. In this study, we investigated the clinical significance of pre-transplant BAFF level as well as post-transplant BAFF levels measured when indication biopsy was done. We checked for anti-HLA antibodies in 115 kidney transplant recipients who required allograft biopsy due to an increase in serum creatinine. With the same serum specimen, we measured BAFF levels, and in 78 of these patients, pre-transplant BAFF and anti-HLA antibody levels were detected as well. Patients in each group were divided into tertiles according to BAFF levels. We investigated the relationship between BAFF levels and the occurrence of anti-HLA antibodies. Pre-transplant BAFF levels showed significant association with pre-transplant sensitization, and also with early rejection (Tertile 3, 26.9% vs. Tertile 1, 11.5%; P<0.05). Post-transplant BAFF levels showed significant association with pre-transplant sensitization, but did not show association with anti-HLA antibodies and positive donor-specific antibodies at the time of biopsy. We did not find any association between post-transplant BAFF levels and allograft biopsy results, Banff scores and microvascular inflammation scores. In conclusion, pre-transplant BAFF levels are associated with pre-transplant sensitization and are useful in predicting allograft rejection. But post-transplant BAFF levels measured at the time of indication biopsy are not associated with the appearance of de novo HLA-DSA, allograft rejection, biopsy findings and other allograft outcomes.
Collapse
Affiliation(s)
- Ji Won Min
- Transplant research center, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Division of Nephrology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Kyoung Woon Kim
- Transplant research center, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Bo-Mi Kim
- Transplant research center, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Kyoung Chan Doh
- Transplant research center, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Min Seok Choi
- Transplant research center, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Bum Soon Choi
- Transplant research center, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Division of Nephrology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Cheol Whee Park
- Transplant research center, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Division of Nephrology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Chul Woo Yang
- Transplant research center, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Division of Nephrology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Yong-Soo Kim
- Transplant research center, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Division of Nephrology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Eun-Jee Oh
- Department of Laboratory Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
- * E-mail: (BHC); (EJO)
| | - Byung Ha Chung
- Transplant research center, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Division of Nephrology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
- * E-mail: (BHC); (EJO)
| |
Collapse
|
14
|
Lu J, Zhang X. Immunological characteristics of renal transplant tolerance in humans. Mol Immunol 2016; 77:71-8. [PMID: 27479171 DOI: 10.1016/j.molimm.2016.07.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 07/13/2016] [Accepted: 07/13/2016] [Indexed: 10/21/2022]
Abstract
Establishing allograft tolerance is a highly desirable therapeutic goal in kidney transplantation, from which recipients would greatly benefit by withdrawing or minimizing immunosuppression. Identifying biomarkers in predicting tolerance or early diagnosing rejection is essential to direct personalized management. Recent findings have revealed that multiple populations of immune cells have involved in promoting long-term graft function or inducing rejection in renal transplant recipients. Thus, roles of immune cells add another level to predict the renal tolerant state; tailoring their functional and/or phenotypic characteristics would provide insights into mechanism involved in transplant tolerance that may aid in designing new therapies. Here, we review these findings and discuss the current understanding immunological characteristics of renal transplant tolerance in humans, and their potential clinical translation to immune tolerance biomarkers.
Collapse
Affiliation(s)
- Jingli Lu
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaojian Zhang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
15
|
Interleukin-10 From Marginal Zone Precursor B-Cell Subset Is Required for Costimulatory Blockade-Induced Transplantation Tolerance. Transplantation 2015; 99:1817-28. [PMID: 25839706 DOI: 10.1097/tp.0000000000000718] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Blocking CD40-CD40L costimulatory signals induces transplantation tolerance. Although B-cell depletion prevents alloantibody formation, nonhumoral functions of B cells in tolerance have not been well characterized. We investigated whether specific subsets of B cell or B cell-derived interleukin (IL)-10 contribute to tolerance. METHODS Wild type C57BL/6, or B cell-specific interleukin (IL)-10 (CD19-Cre::IL-10) mice, received vascularized BALB/c cardiac allografts. BALB/c donor-specific splenocyte transfusion and anti-CD40L monoclonal antibody were used as tolerogen. B cells were depleted with antimouse CD20 monoclonal antibody. Various B-cell subsets were purified and characterized by flow cytometry, reverse transcription polymerase chain reaction, and adoptive transfer. RESULTS B-cell depletion prevented costimulatory blockade-induced allogeneic tolerance. Costimulatory blockade increased IL-10 in marginal zone precursor (MZP) B cells, but not other subsets. In particular, costimulatory blockade did not change other previously defined regulatory B-cell subsets (Breg), including CD5CD1d Breg or expression of TIM1 or TIM4 on these Breg or other Breg cell subsets. Costimulatory blockade also induced IL-21R expression in MZP B cells, and IL-21R MZP B cells expressed even more IL-10. B-cell depletion or IL-10 deficiency in B cells prevented tolerance in a cardiac allograft model, resulting in rapid acute cardiac allograft rejection. Adoptive transfer of wild type MZP B cells but not other subsets to B cell-specific IL-10 deficient mice prevented graft rejection. CONCLUSIONS CD40 costimulatory blockade induces MZP B cell IL-10 which is necessary for tolerance. These observations have implications for understanding tolerance induction and how B cell depletion may prevent tolerance.
Collapse
|
16
|
Lo DJ, Kirk AD. Immune responses to transplants. Transpl Immunol 2015. [DOI: 10.1002/9781119072997.ch8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
17
|
A common gene signature across multiple studies relate biomarkers and functional regulation in tolerance to renal allograft. Kidney Int 2015; 87:984-95. [PMID: 25629549 PMCID: PMC4424816 DOI: 10.1038/ki.2014.395] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Revised: 10/02/2014] [Accepted: 10/23/2014] [Indexed: 01/21/2023]
Abstract
Patients tolerant to a kidney graft display a specific blood cell transcriptional pattern but results from five different studies were inconsistent, raising the question of relevance for future clinical application. To resolve this, we sought to identify a common gene signature, specific functional and cellular components, and discriminating biomarkers for tolerance following kidney transplantation. A meta-analysis of studies identified a robust gene signature involving proliferation of B and CD4 T cells, and inhibition of CD14 monocyte related functions among 96 tolerant samples. This signature was further supported through a cross-validation approach, yielding 92.5% accuracy independent of the study of origin. Experimental validation, performed on new tolerant samples and using a selection of the top-20 biomarkers, returned 91.7% of good classification. Beyond the confirmation of B-cell involvement, our data also indicated participation of other cell subsets in tolerance. Thus, the use of the top 20 biomarkers, mostly centered on B cells, may provide a common and standardized tool towards personalized medicine for the monitoring of tolerant or low-risk patients among kidney allotransplant recipients. These data point to a global preservation of genes favoring the maintenance of a homeostatic and ‘healthy' environment in tolerant patients and may contribute to a better understanding of tolerance maintenance mechanisms.
Collapse
|
18
|
Shimabukuro-Vornhagen A, Zoghi S, Liebig TM, Wennhold K, Chemitz J, Draube A, Kochanek M, Blaschke F, Pallasch C, Holtick U, Scheid C, Theurich S, Hallek M, von Bergwelt-Baildon MS. Inhibition of protein geranylgeranylation specifically interferes with CD40-dependent B cell activation, resulting in a reduced capacity to induce T cell immunity. THE JOURNAL OF IMMUNOLOGY 2014; 193:5294-305. [PMID: 25311809 DOI: 10.4049/jimmunol.1203436] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Ab-independent effector functions of B cells, such as Ag presentation and cytokine production, have been shown to play an important role in a variety of immune-mediated conditions such as autoimmune diseases, transplant rejection, and graft-versus-host disease. Most current immunosuppressive treatments target T cells, are relatively unspecific, and result in profound immunosuppression that places patients at an increased risk of developing severe infections and cancer. Therapeutic strategies, which interfere with B cell activation, could therefore be a useful addition to the current immunosuppressive armamentarium. Using a transcriptomic approach, we identified upregulation of genes that belong to the mevalonate pathway as a key molecular event following CD40-mediated activation of B cells. Inhibition of 3-hydroxy-3-methylglutaryl CoA reductase, the rate-limiting enzyme of the mevalonate pathway, by lipophilic statins such as simvastatin and atorvastatin resulted in a specific inhibition of B cell activation via CD40 and impaired their ability to act as stimulatory APCs for allospecific T cells. Mechanistically, the inhibitory effect resulted from the inhibition of protein geranylgeranylation subsequent to the depletion of mevalonate, the metabolic precursor for geranylgeranyl. Thus, inhibition of geranylgeranylation either directly through geranylgeranyl transferase inhibitors or indirectly through statins represents a promising therapeutic approach for the treatment of diseases in which Ag presentation by B cells plays a role.
Collapse
Affiliation(s)
- Alexander Shimabukuro-Vornhagen
- Cologne Interventional Immunology, University Hospital of Cologne, 50924 Cologne, Germany; Stem Cell Transplantation Program, University Hospital of Cologne, 50924 Cologne, Germany; Intensive Care Unit and Laboratory for Department I of Internal Medicine, University Hospital of Cologne, 50924 Cologne, Germany;
| | - Shahram Zoghi
- Cologne Interventional Immunology, University Hospital of Cologne, 50924 Cologne, Germany
| | - Tanja M Liebig
- Cologne Interventional Immunology, University Hospital of Cologne, 50924 Cologne, Germany
| | - Kerstin Wennhold
- Cologne Interventional Immunology, University Hospital of Cologne, 50924 Cologne, Germany
| | - Jens Chemitz
- Stem Cell Transplantation Program, University Hospital of Cologne, 50924 Cologne, Germany
| | - Andreas Draube
- Cologne Interventional Immunology, University Hospital of Cologne, 50924 Cologne, Germany
| | - Matthias Kochanek
- Stem Cell Transplantation Program, University Hospital of Cologne, 50924 Cologne, Germany; Intensive Care Unit and Laboratory for Department I of Internal Medicine, University Hospital of Cologne, 50924 Cologne, Germany
| | - Florian Blaschke
- Department of Cardiology, Charité Campus Virchow-Klinikum, 13353 Berlin, Germany; and Experimental and Clinical Research Center, a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | - Christian Pallasch
- Stem Cell Transplantation Program, University Hospital of Cologne, 50924 Cologne, Germany
| | - Udo Holtick
- Cologne Interventional Immunology, University Hospital of Cologne, 50924 Cologne, Germany; Stem Cell Transplantation Program, University Hospital of Cologne, 50924 Cologne, Germany
| | - Christof Scheid
- Stem Cell Transplantation Program, University Hospital of Cologne, 50924 Cologne, Germany
| | - Sebastian Theurich
- Cologne Interventional Immunology, University Hospital of Cologne, 50924 Cologne, Germany; Stem Cell Transplantation Program, University Hospital of Cologne, 50924 Cologne, Germany
| | - Michael Hallek
- Stem Cell Transplantation Program, University Hospital of Cologne, 50924 Cologne, Germany; Intensive Care Unit and Laboratory for Department I of Internal Medicine, University Hospital of Cologne, 50924 Cologne, Germany
| | - Michael S von Bergwelt-Baildon
- Cologne Interventional Immunology, University Hospital of Cologne, 50924 Cologne, Germany; Stem Cell Transplantation Program, University Hospital of Cologne, 50924 Cologne, Germany; Intensive Care Unit and Laboratory for Department I of Internal Medicine, University Hospital of Cologne, 50924 Cologne, Germany
| |
Collapse
|
19
|
Abstract
Immunosuppressive therapy in pediatrics continues to evolve. Over the past decade, newer immunosuppressive agents have been introduced into adult and pediatric transplant patients with the goal of improving patient and allograft survival. Unfortunately, large-scale randomized clinical trials are not commonly performed in children. The purpose of this review is to discuss the newer immunosuppressive agents available for induction therapy, maintenance immunosuppression, and the treatment of rejection.
Collapse
Affiliation(s)
- Christina Nguyen
- Division of Pediatric Nephrology, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, United States
| | - Ron Shapiro
- Division of Transplant Surgery, UPMC Thomas E. Starzl Transplantation Institute, Pittsburgh, PA, United States
| |
Collapse
|
20
|
Affiliation(s)
- Börje Haraldsson
- From the Institute of Medicine, University of Gothenburg, Gothenburg, Sweden; and the Department of Medicine, Icahn School of Medicine at Mount Sinai, New York
| |
Collapse
|
21
|
Gao J, McIntyre MSF, D'Souza CA, Zhang L. Pretransplant infusion of donor B cells enhances donor-specific skin allograft survival. PLoS One 2013; 8:e77761. [PMID: 24204953 PMCID: PMC3810130 DOI: 10.1371/journal.pone.0077761] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 09/04/2013] [Indexed: 01/06/2023] Open
Abstract
Pretransplant donor lymphocyte infusion (DLI) has been shown to enhance donor-specific allograft survival in rodents, primates and humans. However, the cell subset that is critical for the DLI effect and the mechanisms involved remain elusive. In this study, we monitored donor cell subsets after DLI in a murine MHC class I Ld-mismatched skin transplantation model. We found that donor B cells, but not DCs, are the major surviving donor APCs in recipients following DLI. Infusing donor B, but not non-B, cells resulted in significantly enhanced donor-specific skin allograft survival. Furthermore, mice that had received donor B cells showed higher expression of Ly6A and CD62L on antigen-specific TCRαβ+CD3+CD4−CD8−NK1.1− double negative (DN) regulatory T cells (Tregs). B cells presented alloantigen to DN Tregs and primed their proliferation in an antigen-specific fashion. Importantly, DN Tregs, activated by donor B cells, showed increased cytotoxicity toward anti-donor CD8+ T cells. These data demonstrate that donor B cells can enhance skin allograft survival, at least partially, by increasing recipient DN Treg-mediated killing of anti-donor CD8+ T cells. These findings provide novel insights into the mechanisms underlying DLI-induced transplant tolerance and suggest that DN Tregs have great potential as an antigen-specific immune therapy to enhance allograft survival.
Collapse
Affiliation(s)
- Julia Gao
- University of Toronto Transplantation Institute, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Megan S. Ford. McIntyre
- University of Toronto Transplantation Institute, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Cheryl A. D'Souza
- University of Toronto Transplantation Institute, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Li Zhang
- University of Toronto Transplantation Institute, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
22
|
Abstract
Islet transplantation is today an accepted modality for treating selected patients with frequent hypoglycemic events or severe glycemic lability. Despite tremendous progress in islet isolation, culture, and preservation, clinical use is still restricted to a limited subset, and lifelong immunosuppression is required. Issues surrounding limited islet revascularization and immune destruction remain. One of the major challenges is to prevent alloreactivity and recurrence of autoimmunity against β-cells. These two hurdles can be effectively reduced by immunosuppressive therapy combining induction and maintenance treatments. The introduction of highly potent and selective biologic agents has significantly reduced the frequency of acute rejection and has prolonged graft survival, while minimizing the complications of this therapeutic scheme. This review will address the most important biological agents used in islet transplantation. We provide a historical perspective of their introduction into clinical practice and their role in current clinical protocols, aiming at improved engraftment efficiency, increased long-term survival, and better overall results of clinical islet transplantation.
Collapse
Affiliation(s)
- Boris Gala-Lopez
- Clinical Islet Transplant Program and Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | | | | |
Collapse
|
23
|
Revascularization of transplanted pancreatic islets and role of the transplantation site. Clin Dev Immunol 2013; 2013:352315. [PMID: 24106517 PMCID: PMC3782812 DOI: 10.1155/2013/352315] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Accepted: 08/09/2013] [Indexed: 12/16/2022]
Abstract
Since the initial reporting of the successful reversal of hyperglycemia through the transplantation of pancreatic islets, significant research efforts have been conducted in elucidating the process of revascularization and the influence of engraftment site on graft function and survival. During the isolation process the intrinsic islet vascular networks are destroyed, leading to impaired revascularization after transplant. As a result, in some cases a significant quantity of the beta cell mass transplanted dies acutely following the infusion into the portal vein, the most clinically used site of engraftment. Subsequently, despite the majority of patients achieving insulin independence after transplant, a proportion of them recommence small, supplemental exogenous insulin over time. Herein, this review considers the process of islet revascularization after transplant, its limiting factors, and potential strategies to improve this critical step. Furthermore, we provide a characterization of alternative transplant sites, analyzing the historical evolution and their role towards advancing transplant outcomes in both the experimental and clinical settings.
Collapse
|
24
|
Banham G, Prezzi D, Harford S, Taylor CJ, Hamer R, Higgins R, Bradley JA, Clatworthy MR. Elevated pretransplantation soluble BAFF is associated with an increased risk of acute antibody-mediated rejection. Transplantation 2013; 96:413-20. [PMID: 23842189 PMCID: PMC4170143 DOI: 10.1097/tp.0b013e318298dd65] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND B cells play an important role in renal allograft pathology, particularly in acute and chronic antibody-mediated rejection (AMR). B-cell activating factor belonging to the tumor necrosis factor family (BAFF; also known as BLyS) is a cytokine that enhances B-cell survival and proliferation. METHODS We analyzed serum BAFF levels in 32 patients undergoing antibody-incompatible (Ai) renal transplantation and 319 antibody-compatible transplant recipients and sought to determine whether there was a correlation with acute rejection and with transplant function and survival. RESULTS We demonstrate that, in patients undergoing Ai transplantation, elevated serum BAFF levels at baseline (before both antibody removal/desensitization and transplantation) are associated with an increased risk of subsequent AMR. In antibody-compatible transplant recipients at lower risk of AMR, no statistically significant association was observed between pretransplantation serum BAFF and AMR. CONCLUSIONS These data raise the possibility that, in high immunologic risk patients undergoing Ai transplantation, the presence of elevated pretransplantation serum BAFF might identify those at increased risk of AMR. BAFF neutralization may be an interesting therapeutic strategy to explore in these patients, particularly because such agents are available and have already been used in the treatment of autoimmunity.
Collapse
Affiliation(s)
- Gemma Banham
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Davide Prezzi
- Department of Surgery, University of Cambridge, Cambridge, UK
| | - Sarah Harford
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Craig J. Taylor
- Tissue Typing Laboratory, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Rizwan Hamer
- Transplant Unit, University Hospitals Coventry and Warwickshire, and Warwick Medical School, University of Warwick, Coventry, West Midlands, UK
| | - Rob Higgins
- Transplant Unit, University Hospitals Coventry and Warwickshire, and Warwick Medical School, University of Warwick, Coventry, West Midlands, UK
| | | | | |
Collapse
|
25
|
Nuclear antigens and auto/alloantibody responses: friend or foe in transplant immunology. Clin Dev Immunol 2013; 2013:267156. [PMID: 23690821 PMCID: PMC3649457 DOI: 10.1155/2013/267156] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Accepted: 03/19/2013] [Indexed: 02/07/2023]
Abstract
In addition to cellular immune responses, humoral immune responses, mediated by natural antibodies, autoantibodies, and alloantibodies, have increasingly been recognized as causes of organ transplant rejection. In our previous studies, we have demonstrated the induction of antinuclear antibodies against histone H1 and high-mobility group box 1 (HMGB1), in both experimental and clinical liver transplant tolerance. The active induction of antinuclear antibodies is usually an undesirable phenomenon, but it is often observed after liver transplantation. However, the release of nuclear antigens and its suppression by neutralizing antibodies are proposed to be important in the initiation and regulation of immune responses. In this review article, we summarize the current understanding of nuclear antigens and corresponding antinuclear regulatory antibodies (Abregs) on infection, injury, inflammation, transplant rejection, and tolerance induction and discuss the significance of nuclear antigens as diagnostic and therapeutic targets.
Collapse
|
26
|
Non-invasive biomarkers to guide management following renal transplantation: the need for a multiplatform approach. Curr Opin Organ Transplant 2013; 18:1-5. [PMID: 23283248 DOI: 10.1097/mot.0b013e32835c8025] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
PURPOSE OF REVIEW Balancing the level of anti-rejection therapy is the key challenge facing clinicians managing recipients of a solid organ transplant. Identification of biomarkers in non-invasive samples will allow serial monitoring which can prompt changes in therapy at an earlier stage without the need for invasive tests, and before significant damage to the graft or side effects have occurred. In this review we provide an update on the present status of such biomarker discovery in renal transplantation. RECENT FINDINGS Previous studies focusing on candidate biomarkers have identified a number of genes that have the potential to identify patients undergoing rejection. Advances in technology now allow screening of samples for markers which have led to identification of further genes as well as adding microRNAs and proteins to the list. Similarly, by studying patients in whom anti-rejection therapy has been withdrawn, a gene signature for operational tolerance has been described. SUMMARY It has become clear that to obtain a test suitable for clinical purposes, combinations of markers are required to identify specific clinical phenotypes. Identification and validation of such marker sets in large cohorts is urgently required to allow progression to clinical trials with the ultimate goal of offering recipients personalized anti-rejection therapy regimes.
Collapse
|
27
|
B-cell-related biomarkers of tolerance are up-regulated in rejection-free kidney transplant recipients. Transplantation 2013; 95:148-54. [PMID: 23222918 DOI: 10.1097/tp.0b013e3182789a24] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Molecular signatures have recently been identified in operationally tolerant long-term kidney transplant patients; however, their expression in patients on immunosuppression remains unclear. METHODS In this prospective study, the gene expression profiles of eight selected tolerance-associated genes (MS4A1, CD79B, TCL1A, TMEM176B, FOXP3, TOAG-1, MAN1A1, and TLR5) in the peripheral blood of 67 kidney transplant recipients at days 0, 7, 14, 21, 28, 60, 90, and at 6 and 12 months, and in graft biopsies were measured. Similarly, using flow cytometry, CD45CD19CD3 B-cell counts were evaluated in the follow-up. Expression patterns were compared among patients with biopsy-proven acute rejection, borderline changes, and in rejection-free patients. A generalized linear mixed model with gamma distribution for repeated measures adjusted for induction therapy was used for statistical analysis of longitudinal data and Kruskal-Wallis test for case biopsy data. RESULTS Compared to patients with rejection, a significantly higher number of peripheral B cells were observed during follow-up in rejection-free patients and in patients with borderline changes. Gene expression patterns of MS4A1 (CD20), TCL1A, CD79B, TOAG-1, and FOXP3 genes were significantly higher in rejection-free patients as compared to rejection group with the highest differences during the first 3 months. In contrast, TMEM176B (TORID) was up-regulated in the rejection group. Similar trends were also observed between patients with borderline changes and acute rejection. Higher intragraft expression of TOAG-1 was observed in rejection-free patients. CONCLUSIONS These observations suggest an association of B-cell signatures, seen also in drug-free tolerant patients, with controlled alloimmune response.
Collapse
|
28
|
Bronchiolitis obliterans syndrome, hypogammaglobulinemia, and infectious complications of lung transplantation. J Heart Lung Transplant 2013; 32:36-43. [DOI: 10.1016/j.healun.2012.10.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Revised: 09/17/2012] [Accepted: 10/17/2012] [Indexed: 11/18/2022] Open
|
29
|
Abu-Elmagd KM, Wu G, Costa G, Lunz J, Martin L, Koritsky DA, Murase N, Irish W, Zeevi A. Preformed and de novo donor specific antibodies in visceral transplantation: long-term outcome with special reference to the liver. Am J Transplant 2012; 12:3047-60. [PMID: 22947059 DOI: 10.1111/j.1600-6143.2012.04237.x] [Citation(s) in RCA: 149] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Despite improvement in early outcome, rejection particularly chronic allograft enteropathy continues to be a major barrier to long-term visceral engraftment. The potential role of donor specific antibodies (DSA) was examined in 194 primary adult recipients. All underwent complement-dependent lymphocytotoxic crossmatch (CDC-XM) with pre- and posttransplant solid phase HLA-DSA assay in 156 (80%). Grafts were ABO-identical with random HLA-match. Liver was included in 71 (37%) allografts. Immunosuppression was tacrolimus-based with antilymphocyte recipient pretreatment in 150 (77%). CDC-XM was positive in 55 (28%). HLA-DSA was detectable before transplant in 49 (31%) recipients with 19 continuing to have circulating antibodies. Another 19 (18%) developed de novo DSA. Ninety percent of patients with preformed DSA harbored HLA Class-I whereas 74% of recipients with de novo antibodies had Class-II. Gender, age, ABO blood-type, cold ischemia, splenectomy and allograft type were significant DSA predictors. Preformed DSA significantly (p < 0.05) increased risk of acute rejection. Persistent and de novo HLA-DSA significantly (p < 0.001) increased risk of chronic rejection and associated graft loss. Inclusion of the liver was a significant predictor of better outcome (p = 0.004, HR = 0.347) with significant clearance of preformed antibodies (p = 0.04, OR = 56) and lower induction of de novo DSA (p = 0.07, OR = 24). Innovative multifaceted anti-DSA strategies are required to further improve long-term survival particularly of liver-free allografts.
Collapse
Affiliation(s)
- K M Abu-Elmagd
- Department of Surgery Department of Pathology, Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, PA, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Cellular Infiltrates and NFκB Subunit c-Rel Signaling in Kidney Allografts of Patients With Clinical Operational Tolerance. Transplantation 2012; 94:729-37. [DOI: 10.1097/tp.0b013e31826032be] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
31
|
Lehnhardt A, Dunst F, van Husen M, Loos S, Oh J, Eiermann T, Koch M, Kemper MJ. Elevated serum levels of B-cell activating factor in pediatric renal transplant patients. Pediatr Nephrol 2012; 27:1389-95. [PMID: 22453734 DOI: 10.1007/s00467-012-2142-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Revised: 02/09/2012] [Accepted: 02/10/2012] [Indexed: 11/29/2022]
Abstract
BACKGROUND B-cells are increasingly recognized as important players in alloimmunity. B cell-activating factor (BAFF) and its receptor BAFF-R are essential for B-cell differentiation and survival. Data on BAFF levels in pediatric renal transplant (RT) patients are scarce. OBJECTIVE It is known from adult data that elevated BAFF levels correlate with an unfavorable outcome in bone marrow and kidney recipients. To analyze this hypothesis in pediatric renal transplant patients we performed a cross-sectional analysis of serum BAFF levels, lymphocyte surface BAFF-R expression, and clinical variables in a cohort of 43 pediatric renal transplant patients. METHODS We studied serum BAFF, CD19+ B-, and FoxP3+ regulatory T-cells (Tregs) and BAFF-R expression in 43 children 2.9 (0.1-12.4) years after RT on maintenance immunosuppression. Twenty-two healthy children and 19 children with chronic kidney disease stage 5 (CKD5) served as controls. RESULTS BAFF levels were significantly higher in RT patients than in healthy children (1,435±574 vs 894±189 pg/mL; p<0.0001) whereas numbers of B-cells and Tregs were significantly lower. BAFF-R expression on B-cells was decreased after RT (531±334 vs 707±257 MFI; p<0.005), BAFF inversely correlated with BAFF-R (r=-0.5022, p<0.006), but not with B-cell count. BAFF was elevated in CKD5 patients (1,276±294 pg/mL). In RT patients BAFF was significantly higher in those with eGFR <60 ml/min/1.73m(2) (1,553±447 vs 1,234±323 pg/mL; p=0.02). BAFF levels and BAFF-R expression did not correlate with HLA antibody status, time after transplantation, age or gender of the patients. CONCLUSION Serum BAFF concentrations were significantly elevated in pediatric RT patients. They correlated with decreased BAFF-R expression on CD19+ B-cells and impaired allograft function. Our findings of a dysregulated BAFF/BAFF-R axis may be of clinical relevance after renal transplantation and therefore underline the importance of further research into BAFF-dependent mechanisms.
Collapse
Affiliation(s)
- Anja Lehnhardt
- University Medical Center Hamburg-Eppendorf, Pediatric Nephrology, Martinistrasse 52, 20246 Hamburg, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Heidt S, Hester J, Shankar S, Friend PJ, Wood KJ. B cell repopulation after alemtuzumab induction-transient increase in transitional B cells and long-term dominance of naïve B cells. Am J Transplant 2012; 12:1784-92. [PMID: 22420490 PMCID: PMC3387484 DOI: 10.1111/j.1600-6143.2012.04012.x] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In organ transplantation, the composition of the B-cell compartment is increasingly identified as an important determinant for graft outcome. Whereas naïve and transitional B cells have been associated with long-term allograft survival and operational tolerance, memory B cells have been linked to decreased allograft survival. Alemtuzumab induction therapy effectively depletes B cells, but is followed by rapid repopulation up to levels exceeding base line. The characteristics of the repopulating B cells are currently unknown. We studied the phenotypic and functional characteristics of B cells longitudinally in 19 kidney transplant recipients, before and at 6, 9 and 12 months after alemtuzumab induction therapy. A transient increase in transitional B cells and cells with phenotypic characteristics of regulatory B cells, as well as a long-term dominance in naïve B cells was found in alemtuzumab-treated kidney transplant recipients, which was not influenced by conversion from tacrolimus to sirolimus. At all time-points after treatment, B cells showed unaltered proliferative and IgM-producing capacity as compared to pretransplant samples, whereas the ability to produce IgG was inhibited long-term. In conclusion, induction therapy with alemtuzumab results in a long-term shift toward naïve B cells with altered phenotypic and functional characteristics.
Collapse
Affiliation(s)
- S Heidt
- Transplant Research Immunology Group, Nuffield Department of Surgical Sciences, University of OxfordOxford, UK
| | - J Hester
- Transplant Research Immunology Group, Nuffield Department of Surgical Sciences, University of OxfordOxford, UK
| | - S Shankar
- Transplant Research Immunology Group, Nuffield Department of Surgical Sciences, University of OxfordOxford, UK
| | - P J Friend
- Oxford Transplant Centre, Nuffield Department of Surgical Sciences, University of OxfordOxford, UK
| | - K J Wood
- Transplant Research Immunology Group, Nuffield Department of Surgical Sciences, University of OxfordOxford, UK,*Corresponding author: Kathryn J. Wood,
| |
Collapse
|
33
|
Heidt S, Hester J, Shankar S, Friend PJ, Wood KJ. B cell repopulation after alemtuzumab induction-transient increase in transitional B cells and long-term dominance of naïve B cells. Am J Transplant 2012. [PMID: 22420490 DOI: 10.1111/j.1600-6143.2012.04 012.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
In organ transplantation, the composition of the B-cell compartment is increasingly identified as an important determinant for graft outcome. Whereas naïve and transitional B cells have been associated with long-term allograft survival and operational tolerance, memory B cells have been linked to decreased allograft survival. Alemtuzumab induction therapy effectively depletes B cells, but is followed by rapid repopulation up to levels exceeding base line. The characteristics of the repopulating B cells are currently unknown. We studied the phenotypic and functional characteristics of B cells longitudinally in 19 kidney transplant recipients, before and at 6, 9 and 12 months after alemtuzumab induction therapy. A transient increase in transitional B cells and cells with phenotypic characteristics of regulatory B cells, as well as a long-term dominance in naïve B cells was found in alemtuzumab-treated kidney transplant recipients, which was not influenced by conversion from tacrolimus to sirolimus. At all time-points after treatment, B cells showed unaltered proliferative and IgM-producing capacity as compared to pretransplant samples, whereas the ability to produce IgG was inhibited long-term. In conclusion, induction therapy with alemtuzumab results in a long-term shift toward naïve B cells with altered phenotypic and functional characteristics.
Collapse
Affiliation(s)
- S Heidt
- Transplant Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | | | | | | | | |
Collapse
|
34
|
Carvello M, Petrelli A, Vergani A, Lee KM, Tezza S, Chin M, Orsenigo E, Staudacher C, Secchi A, Dunussi-Joannopoulos K, Sayegh MH, Markmann JF, Fiorina P. Inotuzumab ozogamicin murine analog-mediated B-cell depletion reduces anti-islet allo- and autoimmune responses. Diabetes 2012; 61:155-65. [PMID: 22076927 PMCID: PMC3237644 DOI: 10.2337/db11-0684] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
B cells participate in the priming of the allo- and autoimmune responses, and their depletion can thus be advantageous for islet transplantation. Herein, we provide an extensive study of the effect of B-cell depletion in murine models of islet transplantation. Islet transplantation was performed in hyperglycemic B-cell-deficient(μMT) mice, in a purely alloimmune setting (BALB/c into hyperglycemic C57BL/6), in a purely autoimmune setting (NOD.SCID into hyperglycemic NOD), and in a mixed allo-/autoimmune setting (BALB/c into hyperglycemic NOD). Inotuzumab ozogamicin murine analog (anti-CD22 monoclonal antibody conjugated with calicheamicin [anti-CD22/cal]) efficiently depleted B cells in all three models of islet transplantation examined. Islet graft survival was significantly prolonged in B-cell-depleted mice compared with control groups in transplants of islets from BALB/c into C57BL/6 (mean survival time [MST]: 16.5 vs. 12.0 days; P = 0.004), from NOD.SCID into NOD (MST: 23.5 vs. 14.0 days; P = 0.03), and from BALB/c into NOD (MST: 12.0 vs. 5.5 days; P = 0.003). In the BALB/c into B-cell-deficient mice model, islet survival was prolonged as well (MST: μMT = 32.5 vs. WT = 14 days; P = 0.002). Pathology revealed reduced CD3(+) cell islet infiltration and confirmed the absence of B cells in treated mice. Mechanistically, effector T cells were reduced in number, concomitant with a peripheral Th2 profile skewing and ex vivo recipient hyporesponsiveness toward donor-derived antigen as well as islet autoantigens. Finally, an anti-CD22/cal and CTLA4-Ig-based combination therapy displayed remarkable prolongation of graft survival in the stringent model of islet transplantation (BALB/c into NOD). Anti-CD22/cal-mediated B-cell depletion promotes the reduction of the anti-islet immune response in various models of islet transplantation.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal, Humanized/chemistry
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibody-Dependent Cell Cytotoxicity/drug effects
- Antibody-Dependent Cell Cytotoxicity/physiology
- Autoimmunity/drug effects
- B-Lymphocytes/cytology
- B-Lymphocytes/drug effects
- B-Lymphocytes/immunology
- Cell Death/drug effects
- Cell Death/immunology
- Cells, Cultured
- Female
- Graft Survival/immunology
- Inotuzumab Ozogamicin
- Islets of Langerhans/drug effects
- Islets of Langerhans/immunology
- Islets of Langerhans Transplantation/immunology
- Islets of Langerhans Transplantation/pathology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Mice, SCID
- Mice, Transgenic
- Transplantation Tolerance/drug effects
- Transplantation Tolerance/immunology
Collapse
Affiliation(s)
- Michele Carvello
- Transplant Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Surgery, San Raffaele Scientific Institute, Milan, Italy
| | - Alessandra Petrelli
- Nephrology Division, Transplantation Research Center, Children’s Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Medicine, San Raffaele Scientific Institute, Milan, Italy
| | - Andrea Vergani
- Nephrology Division, Transplantation Research Center, Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Kang Mi Lee
- Transplant Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Sara Tezza
- Nephrology Division, Transplantation Research Center, Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Melissa Chin
- Nephrology Division, Transplantation Research Center, Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Elena Orsenigo
- Department of Surgery, San Raffaele Scientific Institute, Milan, Italy
| | - Carlo Staudacher
- Department of Surgery, San Raffaele Scientific Institute, Milan, Italy
| | - Antonio Secchi
- Department of Medicine, San Raffaele Scientific Institute, Milan, Italy
| | | | - Mohamed H. Sayegh
- Nephrology Division, Transplantation Research Center, Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - James F. Markmann
- Transplant Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Paolo Fiorina
- Nephrology Division, Transplantation Research Center, Children’s Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Medicine, San Raffaele Scientific Institute, Milan, Italy
- Corresponding author: Paolo Fiorina,
| |
Collapse
|
35
|
Zarkhin V, Sarwal MM. The coin toss of B cells in rejection and tolerance: danger versus defense. Semin Immunol 2011; 24:86-91. [PMID: 22035649 DOI: 10.1016/j.smim.2011.09.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2011] [Accepted: 09/30/2011] [Indexed: 01/27/2023]
Abstract
Transplantation is the preferred therapy for the end stage organ disease. Since the introduction of organ transplantation into medical practice in 1953 [1], significant progress has been achieved in patient and graft survival rates due to improvements in surgical techniques and more targeted immunosuppressive medications [2]. Nevertheless, current gaps in the management of the transplant patient stem from an incomplete understanding about the heterogeneity of the injury response in organ transplantation, at different rates and different time points after transplantation, as well as our inability to monitor the immunologic threshold of risk versus safety in each individual patient. Recent advances in immunology/transplantation biology with the advent of high throughput "omic" assays such as gene microarrays, proteomics, metabolomics, antibiomics, chemical genomics and functional imaging with nanoparticles, offers us unique methods to interrogate and decipher the variability and unpredictability of the immune response in organ transplantation (Fig. 1) [3]. Recent studies using these applications [3-8] have uncovered a critical and pivotal role for specific B cell lineages in organ injury [9] and organ acceptance [10,11] (Fig. 2). The availability of specific therapies against some of these defined B cell populations provides for an exciting new field of B cell targeted manipulation that can both abrogate the allospecific injury response, as well as promote allospecific graft accommodation and health.
Collapse
Affiliation(s)
- Valeriya Zarkhin
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | | |
Collapse
|
36
|
Azimzadeh AM, Lees JR, Ding Y, Bromberg JS. Immunobiology of transplantation: impact on targets for large and small molecules. Clin Pharmacol Ther 2011; 90:229-42. [PMID: 21716276 DOI: 10.1038/clpt.2011.106] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Organ transplantation is the preferred method of treatment for many forms of end-stage organ failure. However, immunosuppressive drugs that are used to avoid rejection can result in numerous undesirable effects (infection, malignancy, hypertension, diabetes, and accelerated arteriosclerosis). Moreover, they are not effective at preventing chronic rejection resulting in late graft loss. This review summarizes the fundamental concepts underlying the rejection of solid-organ allografts with the aim of highlighting potential new targets for therapeutics. Future improvement will depend on new therapeutic moieties, including biologics, to target various pathways of both the innate and adaptive arms of immunity. Results from some of the most recent clinical trials in transplantation and emerging new therapies are also discussed.
Collapse
Affiliation(s)
- A M Azimzadeh
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | | | | |
Collapse
|
37
|
Perucha E, Rebollo-Mesa I, Sagoo P, Hernandez-Fuentes MP. Biomarkers of tolerance: searching for the hidden phenotype. Kidney Int Suppl (2011) 2011; 1:40-46. [PMID: 25018902 PMCID: PMC4089724 DOI: 10.1038/kisup.2011.11] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Induction of transplantation tolerance remains the ideal long-term clinical and logistic solution to the current challenges facing the management of renal allograft recipients. In this review, we describe the recent studies and advances made in identifying biomarkers of renal transplant tolerance, from study inceptions, to the lessons learned and their implications for current and future studies with the same goal. With the age of biomarker discovery entering a new dimension of high-throughput technologies, here we also review the current approaches, developments, and pitfalls faced in the subsequent statistical analysis required to identify valid biomarker candidates.
Collapse
Affiliation(s)
- Esperanza Perucha
- Section of Experimental Immunobiology, Division of Transplantation Immunology and Mucosal Biology, King's College London, London, UK
- Medical Research Centre for Transplantation, King's College London, Guy's Hospital, London, UK
- NIHR Biomedical Research Centre, Guy's and St Thomas' Hospital and King's College London, London, UK
| | - Irene Rebollo-Mesa
- Medical Research Centre for Transplantation, King's College London, Guy's Hospital, London, UK
| | - Pervinder Sagoo
- Section of Experimental Immunobiology, Division of Transplantation Immunology and Mucosal Biology, King's College London, London, UK
- NIHR Biomedical Research Centre, Guy's and St Thomas' Hospital and King's College London, London, UK
- Section of Immunoregulation and Immune Intervention, Division of Transplantation Immunology and Mucosal Biology, King's College London, London, UK
| | - Maria P Hernandez-Fuentes
- Section of Experimental Immunobiology, Division of Transplantation Immunology and Mucosal Biology, King's College London, London, UK
- Medical Research Centre for Transplantation, King's College London, Guy's Hospital, London, UK
- NIHR Biomedical Research Centre, Guy's and St Thomas' Hospital and King's College London, London, UK
| |
Collapse
|
38
|
Abstract
There has been increasing interest in the role played by B cells, plasma cells and their associated antibody in the immune response to an allograft, driven by the need to undertake antibody-incompatible transplantation and evidence suggesting that B cells play a role in acute cellular rejection and in acute and chronic antibody-mediated rejection. A number of immunosuppressive agents have emerged which target B cells, plasma cells and/or antibody, for example, the B cell-depleting CD20 antibody rituximab. This review describes recent developments in the use of such agents, our understanding of the role of B cells in alloimmunity and the application of this knowledge toward novel therapies in transplantation. It also considers the evidence to date suggesting that B cells may act as regulators of an alloimmune response. Thus, future attempts to target B cells will need to address the problem of how to inhibit effector B cells, while enhancing those with regulatory capacity.
Collapse
Affiliation(s)
- M R Clatworthy
- Cambridge Institute for Medical Research, University of Cambridge School of Clinical Medicine, Box 139 Addenbrooke's Hospital, Hills Road, Cambridge CB2 0XY, Cambridge. ;
| |
Collapse
|