1
|
Kenjo A, Sato N, Azuma T, Nishimagi A, Tsukida S, Suzushino S, Muto M, Chiba H, Watanabe J, Haga J, Kofunato Y, Ishigame T, Kimura T, Marubashi S. Clinical outcomes of simultaneous pancreas-kidney transplantation: experience of a single center. Fukushima J Med Sci 2025; 71:119-128. [PMID: 39909445 PMCID: PMC12079050 DOI: 10.5387/fms.24-00052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 11/05/2024] [Indexed: 02/07/2025] Open
Abstract
This study evaluated the outcomes of simultaneous pancreas-kidney transplantation (SPK) at Fukushima Medical University between 2001 and 2024. We retrospectively reviewed ten adult patients who underwent SPK. We aimed to clarify the important aspects of patient management in patients undergoing SPK, focusing on perioperative outcomes and long-term complications.The median postoperative observation period was 1,968 days. Postoperative complications were observed in all patients. Most were classified as Clavien-Dindo (CD) grade I or II, but major complications, including CD grade IIIa or higher, were observed in four patients (40%) with zero mortality. Despite the loss of one kidney graft due to primary non-function, the 5-year survival rates for both patients and pancreatic grafts remained at 100%. However, there was one case each of pancreatic graft loss, kidney graft loss, and patient death after 5 years post-transplantation, all attributed to late-onset complications, including recurrent type 1 diabetes, focal segmental glomerulosclerosis, and cardiovascular disease.The short-term outcomes of SPK at our institution were favorable, with a trend toward a reduction in the comprehensive complication index (CCI) observed in the latter five cases compared with the first five cases, suggesting potential improvements in perioperative management. Long-term monitoring and collaboration with physicians are essential to enhance patient outcomes. .
Collapse
Affiliation(s)
- Akira Kenjo
- Department of Hepato-Biliary-Pancreatic and Transplant Surgery, Fukushima Medical University
| | - Naoya Sato
- Department of Hepato-Biliary-Pancreatic and Transplant Surgery, Fukushima Medical University
| | - Takayasu Azuma
- Department of Hepato-Biliary-Pancreatic and Transplant Surgery, Fukushima Medical University
| | - Atsushi Nishimagi
- Department of Hepato-Biliary-Pancreatic and Transplant Surgery, Fukushima Medical University
| | - Shigeyuki Tsukida
- Department of Hepato-Biliary-Pancreatic and Transplant Surgery, Fukushima Medical University
| | - Seiko Suzushino
- Department of Hepato-Biliary-Pancreatic and Transplant Surgery, Fukushima Medical University
| | - Makoto Muto
- Department of Hepato-Biliary-Pancreatic and Transplant Surgery, Fukushima Medical University
| | - Hiroto Chiba
- Department of Hepato-Biliary-Pancreatic and Transplant Surgery, Fukushima Medical University
| | - Junichiro Watanabe
- Department of Hepato-Biliary-Pancreatic and Transplant Surgery, Fukushima Medical University
| | - Junichiro Haga
- Department of Hepato-Biliary-Pancreatic and Transplant Surgery, Fukushima Medical University
| | - Yasuhide Kofunato
- Department of Hepato-Biliary-Pancreatic and Transplant Surgery, Fukushima Medical University
| | - Teruhide Ishigame
- Department of Hepato-Biliary-Pancreatic and Transplant Surgery, Fukushima Medical University
| | - Takashi Kimura
- Department of Hepato-Biliary-Pancreatic and Transplant Surgery, Fukushima Medical University
| | - Shigeru Marubashi
- Department of Hepato-Biliary-Pancreatic and Transplant Surgery, Fukushima Medical University
| |
Collapse
|
2
|
Burke GW, Mitrofanova A, Fontanella AM, Vendrame F, Ciancio G, Vianna RM, Roth D, Ruiz P, Abitbol CL, Chandar J, Merscher S, Pugliese A, Fornoni A. Transplantation: platform to study recurrence of disease. Front Immunol 2024; 15:1354101. [PMID: 38495894 PMCID: PMC10940352 DOI: 10.3389/fimmu.2024.1354101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 01/29/2024] [Indexed: 03/19/2024] Open
Abstract
Beyond the direct benefit that a transplanted organ provides to an individual recipient, the study of the transplant process has the potential to create a better understanding of the pathogenesis, etiology, progression and possible therapy for recurrence of disease after transplantation while at the same time providing insight into the original disease. Specific examples of this include: 1) recurrence of focal segmental glomerulosclerosis (FSGS) after kidney transplantation, 2) recurrent autoimmunity after pancreas transplantation, and 3) recurrence of disease after orthotopic liver transplantation (OLT) for cirrhosis related to progressive steatosis secondary to jejuno-ileal bypass (JIB) surgery. Our team has been studying these phenomena and their immunologic underpinnings, and we suggest that expanding the concept to other pathologic processes and/or transplanted organs that harbor the risk for recurrent disease may provide novel insight into the pathogenesis of a host of other disease processes that lead to organ failure.
Collapse
Affiliation(s)
- George William Burke
- Division of Kidney-Pancreas Transplantation, Department of Surgery, Miami Transplant Institute, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Alla Mitrofanova
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, United States
| | | | - Francesco Vendrame
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Gaetano Ciancio
- Division of Kidney-Pancreas Transplantation, Department of Surgery, Miami Transplant Institute, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Rodrigo M. Vianna
- Department of Surgery, Miami Transplant Institute, University of Miami Miller School of Medicine, Miami, FL, United States
| | - David Roth
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Phillip Ruiz
- Transplant Pathology, Immunology and Histocompatibility Laboratory University of Miami Department of Surgery, Miami Transplant Institute, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Carolyn L. Abitbol
- Pediatric Nephrology & Hypertension, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Jayanthi Chandar
- Pediatric Kidney Transplant, Miami Transplant Institute, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Sandra Merscher
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, United States
- Peggy and Harold Katz Family Drug Discovery Center, Department of Medicine, University of Miami - Miller School of Medicine, Miami, FL, United States
| | - Alberto Pugliese
- Department of Diabetes Immunology, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, United States
| | - Alessia Fornoni
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, United States
- Peggy and Harold Katz Family Drug Discovery Center, Department of Medicine, University of Miami - Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
3
|
Drachenberg CB, Buettner-Herold M, Aguiar PV, Horsfield C, Mikhailov AV, Papadimitriou JC, Seshan SV, Perosa M, Boggi U, Uva P, Rickels M, Grzyb K, Arend L, Cuatrecasas M, Toniolo MF, Farris AB, Renaudin K, Zhang L, Roufousse C, Gruessner A, Gruessner R, Kandaswamy R, White S, Burke G, Cantarovich D, Parsons RF, Cooper M, Kudva YC, Kukla A, Haririan A, Parajuli S, Merino-Torres JF, Argente-Pla M, Meier R, Dunn T, Ugarte R, Rao JS, Vistoli F, Stratta R, Odorico J. Banff 2022 pancreas transplantation multidisciplinary report: Refinement of guidelines for T cell-mediated rejection, antibody-mediated rejection and islet pathology. Assessment of duodenal cuff biopsies and noninvasive diagnostic methods. Am J Transplant 2024; 24:362-379. [PMID: 37871799 DOI: 10.1016/j.ajt.2023.10.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 10/02/2023] [Accepted: 10/11/2023] [Indexed: 10/25/2023]
Abstract
The Banff pancreas working schema for diagnosis and grading of rejection is widely used for treatment guidance and risk stratification in centers that perform pancreas allograft biopsies. Since the last update, various studies have provided additional insight regarding the application of the schema and enhanced our understanding of additional clinicopathologic entities. This update aims to clarify terminology and lesion description for T cell-mediated and antibody-mediated allograft rejections, in both active and chronic forms. In addition, morphologic and immunohistochemical tools are described to help distinguish rejection from nonrejection pathologies. For the first time, a clinicopathologic approach to islet pathology in the early and late posttransplant periods is discussed. This update also includes a discussion and recommendations on the utilization of endoscopic duodenal donor cuff biopsies as surrogates for pancreas biopsies in various clinical settings. Finally, an analysis and recommendations on the use of donor-derived cell-free DNA for monitoring pancreas graft recipients are provided. This multidisciplinary effort assesses the current role of pancreas allograft biopsies and offers practical guidelines that can be helpful to pancreas transplant practitioners as well as experienced pathologists and pathologists in training.
Collapse
Affiliation(s)
| | - Maike Buettner-Herold
- Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-University Erlangen-Nuremberg (FAU) and University Hospital, Erlangen, Germany
| | | | - Catherine Horsfield
- Department of Histopathology/Cytology, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Alexei V Mikhailov
- Department of Pathology, Atrium Health Wake Forest Baptist, Winston-Salem, North Carolina, USA
| | - John C Papadimitriou
- Department of Pathology, University of Maryland School of Medicine, Maryland, USA
| | - Surya V Seshan
- Division of Renal Pathology, Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, Cornell University, New York, New York, USA
| | - Marcelo Perosa
- Beneficência Portuguesa and Bandeirantes Hospital of São Paulo, São Paulo, Brazil
| | - Ugo Boggi
- Department of Surgery, University of Pisa, Pisa, The province of Pisa, Italy
| | - Pablo Uva
- Kidney/Pancreas Transplant Program, Instituto de Trasplantes y Alta Complejidad (ITAC - Nephrology), Buenos Aires, Argentina
| | - Michael Rickels
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, Philadelphia, USA
| | - Krzyztof Grzyb
- Department of Pathology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Lois Arend
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | | | | | - Alton B Farris
- Department of Pathology, Emory University School of Medicine, Atlanta, Georgia, USA
| | | | - Lizhi Zhang
- Division of Anatomic Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Candice Roufousse
- Department of Immunology and Inflammation, Imperial College of London, London, United Kingdom
| | - Angelika Gruessner
- Department of Nephrology/Medicine, State University of New York, New York, USA
| | - Rainer Gruessner
- Department of Surgery, State University of New York, New York, USA
| | - Raja Kandaswamy
- Division of Solid Organ Transplantation, Department of Surgery, University of Minnesota, Minneapolis, MN, USA
| | - Steven White
- Department of Surgery, Newcastle Upon Tyne NHS Foundation Trust, Newcastle upon Tyne, England, United Kingdom
| | - George Burke
- Division of Kidney-Pancreas Transplantation, Department of Surgery, Miami Transplant Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
| | | | - Ronald F Parsons
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Matthew Cooper
- Division of Transplant Surgery, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Yogish C Kudva
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Aleksandra Kukla
- Department of Medicine, Division of Nephrology and Hypertension, Mayo Clinic College of Medicine and Science, Mayo Clinic, Rochester, Minnesota, USA
| | - Abdolreza Haririan
- Department of Medicine, University of Maryland School of Medicine, Maryland, USA
| | - Sandesh Parajuli
- Department of Medicine, UWHealth Transplant Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Juan Francisco Merino-Torres
- Department of Endocrinology and Nutrition, University Hospital La Fe, La Fe Health Research Institute, University of Valencia, Valencia, Spain
| | - Maria Argente-Pla
- University Hospital La Fe, Health Research Institute La Fe, Valencia, Spain
| | - Raphael Meier
- Department of Surgery, University of Maryland School of Medicine, Maryland, USA
| | - Ty Dunn
- Division of Transplantation, Department of Surgery, Penn Transplant Institute, University of Pennsylvania, Pennsylvania, Philadelphia, USA
| | - Richard Ugarte
- Department of Medicine, University of Maryland School of Medicine, Maryland, USA
| | - Joseph Sushil Rao
- Division of Solid Organ Transplantation, Department of Surgery, University of Minnesota, Minneapolis, MN, USA; Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN, USA
| | - Fabio Vistoli
- Department of Surgery, University of Pisa, Pisa, The province of Pisa, Italy
| | - Robert Stratta
- Department of Surgery, Atrium Health Wake Forest Baptist, Winston-Salem, North Carolina, USA
| | - Jon Odorico
- Division of Transplantation, Department of Surgery, UWHealth Transplant Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| |
Collapse
|
4
|
Zou Y, Li S, Chen W, Xu J. Urine-derived stem cell therapy for diabetes mellitus and its complications: progress and challenges. Endocrine 2024; 83:270-284. [PMID: 37801228 DOI: 10.1007/s12020-023-03552-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 09/24/2023] [Indexed: 10/07/2023]
Abstract
Diabetes mellitus (DM) is a chronic and relentlessly progressive metabolic disease characterized by a relative or absolute deficiency of insulin in the body, leading to increased production of advanced glycosylation end products that further enhance oxidative and nitrosative stresses, often leading to multiple macrovascular (cardiovascular disease) and microvascular (e.g., diabetic nephropathy, diabetic retinopathy, and neuropathy) complications, representing the ninth leading cause of death worldwide. Existing medical treatments do not provide a complete cure for DM; thus, stem cell transplantation therapy has become the focus of research on DM and its complications. Urine-derived stem cells (USCs), which are isolated from fresh urine and have biological properties similar to those of mesenchymal stem cells (MSCs), were demonstrated to exert antiapoptotic, antifibrotic, anti-inflammatory, and proangiogenic effects through direct differentiation or paracrine mechanisms and potentially treat patients with DM. USCs also have the advantages of simple noninvasive sample collection procedures, minimal ethical issues, low cost, and easy cell isolation methods and thus have received more attention in regenerative therapies in recent years. This review outlines the biological properties of USCs and the research progress and current limitations of their role in DM and related complications. In summary, USCs have shown good versatility in treating hyperglycemia-impaired target organs in preclinical models, and many challenges remain in translating USC therapies to the clinic.
Collapse
Affiliation(s)
- Yun Zou
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Shanshan Li
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Wen Chen
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jixiong Xu
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Nanchang University, Nanchang, China.
| |
Collapse
|
5
|
Forbes S, Halpin A, Lam A, Grynoch D, Parker R, Hidalgo L, Bigam D, Anderson B, Dajani K, Kin T, O'Gorman D, Senior PA, Campbell P, Shapiro AJ. Islet transplantation outcomes in type 1 diabetes and transplantation of HLA-DQ8/DR4: results of a single-centre retrospective cohort in Canada. EClinicalMedicine 2024; 67:102333. [PMID: 38169703 PMCID: PMC10758748 DOI: 10.1016/j.eclinm.2023.102333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 11/07/2023] [Accepted: 11/09/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND In solid organ transplantation, HLA matching between donor and recipient is associated with superior outcomes. In islet transplantation, an intervention for Type 1 diabetes, HLA matching between donor and recipient is not performed as part of allocation. Susceptibility to Type 1 diabetes is associated with the presence of certain HLA types. This study was conducted to determine the impact of these susceptibility antigens on islet allograft survival. METHODS This is a single-centre retrospective cohort study. This cohort of transplant recipients (n = 268) received islets from 661 donor pancreases between March 11th, 1999 and August 29th, 2018 at the University of Alberta Hospital (Edmonton, AB, Canada). The frequency of the Type 1 diabetes susceptibility HLA antigens (HLA-A24, -B39, -DQ8, -DQ2 and-DQ2-DQA1∗05) in recipients and donors were determined. Recipient and donor HLA antigens were examined in relation to time to first C-peptide negative status/graft failure or last observation point. Taking into account multiple transplants per patient, we fitted a Gaussian frailty survival analysis model with baseline hazard function stratified by transplant number, adjusted for cumulative islet dose and other confounders. FINDINGS Across all transplants recipients of donors positive for HLA-DQ8 had significantly better graft survival (adjusted HRs 0.33 95% CI 0.17-0.66; p = 0.002). At first transplant only, donors positive for HLA-DQ2-DQA1∗05 had inferior graft survival (adjusted HR 1.96 95% CI 1.10-3.46); p = 0.02), although this was not significant in the frailty analysis taking multiple transplants into account (adjusted HR 1.46 95% CI 0.77-2.78; p = 0.25). Other HLA antigens were not associated with graft survival after adjustment for confounders. INTERPRETATION Our findings suggest islet transplantation from HLA-DQ8 donors is associated with superior graft outcomes. A donor positive for HLA-DQ2-DQA1∗05 at first transplant was associated with inferior graft survival but not when taking into account multiple transplants per recipient. The relevance of HLA-antigens on organ allocation needs further evaluation and inclusion in islet transplant registries and additional observational and interventional studies to evaluate the role of HLA-DQ8 in islet graft survival are required. FUNDING None.
Collapse
Affiliation(s)
- Shareen Forbes
- Clinical Islet Transplant Programme, University of Alberta, Edmonton, Canada
- Department of Surgery, University of Alberta, Edmonton, Canada
- Queen's Medical Research Institute, BHF Centre for Cardiovascular Science, University of Edinburgh, Scotland, UK
| | - Anne Halpin
- Alberta Precision Laboratories, University of Alberta, Laboratory Medicine and Pathology, University of Alberta, Edmonton, Canada
| | - Anna Lam
- Clinical Islet Transplant Programme, University of Alberta, Edmonton, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, University of Alberta, Edmonton, Canada
- Alberta Diabetes Institute, University of Alberta, Edmonton, Canada
| | - Don Grynoch
- Alberta Precision Laboratories, University of Alberta, Laboratory Medicine and Pathology, University of Alberta, Edmonton, Canada
| | - Richard Parker
- Edinburgh Clinical Trials Unit, Usher Institute, University of Edinburgh, Scotland, UK
| | - Luis Hidalgo
- Department of Surgery, University of Wisconsin, Madison, WI, USA
| | - David Bigam
- Department of Surgery, University of Alberta, Edmonton, Canada
| | - Blaire Anderson
- Department of Surgery, University of Alberta, Edmonton, Canada
| | - Khaled Dajani
- Department of Surgery, University of Alberta, Edmonton, Canada
| | - Tatsuya Kin
- Clinical Islet Transplant Programme, University of Alberta, Edmonton, Canada
| | - Doug O'Gorman
- Clinical Islet Transplant Programme, University of Alberta, Edmonton, Canada
| | - Peter A. Senior
- Clinical Islet Transplant Programme, University of Alberta, Edmonton, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, University of Alberta, Edmonton, Canada
- Alberta Diabetes Institute, University of Alberta, Edmonton, Canada
| | - Patricia Campbell
- Alberta Precision Laboratories, University of Alberta, Laboratory Medicine and Pathology, University of Alberta, Edmonton, Canada
| | - A.M. James Shapiro
- Clinical Islet Transplant Programme, University of Alberta, Edmonton, Canada
- Department of Surgery, University of Alberta, Edmonton, Canada
- Alberta Diabetes Institute, University of Alberta, Edmonton, Canada
| |
Collapse
|
6
|
Lemos JRN, Poggioli R, Ambut J, Bozkurt NC, Alvarez AM, Padilla N, Vendrame F, Ricordi C, Baidal DA, Alejandro R. Impact of GAD65 and IA2 autoantibodies on islet allograft survival. FRONTIERS IN CLINICAL DIABETES AND HEALTHCARE 2023; 4:1269758. [PMID: 38028981 PMCID: PMC10679328 DOI: 10.3389/fcdhc.2023.1269758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 10/02/2023] [Indexed: 12/01/2023]
Abstract
Introduction Islet transplantation (ITx) shows promise in treating T1D, but the role of islet autoantibodies on graft survival has not been clearly elucidated. We aimed to analyze the effect of GAD65 and IA2 autoantibody status on graft survival and attainment of insulin independence in subjects with T1D who underwent ITx. Method We conducted a retrospective cohort study on 47 ITx recipients from 2000 to 2018. Islet infusion was performed via intrahepatic portal (n=44) or onto the omentum via laparoscopic approach (n=3). Immunosuppression involved anti-IL2 receptor antibody, anti-TNF, and dual combinations of sirolimus, tacrolimus, or mycophenolate mofetil (Edmonton-like) in 38 subjects (80.9%). T-cell depletion induction with Edmonton-like maintenance was used in 9 subjects (19%). GAD65 and IA2 autoantibodies were assessed pre-transplant and post-transplant (monthly) until graft failure, and categorized as persistently negative, persistently positive, or seroconverters. Graft survival was analyzed using U-Mann-Whitney test, and Quade's nonparametric ANCOVA adjusted for confounders. Kaplan-Meier and Log-Rank tests were employed to analyze attainment of insulin independence. P value <0.05 indicated statistical significance. Results ITx recipients with persistent autoantibody negativity (n = 21) showed longer graft function (98 [61 - 182] months) than those with persistent autoantibody positivity (n = 18; 38 [13 - 163] months), even after adjusting for immunosuppressive induction protocol (P = 0.027). Seroconverters (n=8) had a median graft survival time of 73 (7.7 - 167) months, which did not significantly differ from the other 2 groups. Subjects with persistently single antibody positivity to GAD65 (n = 8) had shorter graft survival compared to negative islet autoantibody (GAD65/IA2) subjects (n = 21; P = 0.016). Time of graft survival did not differ in subjects with single antibody positivity to IA2. The proportion of insulin independence attainment was similar irrespective of autoantibody status. Conclusion The persistence of islet autoantibodies, as markers of islet autoimmunity, may represent an underappreciated contributing factor to the failure of transplanted β cells. Whether induction with T-cell depletion may lead to improved graft survival, independent of islet autoantibody status, could not be evaluated in our cohort. Larger prospective studies are needed to further address the role of islet autoantibody status on islet graft survival.
Collapse
Affiliation(s)
- Joana R. N. Lemos
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Raffaella Poggioli
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Jonathan Ambut
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Nujen C. Bozkurt
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Ana M. Alvarez
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Nathalia Padilla
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Francesco Vendrame
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, FL, United States
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Camillo Ricordi
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, FL, United States
- Division of Cellular Transplantation, Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
| | - David A. Baidal
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, FL, United States
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Rodolfo Alejandro
- Diabetes Research Institute (DRI) and Clinical Cell Transplant Program, University of Miami Miller School of Medicine, Miami, FL, United States
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
7
|
Battle R, Pritchard D, Peacock S, Hastie C, Worthington J, Jordan S, McCaughlan JA, Barnardo M, Cope R, Collins C, Diaz-Burlinson N, Rosser C, Foster L, Kallon D, Shaw O, Briggs D, Turner D, Anand A, Akbarzad-Yousefi A, Sage D. BSHI and BTS UK guideline on the detection of alloantibodies in solid organ (and islet) transplantation. Int J Immunogenet 2023; 50 Suppl 2:3-63. [PMID: 37919251 DOI: 10.1111/iji.12641] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 10/04/2023] [Indexed: 11/04/2023]
Abstract
Solid organ transplantation represents the best (and in many cases only) treatment option for patients with end-stage organ failure. The effectiveness and functioning life of these transplants has improved each decade due to surgical and clinical advances, and accurate histocompatibility assessment. Patient exposure to alloantigen from another individual is a common occurrence and takes place through pregnancies, blood transfusions or previous transplantation. Such exposure to alloantigen's can lead to the formation of circulating alloreactive antibodies which can be deleterious to solid organ transplant outcome. The purpose of these guidelines is to update to the previous BSHI/BTS guidelines 2016 on the relevance, assessment, and management of alloantibodies within solid organ transplantation.
Collapse
Affiliation(s)
- Richard Battle
- Scottish National Blood Transfusion Service, Edinburgh, UK
| | | | - Sarah Peacock
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | | | | | - Sue Jordan
- National Blood Service Tooting, London, UK
| | | | - Martin Barnardo
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Rebecca Cope
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | | | | | | | - Luke Foster
- Birmingham Blood Donor Centre, Birmingham, UK
| | | | - Olivia Shaw
- Guy's and St Thomas' NHS Foundation Trust, London, UK
| | | | - David Turner
- Scottish National Blood Transfusion Service, Edinburgh, UK
| | - Arthi Anand
- Imperial College Healthcare NHS Trust, London, UK
| | | | | |
Collapse
|
8
|
Stathi D, Johnston T, Hyslop R, Brackenridge A, Karalliedde J. Diabetes technology including automated insulin delivery systems to manage hyperglycemia in a failing pancreatic graft: Case series of people with type 1 diabetes and a pancreas kidney or pancreas-only transplant. J Diabetes Investig 2023. [PMID: 37191402 DOI: 10.1111/jdi.14019] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/28/2023] [Accepted: 04/04/2023] [Indexed: 05/17/2023] Open
Abstract
We share our experience of using continuous subcutaneous insulin infusion (CSII) therapy and diabetes technology in six people (5 men) with type 1 diabetes (mean duration 36 years), who developed hyperglycemia post-simultaneous kidney/pancreas (n = 5) or pancreas only (n = 1) transplant. All were on immunosuppression and multiple daily injections of insulin prior to CSII. Four people were started on automated insulin delivery, and two people on CSII and intermittently scanned continuous glucose monitoring. With diabetes technology, the median time in range glucose improved from 37% (24-49%) to 56.6% (48-62%), and similarly, glycated hemoglobin fell from 72.7 mmol/mol (72-79 mmol/mol) to 64 mmol/mol (42-67 mmol/mol; P < 0.05 for both) with no concomitant increase in hypoglycemia. Use of diabetes technology improved glycemic parameters in people with type 1 diabetes with failing pancreatic graft function. Early use of such technology should be considered to improve diabetes control in this complex cohort.
Collapse
Affiliation(s)
- Dimitra Stathi
- Department of Diabetes and Endocrinology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Thomas Johnston
- Department of Diabetes and Endocrinology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Rebecca Hyslop
- Department of Diabetes and Endocrinology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Anna Brackenridge
- Department of Diabetes and Endocrinology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Janaka Karalliedde
- Department of Diabetes and Endocrinology, Guy's and St Thomas' NHS Foundation Trust, London, UK
- School of Cardiovascular and Metabolic Medicine and Sciences, King's College London, London, UK
| |
Collapse
|
9
|
Ballou C, Barton F, Payne EH, Berney T, Villard J, Meier RPH, Baidal D, Alejandro R, Robien M, Eggerman TL, Kamoun M, Muller YD. Matching for HLA-DR excluding diabetogenic HLA-DR3 and HLA-DR4 predicts insulin independence after pancreatic islet transplantation. Front Immunol 2023; 14:1110544. [PMID: 37026004 PMCID: PMC10070978 DOI: 10.3389/fimmu.2023.1110544] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 02/16/2023] [Indexed: 04/08/2023] Open
Abstract
Introduction In pancreatic islet transplantation, the exact contribution of human leukocyte antigen (HLA) matching to graft survival remains unclear. Islets may be exposed to allogenic rejection but also the recurrence of type 1 diabetes (T1D). We evaluated the HLA-DR matching, including the impact of diabetogenic HLA-DR3 or HLA-DR4 matches. Methods We retrospectively examined the HLA profile in 965 transplant recipients and 2327 islet donors. The study population was obtained from patients enrolled in the Collaborative Islet Transplant Registry. We then identified 87 recipients who received a single-islet infusion. Islet-kidney recipients, 2nd islet infusion, and patients with missing data were excluded from the analysis (n=878). Results HLA-DR3 and HLA-DR4 were present in 29.7% and 32.6% of T1D recipients and 11.6% and 15.8% of the donors, respectively. We identified 52 T1D islet recipients mismatched for HLA-DR (group A), 11 with 1 or 2 HLA-DR-matches but excluding HLA-DR3 and HLA- DR4 (group B), and 24 matched for HLA-DR3 or HLA-DR4 (group C). Insulin-independence was maintained in a significantly higher percentage of group B recipients from year one through five post-transplantation (p<0.01). At five-year post-transplantation, 78% of group B was insulin-independent compared to 24% (group A) and 35% (group C). Insulin-independence correlated with significantly better glycemic control (HbA1c <7%), fasting blood glucose, and reduced severe hypoglycemic events. Matching HLA-A-B-DR (≥3) independently of HLA- DR3 or HLA-DR4 matching did not improve graft survival. Conclusion This study suggests that matching HLA-DR but excluding the diabetogenic HLA-DR3 and/or 4 is a significant predictor for long-term islet survival.
Collapse
Affiliation(s)
- Cassandra Ballou
- Collaborative Islet Transplant Registry Coordinating Center, The EMMES Company, LLC, Rockville, MD, United States
- *Correspondence: Yannick D. Muller, ; Cassandra Ballou,
| | - Franca Barton
- Collaborative Islet Transplant Registry Coordinating Center, The EMMES Company, LLC, Rockville, MD, United States
| | - Elizabeth H. Payne
- Collaborative Islet Transplant Registry Coordinating Center, The EMMES Company, LLC, Rockville, MD, United States
| | - Thierry Berney
- Division of Transplantation, Department of Surgery, Geneva University Hospitals, Geneva, Switzerland
| | - Jean Villard
- Department of Genetic, Laboratory and Pathology Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Raphael P. H. Meier
- Department of Surgery, School of Medicine, University of Maryland, Baltimore, MD, United States
| | - David Baidal
- Department of Medicine and the Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Rodolfo Alejandro
- Department of Medicine and the Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Mark Robien
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Thomas L. Eggerman
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Malek Kamoun
- Immunology and Histocompatibility Testing Laboratory, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Yannick D. Muller
- Division of Immunology and Allergy, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
- *Correspondence: Yannick D. Muller, ; Cassandra Ballou,
| |
Collapse
|
10
|
Lasting Over Time: 20-Year Follow-Up of Living Related Simultaneous Pancreas-Kidney Transplant Between Identical Twins. Transplant Proc 2022; 54:2344-2346. [PMID: 36163084 DOI: 10.1016/j.transproceed.2022.08.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/25/2022] [Accepted: 08/29/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND Twenty-year follow-up of the first reported simultaneous pancreas-kidney transplant from living donor between identical twins. CASE We present a case of a patient after the world's first simultaneous pancreas and kidney transplant between identical twins. The transplant was performed because of diabetes mellitus type (DM1) related renal failure. Now in her sixties, the patient has had exceptional prolonged bi-graft function without immunosuppression. She is free from DM1 recurrence, does not take insulin, and her creatinine level has stayed with the normal limit. CONCLUSIONS Simultaneous pancreas and kidney transplant can be performed successfully with excellent long-term outcomes.
Collapse
|
11
|
Nakamura T, Shirouzu T. Antibody-Mediated Rejection and Recurrent Primary Disease: Two Main Obstacles in Abdominal Kidney, Liver, and Pancreas Transplants. J Clin Med 2021; 10:5417. [PMID: 34830699 PMCID: PMC8619797 DOI: 10.3390/jcm10225417] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 02/08/2023] Open
Abstract
The advances in acute phase care have firmly established the practice of organ transplantation in the last several decades. Then, the next issues that loom large in the field of transplantation include antibody-mediated rejection (ABMR) and recurrent primary disease. Acute ABMR is a daunting hurdle in the performance of organ transplantation. The recent progress in desensitization and preoperative monitoring of donor-specific antibodies enables us to increase positive outcomes. However, chronic active ABMR is one of the most significant problems we currently face. On the other hand, recurrent primary disease is problematic for many recipients. Notably, some recipients, unfortunately, lost their vital organs due to this recurrence. Although some progress has been achieved in these two areas, many other factors remain largely obscure. In this review, these two topics will be discussed in light of recent discoveries.
Collapse
Affiliation(s)
- Tsukasa Nakamura
- Department of Organ Transplantation and General Surgery, Kyoto Prefectural University of Medicine, Kajii-cho 465, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Takayuki Shirouzu
- Molecular Diagnositcs Division, Wakunaga Pharmaceutical Co., Ltd., 13-4 Arakicho, shinjyuku-ku, Tokyo 160-0007, Japan;
| |
Collapse
|
12
|
Samojlik MM, Stabler CL. Designing biomaterials for the modulation of allogeneic and autoimmune responses to cellular implants in Type 1 Diabetes. Acta Biomater 2021; 133:87-101. [PMID: 34102338 PMCID: PMC9148663 DOI: 10.1016/j.actbio.2021.05.039] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 05/05/2021] [Accepted: 05/20/2021] [Indexed: 12/15/2022]
Abstract
The effective suppression of adaptive immune responses is essential for the success of allogeneic cell therapies. In islet transplantation for Type 1 Diabetes, pre-existing autoimmunity provides an additional hurdle, as memory autoimmune T cells mediate both an autoantigen-specific attack on the donor beta cells and an alloantigen-specific attack on the donor graft cells. Immunosuppressive agents used for islet transplantation are generally successful in suppressing alloimmune responses, but dramatically hinder the widespread adoption of this therapeutic approach and fail to control memory T cell populations, which leaves the graft vulnerable to destruction. In this review, we highlight the capacity of biomaterials to provide local and nuanced instruction to suppress or alter immune pathways activated in response to an allogeneic islet transplant. Biomaterial immunoisolation is a common approach employed to block direct antigen recognition and downstream cell-mediated graft destruction; however, immunoisolation alone still permits shed donor antigens to escape into the host environment, resulting in indirect antigen recognition, immune cell activation, and the creation of a toxic graft site. Designing materials to decrease antigen escape, improve cell viability, and increase material compatibility are all approaches that can decrease the local release of antigen and danger signals into the implant microenvironment. Implant materials can be further enhanced through the local delivery of anti-inflammatory, suppressive, chemotactic, and/or tolerogenic agents, which serve to control both the innate and adaptive immune responses to the implant with a benefit of reduced systemic effects. Lessons learned from understanding how to manipulate allogeneic and autogenic immune responses to pancreatic islets can also be applied to other cell therapies to improve their efficacy and duration. STATEMENT OF SIGNIFICANCE: This review explores key immunologic concepts and critical pathways mediating graft rejection in Type 1 Diabetes, which can instruct the future purposeful design of immunomodulatory biomaterials for cell therapy. A summary of immunological pathways initiated following cellular implantation, as well as current systemic immunomodulatory agents used, is provided. We then outline the potential of biomaterials to modulate these responses. The capacity of polymeric encapsulation to block some powerful rejection pathways is covered. We also highlight the role of cellular health and biocompatibility in mitigating immune responses. Finally, we review the use of bioactive materials to proactively modulate local immune responses, focusing on key concepts of anti-inflammatory, suppressive, and tolerogenic agents.
Collapse
Affiliation(s)
- Magdalena M Samojlik
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Cherie L Stabler
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA; University of Florida Diabetes Institute, Gainesville, FL, USA; Graduate Program in Biomedical Sciences, College of Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
13
|
Boggi U, Vistoli F, Andres A, Arbogast HP, Badet L, Baronti W, Bartlett ST, Benedetti E, Branchereau J, Burke GW, Buron F, Caldara R, Cardillo M, Casanova D, Cipriani F, Cooper M, Cupisti A, Davide J, Drachenberg C, de Koning EJP, Ettorre GM, Fernandez Cruz L, Fridell JA, Friend PJ, Furian L, Gaber OA, Gruessner AC, Gruessner RW, Gunton JE, Han D, Iacopi S, Kauffmann EF, Kaufman D, Kenmochi T, Khambalia HA, Lai Q, Langer RM, Maffi P, Marselli L, Menichetti F, Miccoli M, Mittal S, Morelon E, Napoli N, Neri F, Oberholzer J, Odorico JS, Öllinger R, Oniscu G, Orlando G, Ortenzi M, Perosa M, Perrone VG, Pleass H, Redfield RR, Ricci C, Rigotti P, Paul Robertson R, Ross LF, Rossi M, Saudek F, Scalea JR, Schenker P, Secchi A, Socci C, Sousa Silva D, Squifflet JP, Stock PG, Stratta RJ, Terrenzio C, Uva P, Watson CJ, White SA, Marchetti P, Kandaswamy R, Berney T. First World Consensus Conference on pancreas transplantation: Part II - recommendations. Am J Transplant 2021; 21 Suppl 3:17-59. [PMID: 34245223 PMCID: PMC8518376 DOI: 10.1111/ajt.16750] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 06/25/2021] [Accepted: 06/26/2021] [Indexed: 02/07/2023]
Abstract
The First World Consensus Conference on Pancreas Transplantation provided 49 jury deliberations regarding the impact of pancreas transplantation on the treatment of diabetic patients, and 110 experts' recommendations for the practice of pancreas transplantation. The main message from this consensus conference is that both simultaneous pancreas-kidney transplantation (SPK) and pancreas transplantation alone can improve long-term patient survival, and all types of pancreas transplantation dramatically improve the quality of life of recipients. Pancreas transplantation may also improve the course of chronic complications of diabetes, depending on their severity. Therefore, the advantages of pancreas transplantation appear to clearly surpass potential disadvantages. Pancreas after kidney transplantation increases the risk of mortality only in the early period after transplantation, but is associated with improved life expectancy thereafter. Additionally, preemptive SPK, when compared to SPK performed in patients undergoing dialysis, appears to be associated with improved outcomes. Time on dialysis has negative prognostic implications in SPK recipients. Increased long-term survival, improvement in the course of diabetic complications, and amelioration of quality of life justify preferential allocation of kidney grafts to SPK recipients. Audience discussions and live voting are available online at the following URL address: http://mediaeventi.unipi.it/category/1st-world-consensus-conference-of-pancreas-transplantation/246.
Collapse
|
14
|
Korpos É, Kadri N, Loismann S, Findeisen CR, Arfuso F, Burke GW, Richardson SJ, Morgan NG, Bogdani M, Pugliese A, Sorokin L. Identification and characterisation of tertiary lymphoid organs in human type 1 diabetes. Diabetologia 2021; 64:1626-1641. [PMID: 33912981 PMCID: PMC8187221 DOI: 10.1007/s00125-021-05453-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 01/28/2021] [Indexed: 12/13/2022]
Abstract
AIMS/HYPOTHESIS We and others previously reported the presence of tertiary lymphoid organs (TLOs) in the pancreas of NOD mice, where they play a role in the development of type 1 diabetes. Our aims here are to investigate whether TLOs are present in the pancreas of individuals with type 1 diabetes and to characterise their distinctive features, in comparison with TLOs present in NOD mouse pancreases, in order to interpret their functional significance. METHODS Using immunofluorescence confocal microscopy, we examined the extracellular matrix (ECM) and cellular constituents of pancreatic TLOs from individuals with ongoing islet autoimmunity in three distinct clinical settings of type 1 diabetes: at risk of diabetes; at/after diagnosis; and in the transplanted pancreas with recurrent diabetes. Comparisons were made with TLOs from 14-week-old NOD mice, which contain islets exhibiting mild to heavy leucocyte infiltration. We determined the frequency of the TLOs in human type 1diabetes with insulitis and investigated the presence of TLOs in relation to age of onset, disease duration and disease severity. RESULTS TLOs were identified in preclinical and clinical settings of human type 1 diabetes. The main characteristics of these TLOs, including the cellular and ECM composition of reticular fibres (RFs), the presence of high endothelial venules and immune cell subtypes detected, were similar to those observed for TLOs from NOD mouse pancreases. Among 21 donors with clinical type 1 diabetes who exhibited insulitis, 12 had TLOs and had developed disease at younger age compared with those lacking TLOs. Compartmentalised TLOs with distinct T cell and B cell zones were detected in donors with short disease duration. Overall, TLOs were mainly associated with insulin-containing islets and their frequency decreased with increasing severity of beta cell loss. Parallel studies in NOD mice further revealed some differences in so far as regulatory T cells were essentially absent from human pancreatic TLOs and CCL21 was not associated with RFs. CONCLUSIONS/INTERPRETATION We demonstrate a novel feature of pancreas pathology in type 1 diabetes. TLOs represent a potential site of autoreactive effector T cell generation in islet autoimmunity and our data from mouse and human tissues suggest that they disappear once the destructive process has run its course. Thus, TLOs may be important for type 1 diabetes progression.
Collapse
Affiliation(s)
- Éva Korpos
- Institute of Physiological Chemistry and Pathobiochemistry, University of Muenster, Muenster, Germany.
- Cells-in-Motion Interfaculty Centre, University of Muenster, Muenster, Germany.
| | - Nadir Kadri
- Science for Life Laboratory, Department of Medicine, Karolinska Institute, Karolinska University Hospital, Solna, Stockholm, Sweden
| | - Sophie Loismann
- Institute of Physiological Chemistry and Pathobiochemistry, University of Muenster, Muenster, Germany
- Cells-in-Motion Interfaculty Centre, University of Muenster, Muenster, Germany
| | - Clais R Findeisen
- Institute of Physiological Chemistry and Pathobiochemistry, University of Muenster, Muenster, Germany
- Cells-in-Motion Interfaculty Centre, University of Muenster, Muenster, Germany
| | - Frank Arfuso
- Institute of Physiological Chemistry and Pathobiochemistry, University of Muenster, Muenster, Germany
- Cells-in-Motion Interfaculty Centre, University of Muenster, Muenster, Germany
| | - George W Burke
- Department of Surgery, Division of Transplantation, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Sarah J Richardson
- Institute of Biomedical & Clinical Science, University of Exeter, Exeter, UK
| | - Noel G Morgan
- Institute of Biomedical & Clinical Science, University of Exeter, Exeter, UK
| | - Marika Bogdani
- Matrix Biology Program, Benaroya Research Institute, Seattle, WA, USA
| | - Alberto Pugliese
- Diabetes Research Institute, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
- Division of Endocrinology and Metabolism, Department of Medicine, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
- Department of Microbiology and Immunology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Lydia Sorokin
- Institute of Physiological Chemistry and Pathobiochemistry, University of Muenster, Muenster, Germany
- Cells-in-Motion Interfaculty Centre, University of Muenster, Muenster, Germany
| |
Collapse
|
15
|
Anteby R, Lucander A, Bachul PJ, Pyda J, Grybowski D, Basto L, Generette GS, Perea L, Golab K, Wang LJ, Tibudan M, Thomas C, Fung J, Witkowski P. Evaluating the Prognostic Value of Islet Autoantibody Monitoring in Islet Transplant Recipients with Long-Standing Type 1 Diabetes Mellitus. J Clin Med 2021; 10:jcm10122708. [PMID: 34205321 PMCID: PMC8233942 DOI: 10.3390/jcm10122708] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/15/2021] [Accepted: 06/17/2021] [Indexed: 12/03/2022] Open
Abstract
(1) Background: The correlation between titers of islet autoantibodies (IAbs) and the loss of transplanted islets remains controversial. We sought to evaluate the prognostic utility of monitoring IAbs in diabetic patients after islet transplantation (ITx); (2) Methods: Twelve patients with Type 1 diabetes mellitus and severe hypoglycemia underwent ITx. Serum concentration of glutamic acid decarboxylase (GAD), insulinoma antigen 2 (IA-2), and zinc transport 8 (ZnT8) autoantibodies was assessed before ITx and 0, 7, and 75 days and every 3 months post-operatively; (3) Results: IA-2A (IA-2 antibody) and ZnT8A (ZnT8 antibody) levels were not detectable before or after ITx in all patients (median follow-up of 53 months (range 24–61)). Prior to ITx, GAD antibody (GADA) was undetectable in 67% (8/12) of patients. Of those, 75% (6/8) converted to GADA+ after ITx. In 67% (4/6) of patients with GADA+ seroconversion, GADA level peaked within 3 months after ITx and subsequently declined. All patients with GADA+ seroconversion maintained long-term partial or complete islet function (insulin independence) after 1 or 2 ITx. There was no correlation between the presence of IAb-associated HLA haplotypes and the presence of IAbs before or after ITx; (4) Conclusions: There is no association between serum GADA trends and ITx outcomes. IA-2A and ZnT8A were not detectable in any of our patients before or after ITx.
Collapse
Affiliation(s)
- Roi Anteby
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA; (R.A.); (A.L.); (P.J.B.); (D.G.); (L.B.); (G.S.G.); (L.P.); (K.G.); (L.-j.W.); (M.T.); (J.F.)
- Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Harvard School of Public Health, Boston, MA 02115, USA
| | - Aaron Lucander
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA; (R.A.); (A.L.); (P.J.B.); (D.G.); (L.B.); (G.S.G.); (L.P.); (K.G.); (L.-j.W.); (M.T.); (J.F.)
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Piotr J. Bachul
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA; (R.A.); (A.L.); (P.J.B.); (D.G.); (L.B.); (G.S.G.); (L.P.); (K.G.); (L.-j.W.); (M.T.); (J.F.)
| | - Jordan Pyda
- Department of Surgery, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA;
| | - Damian Grybowski
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA; (R.A.); (A.L.); (P.J.B.); (D.G.); (L.B.); (G.S.G.); (L.P.); (K.G.); (L.-j.W.); (M.T.); (J.F.)
| | - Lindsay Basto
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA; (R.A.); (A.L.); (P.J.B.); (D.G.); (L.B.); (G.S.G.); (L.P.); (K.G.); (L.-j.W.); (M.T.); (J.F.)
| | - Gabriela S. Generette
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA; (R.A.); (A.L.); (P.J.B.); (D.G.); (L.B.); (G.S.G.); (L.P.); (K.G.); (L.-j.W.); (M.T.); (J.F.)
| | - Laurencia Perea
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA; (R.A.); (A.L.); (P.J.B.); (D.G.); (L.B.); (G.S.G.); (L.P.); (K.G.); (L.-j.W.); (M.T.); (J.F.)
| | - Karolina Golab
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA; (R.A.); (A.L.); (P.J.B.); (D.G.); (L.B.); (G.S.G.); (L.P.); (K.G.); (L.-j.W.); (M.T.); (J.F.)
| | - Ling-jia Wang
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA; (R.A.); (A.L.); (P.J.B.); (D.G.); (L.B.); (G.S.G.); (L.P.); (K.G.); (L.-j.W.); (M.T.); (J.F.)
| | - Martin Tibudan
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA; (R.A.); (A.L.); (P.J.B.); (D.G.); (L.B.); (G.S.G.); (L.P.); (K.G.); (L.-j.W.); (M.T.); (J.F.)
| | - Celeste Thomas
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA;
| | - John Fung
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA; (R.A.); (A.L.); (P.J.B.); (D.G.); (L.B.); (G.S.G.); (L.P.); (K.G.); (L.-j.W.); (M.T.); (J.F.)
| | - Piotr Witkowski
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA; (R.A.); (A.L.); (P.J.B.); (D.G.); (L.B.); (G.S.G.); (L.P.); (K.G.); (L.-j.W.); (M.T.); (J.F.)
- Correspondence: ; Tel.: +1-773-702-2447
| |
Collapse
|
16
|
Buron F, Reffet S, Badet L, Morelon E, Thaunat O. Immunological Monitoring in Beta Cell Replacement: Towards a Pathophysiology-Guided Implementation of Biomarkers. Curr Diab Rep 2021; 21:19. [PMID: 33895937 DOI: 10.1007/s11892-021-01386-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/25/2021] [Indexed: 01/23/2023]
Abstract
PURPOSE OF REVIEW Grafted beta cells are lost because of recurrence of T1D and/or allograft rejection, two conditions diagnosed with pancreas graft biopsy, which is invasive and impossible in case of islet transplantation. This review synthetizes the current pathophysiological knowledge and discusses the interest of available immune biomarkers. RECENT FINDINGS Despite the central role of auto-(recurrence of T1D) and allo-(T-cell mediated rejection) immune cellular responses, the latter are not directly monitored in routine. In striking contrast, there have been undisputable progresses in monitoring of auto and alloantibodies. Except for pancreas recipients in whom anti-donor HLA antibodies can be directly responsible for antibody-mediated rejection, autoantibodies (and alloantibodies in islet recipients) have no direct pathogenic effect. However, their fluctuation offers a surrogate marker for the activation status of T cells (because antibody generation depends on T cells). This illustrates the necessity to understand the pathophysiology when interpreting a biomarker and selecting the appropriate treatment.
Collapse
Affiliation(s)
- Fanny Buron
- Department of Transplantation, Nephrology and Clinical Immunology, Edouard Herriot Hospital, Hospices Civils de Lyon, 5 Place d'Arsonval, 69003, Lyon, France
| | - Sophie Reffet
- Department of Endocrinology and Diabetes, Lyon-Sud Hospital, Hospices Civils de Lyon, 69310, Pierre-Bénite, France
| | - Lionel Badet
- Department of Urology and Transplantation surgery, Edouard Herriot Hospital, Hospices Civils de Lyon, Lyon, France
| | - Emmanuel Morelon
- Department of Transplantation, Nephrology and Clinical Immunology, Edouard Herriot Hospital, Hospices Civils de Lyon, 5 Place d'Arsonval, 69003, Lyon, France
- French National Institute of Health and Medical Research (Inserm) Unit 1111, Lyon, France
- Lyon-Est Medical Faculty, Claude Bernard University (Lyon 1), Lyon, France
| | - Olivier Thaunat
- Department of Transplantation, Nephrology and Clinical Immunology, Edouard Herriot Hospital, Hospices Civils de Lyon, 5 Place d'Arsonval, 69003, Lyon, France.
- French National Institute of Health and Medical Research (Inserm) Unit 1111, Lyon, France.
- Lyon-Est Medical Faculty, Claude Bernard University (Lyon 1), Lyon, France.
- Service de Transplantation, Néphrologie et Immunologie Clinique, Hôpital Edouard Herriot, 5 Place d'Arsonval, 69003, Lyon, France.
| |
Collapse
|
17
|
Masset C, Branchereau J, Karam G, Hourmant M, Dantal J, Giral M, Garandeau C, Meurette A, Kerleau C, Kervella D, Ville S, Blancho G, Cantarovich D. Clinical utility of C-peptide measurement after pancreas transplantation with especial focus on early graft thrombosis. Transpl Int 2021; 34:942-953. [PMID: 33733553 DOI: 10.1111/tri.13866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 03/09/2021] [Accepted: 03/13/2021] [Indexed: 11/26/2022]
Abstract
Since the beginning of our pancreas transplant programme, plasma C-peptide was routinely measured daily during the postoperative period. We aimed to evaluate the clinical interest of the C-peptide in the follow-up of pancreas transplantation with a particular look on early graft failure. From 2000 to 2016, 384 pancreas transplantations were evaluated. We collected and compared C-peptide, glycaemia and adjusted C-peptide (aCP; calculated based on C-peptide, glycaemia and creatininaemia) in patients with and without pancreas failure within 30 days after surgery. Variations of glycaemia, C-peptide and aCP between the day before and the day of failure were also recorded. The difference of aCP was significant during the first week after transplantation between patients with thrombosis and those with functional allograft: 63.2 vs. 26.7 on day 1, P = 0.0003; 61.4 vs. 26.7 on day 3, P < 0.0001; 64.8 vs. 5.7 on day 7, P < 0.0001, respectively. Glycaemia had a median increase of 8% on the day of failure, whereas C-peptide and aCP had, respectively, a median decrease of 88% and 83%. C-peptide monitoring after pancreas transplantation may help to identify graft function and early failure. This sensitive biomarker could allow pre-emptive diagnosis of an early thrombotic event allowing the possibility of rescue interventions.
Collapse
Affiliation(s)
- Christophe Masset
- Institut de Transplantation-Urologie-Néphrologie (ITUN), Nantes University Hospital, Nantes, France
| | - Julien Branchereau
- Institut de Transplantation-Urologie-Néphrologie (ITUN), Nantes University Hospital, Nantes, France
| | - Georges Karam
- Institut de Transplantation-Urologie-Néphrologie (ITUN), Nantes University Hospital, Nantes, France
| | - Maryvonne Hourmant
- Institut de Transplantation-Urologie-Néphrologie (ITUN), Nantes University Hospital, Nantes, France
| | - Jacques Dantal
- Institut de Transplantation-Urologie-Néphrologie (ITUN), Nantes University Hospital, Nantes, France
| | - Magali Giral
- Institut de Transplantation-Urologie-Néphrologie (ITUN), Nantes University Hospital, Nantes, France
| | - Claire Garandeau
- Institut de Transplantation-Urologie-Néphrologie (ITUN), Nantes University Hospital, Nantes, France
| | - Aurélie Meurette
- Institut de Transplantation-Urologie-Néphrologie (ITUN), Nantes University Hospital, Nantes, France
| | - Clarisse Kerleau
- Institut de Transplantation-Urologie-Néphrologie (ITUN), Nantes University Hospital, Nantes, France
| | - Delphine Kervella
- Institut de Transplantation-Urologie-Néphrologie (ITUN), Nantes University Hospital, Nantes, France
| | - Simon Ville
- Institut de Transplantation-Urologie-Néphrologie (ITUN), Nantes University Hospital, Nantes, France
| | - Gilles Blancho
- Institut de Transplantation-Urologie-Néphrologie (ITUN), Nantes University Hospital, Nantes, France
| | - Diego Cantarovich
- Institut de Transplantation-Urologie-Néphrologie (ITUN), Nantes University Hospital, Nantes, France
| |
Collapse
|
18
|
Pancreatic Islet Changes in Human Whole Organ Pancreas Explants: What Can Be Learned From Explanted Samples? Transplant Direct 2020; 6:e613. [PMID: 33134489 PMCID: PMC7575169 DOI: 10.1097/txd.0000000000001059] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 06/24/2020] [Accepted: 07/14/2020] [Indexed: 01/09/2023] Open
Abstract
Background. Whole pancreas transplantation (Tx) is a successful treatment for type 1 diabetes resulting in independence from antidiabetic therapies. Transplant-related factors contributing to pancreatic islet failure are largely unknown; both recurring insulitis and pancreatitis have been implicated. The aim was to determine if cellular changes in islets and exocrine tissue are evident early in Tx, which could contribute to eventual graft failure using well-preserved tissue of grafts explanted from largely normoglycemic recipients. Methods. Histological specimens of explants (n = 31), Tx duration 1 day–8 years (median 29 d), cold ischemia time 7.2–17.3 hours (median 11.1 h), donor age 13–54 years (median 38 y) were examined; sections were labeled for inflammation, islet amyloidosis, and tissue fibrosis, and morphometry performed on immunolabeled insulin and glucagon positive islet cells. Data were related to clinical details of donor, recipient, and features of Tx. Results. Islet inflammation consistent with recurrent insulitis was not seen in any sample. Insulin-labeled islet cell proportion decreased with donor age (P < 0.05) and cold ischemia (P < 0.01) in explants from 26 normoglycemic patients; glucagon-labeled area proportion increased with cold ischemia (P < 0.05). Clinical pancreatitis was the explant reason in 12 of 28 normoglycemic cases. Exocrine fibrotic area/pancreas was variable (0.7%–55%) and unrelated to clinical/pathological features. Islet amyloid was present in 3 normoglycemic cases (donor ages 58, 42, and 31 y; Tx duration 8 y, 31 and 33 d, respectively). In 1 patient receiving antidiabetic therapy, the insulin-labeled area was reduced but with no evidence of islet inflammation. Conclusions. Explant histological changes after short-term Tx are similar to those seen in type 2 diabetes and occur in the absence of immunologic rejection without causing hyperglycemia. This suggests that factors associated with Tx affect islet stability; persistent deterioration of islet integrity and exocrine tissue fibrosis could impact on sustainability of islet function.
Collapse
|
19
|
El-Kersh AOFO, El-Akabawy G, Al-Serwi RH. Transplantation of human dental pulp stem cells in streptozotocin-induced diabetic rats. Anat Sci Int 2020; 95:523-539. [PMID: 32476103 DOI: 10.1007/s12565-020-00550-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 05/20/2020] [Indexed: 12/11/2022]
Abstract
Type 1 diabetes mellitus (T1DM) is a chronic metabolic disease caused by the destruction of pancreatic β-cells. Human dental pulp stem cells represent a promising source for cell-based therapies, owing to their easy, minimally invasive surgical access, and high proliferative capacity. It was reported that human dental pulp stem cells can differentiate into a pancreatic cell lineage in vitro; however, few studies have investigated their effects on diabetes. Our study aimed to investigate the therapeutic potential of intravenous and intrapancreatic transplantation of human dental pulp stem cells in a rat model of streptozotocin-induced type 1 diabetes. Forty Sprague Dawley male rats were randomly categorized into four groups: control, diabetic (STZ), intravenous treatment group (IV), and intrapancreatic treatment group (IP). Human dental pulp stem cells (1 × 106 cells) or vehicle were injected into the pancreas or tail vein 7 days after streptozotocin injection. Fasting blood glucose levels were monitored weekly. Glucose tolerance test, rat and human serum insulin and C-peptide, pancreas histology, and caspase-3, vascular endothelial growth factor, and Ki67 expression in pancreatic tissues were assessed 28 days post-transplantation. We found that both IV and IP transplantation of human dental pulp stem cells reduced blood glucose and increased levels of rat and human serum insulin and C-peptide. The cells engrafted and survived in the streptozotocin-injured pancreas. Islet-like clusters and scattered human dental pulp stem cells expressing insulin were observed in the pancreas of diabetic rats with some difference in the distribution pattern between the two injection routes. RT-PCR analyses revealed the expression of the human-specific pancreatic β-cell genes neurogenin 3 (NGN3), paired box 4 (PAX4), glucose transporter 2 (GLUT2), and insulin in the pancreatic tissues of both the IP and IV groups. In addition, the transplanted cells downregulated the expression of caspase-3 and upregulated the expression of vascular endothelial growth factor and Ki67, suggesting that the injected cells exerted pro-angiogenetic and antiapoptotic effects, and promoted endogenous β-cell replication. Our study is the first to show that human dental pulp stem cells can migrate and survive within streptozotocin-injured pancreas, and induce antidiabetic effects through the differentiation and replacement of lost β-cells and paracrine-mediated pancreatic regeneration. Thus, human dental pulp stem cells may have therapeutic potential to treat patients with long term T1DM.
Collapse
Affiliation(s)
| | - Gehan El-Akabawy
- Department of Basic Sciences, College of Medicine, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia. .,Department of Anatomy and Embryology, Faculty of Medicine, Menoufia University, Menoufia, Egypt.
| | - Rasha H Al-Serwi
- Basic Dental Sciences, College of Dentistry, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia.,Oral Biology Department, Faculty of Dentistry, Mansoura University, Mansoura, Egypt
| |
Collapse
|
20
|
Pestana N, Malheiro J, Silva F, Silva A, Ribeiro C, Pedroso S, Almeida M, Dias L, Henriques AC, Martins LS. Impact of Pancreatic Autoantibodies in Pancreas Graft Survival After Pancreas-Kidney Transplantation. Transplant Proc 2020; 52:1370-1375. [PMID: 32245621 DOI: 10.1016/j.transproceed.2020.02.035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 02/07/2020] [Indexed: 01/09/2023]
Abstract
BACKGROUND In simultaneous pancreas-kidney transplantation (SPKT), persistence or recurrence of pancreatic autoantibodies (PAs) has been associated with pancreas graft (PG) autoimmune-driven injury. Our aim was to analyze the impact of PAs on PG survival. METHODS Between January 1, 2000, and December 31, 2017, we studied 139 patients with post-SPKT anti-glutamic acid decarboxylase (GAD) autoantibody. Alloimmune (ALI) events were defined as PG rejection and/or de novo donor-specific antibodies (DSA). Hence, 3 groups were defined: patients without ALI events or anti-GAD (n = 42), those with ALI events (n = 14), or those only with autoimmune events (positive for anti-GAD and no ALI events; n = 83). RESULTS Male sex was predominant (n = 72, 52%). Median age was 35 years (interquartile range: 31-39) and median follow-up was 6-7 years (interquartile range: 4.1-9.2). Regarding anti-GAD positivity post-SPKT (n = 90, 65%), no differences were observed concerning age, sex, anti-HLA antibodies, HLA mismatch number and de novo DSA. ALI events were present in 10% (n = 14). PG survival 15 years post-SPKT was better in patients without immune events (96%) followed by those with ALI (69%) and autoimmune events (63%) (P = .025). Anti-GAD was associated to higher annualized mean Hb1AC (P = .006) and lower mean C-peptide (P = .013). According to pre- and post-SPKT anti-GAD status, conversion from negative to positive was associated to worse (63%) 10-year PG survival (P = .044), compared to persistence of negative (100%) or positive anti-GAD (88%). Anti-islet cell and anti-insulin autoantibodies had no impact. CONCLUSION Anti-GAD presence post-SPKT was associated to higher pancreas disfunction and lower PG survival. De novo anti-GAD seems to offer a particular risk of PG failure.
Collapse
Affiliation(s)
- Nicole Pestana
- Nephrology Department, Hospital Central do Funchal, Funchal, Portugal.
| | - Jorge Malheiro
- Nephrology Department, Renal and Pancreatic Transplant Units, Centro Hospitalar Universitário do Porto, Lg Prof Abel Salazar, Portugal
| | - Filipa Silva
- Nephrology Department, Renal and Pancreatic Transplant Units, Centro Hospitalar Universitário do Porto, Lg Prof Abel Salazar, Portugal
| | - Andreia Silva
- Nephrology Department, Centro Hospitalar Tondela-Viseu, Viseu, Portugal
| | - Catarina Ribeiro
- Nephrology Department, Centro Hospitalar Vila Nova de Gaia/Espinho, Vila Nova de Gaia, Porto, Portugal
| | - Sofia Pedroso
- Nephrology Department, Renal and Pancreatic Transplant Units, Centro Hospitalar Universitário do Porto, Lg Prof Abel Salazar, Portugal
| | - Manuela Almeida
- Nephrology Department, Renal and Pancreatic Transplant Units, Centro Hospitalar Universitário do Porto, Lg Prof Abel Salazar, Portugal
| | - Leonídio Dias
- Nephrology Department, Renal and Pancreatic Transplant Units, Centro Hospitalar Universitário do Porto, Lg Prof Abel Salazar, Portugal
| | - António Castro Henriques
- Nephrology Department, Renal and Pancreatic Transplant Units, Centro Hospitalar Universitário do Porto, Lg Prof Abel Salazar, Portugal
| | - La Salete Martins
- Nephrology Department, Renal and Pancreatic Transplant Units, Centro Hospitalar Universitário do Porto, Lg Prof Abel Salazar, Portugal
| |
Collapse
|
21
|
Argente-Pla M, Martínez-Millana A, Del Olmo-García MI, Espí-Reig J, Pérez-Rojas J, Traver-Salcedo V, Merino-Torres JF. Autoimmune Diabetes Recurrence After Pancreas Transplantation: Diagnosis, Management, and Literature Review. Ann Transplant 2019; 24:608-616. [PMID: 31767825 PMCID: PMC6896746 DOI: 10.12659/aot.920106] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Background Pancreas transplantation can be a viable treatment option for patients with type 1 diabetes mellitus (T1DM), especially for those who are candidates for kidney transplantation. T1DM may rarely recur after pancreas transplantation, causing the loss of pancreatic graft. The aim of this study was to describe the prevalence of T1DM recurrence after pancreas transplantation in our series. Material/Methods Eighty-one patients transplanted from 2002 to 2015 were included. Autoantibody testing (GADA and IA-2) was performed before pancreas transplantation and during the follow-up. Results The series includes 48 males and 33 females, mean age 37.4±5.7 years and mean duration of diabetes 25.5±6.5 years. Patients received simultaneous pancreas kidney (SPK) transplantation. After SPK transplantation, 56 patients retained pancreatic graft, 8 patients died, and 17 patients lost their pancreatic graft. T1DM recurrence occurred in 2 of the 81 transplanted patients, yielding a prevalence of 2.5%, with an average time of appearance of 3.3 years after transplant. Pancreatic enzymes were normal in the 2 patients, ruling out pancreatic rejection. T1DM recurrence was confirmed histologically, showing selective lymphoid infiltration of the pancreatic islets. Conclusions T1DM recurrence after pancreas transplantation is infrequent; however, it is one of the causes of pancreatic graft loss that should always be ruled out. Negative autoimmunity prior to transplantation does not ensure that T1DM does not recur.
Collapse
Affiliation(s)
- María Argente-Pla
- Department of Endocrinology and Nutrition, La Fe University and Polytechnic Hospital, Valencia, Spain.,Mixed Research Unit of Endocrinology, Nutrition and Dietetics, La Fe Health Research Institute, Valencia, Spain
| | | | - María Isabel Del Olmo-García
- Department of Endocrinology and Nutrition, La Fe University and Polytechnic Hospital, Valencia, Spain.,Mixed Research Unit of Endocrinology, Nutrition and Dietetics, La Fe Health Research Institute, Valencia, Spain
| | - Jordi Espí-Reig
- Department of Nephrology, La Fe University and Polytechnic Hospital, Valencia, Spain
| | - Judith Pérez-Rojas
- Department of Pathological Anatomy, La Fe University and Polytechnic Hospital, Valencia, Spain
| | | | - Juan Francisco Merino-Torres
- Department of Endocrinology and Nutrition, La Fe University and Polytechnic Hospital, Valencia, Spain.,Mixed Research Unit of Endocrinology, Nutrition and Dietetics, La Fe Health Research Institute, Valencia, Spain.,Department of Medicine, University of Valencia, Valencia, Spain
| |
Collapse
|
22
|
Abstract
PURPOSE OF REVIEW Pancreas retransplantations are rarely carried out, and their outcomes are still debatable because of a lack of studies and clinical series on this issue. RECENT FINDINGS In general, pancreas retransplantations achieve similar or even higher patient survival than primary transplantations; however, it should be noted that this finding may be biased, as only healthier patients are selected for retransplantation. Graft survival in retransplantations is usually lower than that in primary transplantation, but this comparison may also be biased, as most retransplantations are solitary pancreas transplantations (which are known to have lower graft survival), whereas primary transplantations are mostly simultaneous kidney-pancreas transplantations. Technical loss is similar between primary pancreas transplantations and pancreas retransplantations, but the occurrence of surgical complications is greater in the latter. SUMMARY This review summarizes the literature on pancreas retransplantations, comparing them with primary transplantations, and demonstrates that in selected patients in experienced centres, retransplantation can be a valid and effective option for returning the patient to an insulin-free state.
Collapse
|
23
|
Rodelo-Haad C, Agüera ML, Carmona A, Navarro MD, Carracedo J, Rodriguez-Benot A, Aljama P. Pancreatic autoantibodies and CD14+CD16+ monocytes subset are associated with the impairment of ß-cell function after simultaneous pancreas-kidney transplantation. PLoS One 2019; 14:e0212547. [PMID: 30794611 PMCID: PMC6386378 DOI: 10.1371/journal.pone.0212547] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 02/05/2019] [Indexed: 12/18/2022] Open
Abstract
Pancreatic autoantibodies (AAb) has been associated with a worse pancreas graft survival after simultaneous pancreas-kidney transplantation (SPK). However, due to the variable time for AAb to become positive and the lack of early biomarkers suggesting such autoimmune activation, the mechanisms leading ß-cell destruction remain uncertain. The present study aimed to evaluate the association between post-transplant AAb and the functional impairment of the pancreatic ß-cell and also the association of such AAb with inflammation after SPK. In a longitudinal study, we analyzed the impact of post-transplant glutamic acid decarboxylase (GAD-65) and the insulinoma-associated autoantigen 2 (IA-2) AAb on pancreas graft function. Serum Hb1Ac and C-peptide (C-pep) were longitudinally compared between a group with positive posttransplant AAb (AAb+; n = 40) and another matched group with negative AAb (AAb-; n = 40) until the fifth year following seroconversion. In the cross-sectional analysis, we further evaluated the systemic signatures of inflammation by measuring pro-inflammatory CD14+CD16+ monocytes by flow-cytometry and interleukin 17-A serum levels in 38 SPK recipients and ten healthy controls. In the longitudinal study, patients with AAb+ showed higher levels of Hb1Ac (p<0.001) and lower C-pep levels (p<0.001) compared to those who remained AAb- throughout the follow-up. In the cross-sectional study, AAb+ patients showed a higher percentage of CD14+CD16+ monocytes compared with those with AAb- and the healthy controls (6.70±4.19% versus 4.0±1.84% and 3.44±0.93%; p = 0.026 and 0.009 respectively). Also, CD14+CD16+ monocytes correlated with Hb1Ac and C-pep serum levels. Multivariate logistic regression showed that posttransplant AAb+ was independently associated with a higher percentage of pro-inflammatory monocytes (adjusted-OR 1.59, 95%CI 1.05–2.40, p = 0.027). The group of patients with positive AAb also showed higher levels of IL17A as compared with the other groups (either healthy control or the negative AAb subjects). In conclusion, pancreatic AAb+ after SPK were not only associated with higher Hb1Ac and lower c-peptide serum levels but also with an increased percentage of CD14+CD16+ monocytes and higher levels of circulating IL17-A.
Collapse
Affiliation(s)
- Cristian Rodelo-Haad
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC)/Reina Sofia University Hospital/University of Cordoba, Cordoba, Spain
- Nephrology Unit. Reina Sofia University Hospital, Cordoba, Spain
- RETICs Red Renal (Instituto de Salud Carlos III), Madrid, Spain
- * E-mail:
| | - Maria Luisa Agüera
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC)/Reina Sofia University Hospital/University of Cordoba, Cordoba, Spain
- Nephrology Unit. Reina Sofia University Hospital, Cordoba, Spain
- RETICs Red Renal (Instituto de Salud Carlos III), Madrid, Spain
| | - Andres Carmona
- Nephrology Unit. Reina Sofia University Hospital, Cordoba, Spain
- RETICs Red Renal (Instituto de Salud Carlos III), Madrid, Spain
| | - Maria Dolores Navarro
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC)/Reina Sofia University Hospital/University of Cordoba, Cordoba, Spain
- Nephrology Unit. Reina Sofia University Hospital, Cordoba, Spain
- RETICs Red Renal (Instituto de Salud Carlos III), Madrid, Spain
| | - Julia Carracedo
- RETICs Red Renal (Instituto de Salud Carlos III), Madrid, Spain
- Departamento de Genética, Fisiología y Microbiología, Facultad de Biología, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Alberto Rodriguez-Benot
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC)/Reina Sofia University Hospital/University of Cordoba, Cordoba, Spain
- Nephrology Unit. Reina Sofia University Hospital, Cordoba, Spain
- RETICs Red Renal (Instituto de Salud Carlos III), Madrid, Spain
| | - Pedro Aljama
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC)/Reina Sofia University Hospital/University of Cordoba, Cordoba, Spain
- Nephrology Unit. Reina Sofia University Hospital, Cordoba, Spain
- RETICs Red Renal (Instituto de Salud Carlos III), Madrid, Spain
| |
Collapse
|
24
|
Sun Y, Mao Q, Shen C, Wang C, Jia W. Exosomes from β-cells alleviated hyperglycemia and enhanced angiogenesis in islets of streptozotocin-induced diabetic mice. Diabetes Metab Syndr Obes 2019; 12:2053-2064. [PMID: 31632115 PMCID: PMC6790122 DOI: 10.2147/dmso.s213400] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 08/29/2019] [Indexed: 12/20/2022] Open
Abstract
PURPOSE Exosomes are small nanoscale vesicles secreted from cells. Exosome-based therapeutic approaches have been evaluated in treating ischemic diseases. In the present study, we explored the effect of exosomes on streptozotozin (STZ)-induced diabetic mouse and its underlying mechanisms. METHODS Exosomes were isolated from MIN6 cells. Transmission electron microscopy, dynamic light scattering and Western blot were used to identify the exosomes. STZ was used to establish diabetic or abnormal glucose tolerance mouse model. Histology study and flow cytometry were applied to detect the changes in immune responses. RESULTS Transplantation of the exosomes into diabetic mice resulted in a longer median survival time compared with the untreated diabetic mice (P<0.01). Transplantation of the exosomes improved glucose tolerance, increased insulin content and preserved the architectures of islets in mice with abnormal glucose tolerance. Moreover, exosome treatment enhanced the expression of CD31, a marker of endothelial cells, and tended to reduce macrophage infiltration in islets of STZ-treated mice. CONCLUSION Exosomes derived from β-cells play a role in preserving pancreatic islet architecture and its function, and in inducing islet angiogenesis, which implicates that exosome treatment could be a novel therapeutic strategy for diabetes.
Collapse
Affiliation(s)
- Yun Sun
- Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai200233, People’s Republic of China
| | - Qianyun Mao
- Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai200233, People’s Republic of China
| | - Chao Shen
- Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai200233, People’s Republic of China
| | - Chen Wang
- Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai200233, People’s Republic of China
- Correspondence: Chen Wang Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Yishan Road, Shanghai200233, People’s Republic of ChinaTel +86 212 405 8657Fax +86 216 436 8031 Email
| | - Weiping Jia
- Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Diabetes Institute, Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai200233, People’s Republic of China
| |
Collapse
|
25
|
Burke GW, Ciancio G, Morsi M, Figueiro J, Chen L, Vendrame F, Pugliese A. Type 1 Diabetes Recurrence After Simultaneous Pancreas-Kidney
Transplantation. CURRENT TRANSPLANTATION REPORTS 2018. [DOI: 10.1007/s40472-018-0210-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
26
|
Kervella D, Scharbarg E, Mahot-Moreau P, Renaudin K, Branchereau J, Karam G, Chaillous L, Bach K, Cantarovich D. Remission of type 1 diabetes mellitus recurrence 6 years after simultaneous pancreas and kidney transplantation. Am J Transplant 2018; 18:2610-2611. [PMID: 29745003 DOI: 10.1111/ajt.14927] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- Delphine Kervella
- Centre de Recherche en Transplantation et Immunologie, Inserm Nantes, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie, CHU Nantes, Nantes, France
| | - Emeric Scharbarg
- Service d'endocrinologie, maladies métaboliques, et nutrition, CHU Nantes, Nantes, France
| | - Pascale Mahot-Moreau
- Service d'endocrinologie, maladies métaboliques, et nutrition, CHU Nantes, Nantes, France
| | - Karine Renaudin
- Centre de Recherche en Transplantation et Immunologie, Inserm Nantes, Université de Nantes, Nantes, France.,Service d'anatomie et cytologie pathologique Nantes, CHU Nantes, Nantes, France
| | - Julien Branchereau
- Centre de Recherche en Transplantation et Immunologie, Inserm Nantes, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie, CHU Nantes, Nantes, France
| | - Georges Karam
- Institut de Transplantation Urologie Néphrologie, CHU Nantes, Nantes, France
| | - Lucy Chaillous
- Service d'endocrinologie, maladies métaboliques, et nutrition, CHU Nantes, Nantes, France
| | - Kalyane Bach
- Service de Biochimie, CHU Nantes, Nantes, France
| | - Diego Cantarovich
- Centre de Recherche en Transplantation et Immunologie, Inserm Nantes, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie, CHU Nantes, Nantes, France
| |
Collapse
|
27
|
Preoperative Muscle Volume Predicts Graft Survival After Pancreas Transplantation: A Retrospective Observational Cohort Study. Transplant Proc 2018; 50:1482-1488. [DOI: 10.1016/j.transproceed.2018.03.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 03/06/2018] [Indexed: 12/15/2022]
|
28
|
Human urine-derived stem cells play a novel role in the treatment of STZ-induced diabetic mice. J Mol Histol 2018; 49:419-428. [PMID: 29675567 DOI: 10.1007/s10735-018-9772-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 04/16/2018] [Indexed: 12/19/2022]
Abstract
Human urine-derived stem cells (hUSCs) are a potential stem cell source for cell therapy. However, the effect of hUSCs on glucose metabolism regulation in type 1 diabetes was not clear. Therefore, the aim of the study was to evaluate whether hUSCs have protective effect on streptozotocin (STZ)-induced diabetes. hUSCs were extracted and cultivated with a special culture medium. Flow cytometry analysis was applied to detect cell surface markers. BALB/c male nude mice were either injected with high-dose STZ (HD-STZ) or multiple low-dose STZ (MLD-STZ). Serum and pancreatic insulin were measured, islet morphology and its vascularization were investigated. hUSCs highly expressed CD29, CD73, CD90 and CD146, and could differentiate into, at least, bone and fat in vitro. Transplantation of hUSCs into HD-STZ treated mice prolonged the median survival time and improved their blood glucose, and into those with MLD-STZ improved the glucose tolerance, islet morphology and increased the serum and pancreas insulin content. Furthermore, CD31 expression increased significantly in islets of BALB/c nude mice treated with hUSCs compared to those of un-transplanted MLD-STZ mice. hUSCs could improve the median survival time and glucose homeostasis in STZ-treated mice through promoting islet vascular regeneration and pancreatic beta-cell survival.
Collapse
|
29
|
Murai N, Ohtaki H, Watanabe J, Xu Z, Sasaki S, Yagura K, Shioda S, Nagasaka S, Honda K, Izumizaki M. Intrapancreatic injection of human bone marrow-derived mesenchymal stem/stromal cells alleviates hyperglycemia and modulates the macrophage state in streptozotocin-induced type 1 diabetic mice. PLoS One 2017; 12:e0186637. [PMID: 29073149 PMCID: PMC5657972 DOI: 10.1371/journal.pone.0186637] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 10/04/2017] [Indexed: 12/11/2022] Open
Abstract
Type 1 diabetes mellitus is a progressive disease caused by the destruction of pancreatic β-cells, resulting in insulin dependency and hyperglycemia. While transplanted bone marrow-derived mesenchymal stem/stromal cells (BMMSCs) have been explored as an alternative therapeutic approach for diseases, the choice of delivery route may be a critical factor determining their sustainability. This study evaluated the effects of intrapancreatic and intravenous injection of human BMMSCs (hBMMSCs) in streptozotocin (STZ)-induced type 1 diabetic mouse model. C57/BL6 mice were intraperitoneally injected with 115 mg/kg STZ on day 0. hBMMSCs (1 × 106 cells) or vehicle were injected into the pancreas or jugular vein on day 7. Intrapancreatic, but not intravenous, hBMMSC injection significantly reduced blood glucose levels on day 28 compared with vehicle injection by the same route. This glucose-lowering effect was not induced by intrapancreatic injection of human fibroblasts as the xenograft control. Intrapancreatically injected fluorescence-labeled hBMMSCs were observed in the intra- and extra-lobular spaces of the pancreas, and intravenously injected cells were in the lung region, although the number of cells mostly decreased within 2 weeks of injection. For hBMMSCs injected twice into the pancreatic region on days 7 and 28, the injected mice had further reduced blood glucose to borderline diabetic levels on day 56. Animals injected with hBMMSCs twice exhibited increases in the plasma insulin level, number and size of islets, insulin-positive proportion of the total pancreas area, and intensity of insulin staining compared with vehicle-injected animals. We found a decrease of Iba1-positive cells in islets and an increase of CD206-positive cells in both the endocrine and exocrine pancreas. The hBMMSC injection also reduced the number of CD40-positive cells merged with glucagon immunoreactions in the islets. These results suggest that intrapancreatic injection may be a better delivery route of hBMMSCs for the treatment of type 1 diabetes mellitus.
Collapse
Affiliation(s)
- Norimitsu Murai
- Department of Physiology, Showa University School of Medicine, Tokyo, Japan
- Department of Anatomy, Showa University School of Medicine, Tokyo, Japan
- Division of Diabetes, Metabolism and Endocrinology, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Hirokazu Ohtaki
- Department of Anatomy, Showa University School of Medicine, Tokyo, Japan
| | - Jun Watanabe
- Department of Anatomy, Showa University School of Medicine, Tokyo, Japan
- Center for Biotechnology, Showa University, Tokyo, Japan
| | - Zhifang Xu
- Department of Anatomy, Showa University School of Medicine, Tokyo, Japan
| | - Shun Sasaki
- Department of Anatomy, Showa University School of Medicine, Tokyo, Japan
| | - Kazumichi Yagura
- Department of Anatomy, Showa University School of Medicine, Tokyo, Japan
| | - Seiji Shioda
- Peptide Drug Innovation, Global Research Center for Innovative Life Science, Hoshi University School of Pharmacy and Pharmaceutical Sciences, Tokyo, Japan
| | - Shoichiro Nagasaka
- Division of Diabetes, Metabolism and Endocrinology, Showa University Fujigaoka Hospital, Yokohama, Japan
| | - Kazuho Honda
- Department of Anatomy, Showa University School of Medicine, Tokyo, Japan
| | - Masahiko Izumizaki
- Department of Physiology, Showa University School of Medicine, Tokyo, Japan
- * E-mail:
| |
Collapse
|
30
|
Kent SC, Mannering SI, Michels AW, Babon JAB. Deciphering the Pathogenesis of Human Type 1 Diabetes (T1D) by Interrogating T Cells from the "Scene of the Crime". Curr Diab Rep 2017; 17:95. [PMID: 28864875 PMCID: PMC5600889 DOI: 10.1007/s11892-017-0915-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE OF REVIEW Autoimmune-mediated destruction of insulin-producing β-cells within the pancreas results in type 1 diabetes (T1D), which is not yet preventable or curable. Previously, our understanding of the β-cell specific T cell repertoire was based on studies of autoreactive T cell responses in the peripheral blood of patients at risk for, or with, T1D; more recently, investigations have included immunohistochemical analysis of some T cell specificities in the pancreas from organ donors with T1D. Now, we are able to examine live, islet-infiltrating T cells from donors with T1D. RECENT FINDINGS Analysis of the T cell repertoire isolated directly from the pancreatic islets of donors with T1D revealed pro-inflammatory T cells with targets of known autoantigens, including proinsulin and glutamic acid decarboxylase, as well as modified autoantigens. We have assayed the islet-infiltrating T cell repertoire for autoreactivity and function directly from the inflamed islets of T1D organ donors. Design of durable treatments for prevention of or therapy for T1D requires understanding this repertoire.
Collapse
Affiliation(s)
- Sally C Kent
- Department of Medicine, Division of Diabetes, Diabetes Center of Excellence, ASC7-2041, University of Massachusetts Medical School, Worcester, MA, 01605, USA.
| | - Stuart I Mannering
- Immunology and Diabetes Unit, St. Vincent's Institute of Medical Research, 9 Princes Street, Fitzroy, Victoria, 3065, Australia
- Department of Medicine, University of Melbourne, St. Vincent's Hospital, Fitzroy, Victoria, 3065, Australia
| | - Aaron W Michels
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora, CO, USA
| | - Jenny Aurielle B Babon
- Department of Medicine, Division of Diabetes, Diabetes Center of Excellence, ASC7-2041, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| |
Collapse
|
31
|
Kolb H, von Herrath M. Immunotherapy for Type 1 Diabetes: Why Do Current Protocols Not Halt the Underlying Disease Process? Cell Metab 2017; 25:233-241. [PMID: 27839907 DOI: 10.1016/j.cmet.2016.10.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
T cell-directed immunosuppression only transiently delays the loss of β cell function in recent-onset type 1 diabetes. We argue here that the underlying disease process is carried by innate immune reactivity. Inducing a non-polarized functional state of local innate immunity will support regulatory T cell development and β cell proliferation.
Collapse
Affiliation(s)
- Hubert Kolb
- West-German Centre of Diabetes and Health, Düsseldorf Catholic Hospital Group, Hohensandweg 37, 40591 Düsseldorf, Germany; Faculty of Medicine, University of Düsseldorf, 40225 Düsseldorf, Germany.
| | - Matthias von Herrath
- Type 1 Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92014, USA; Novo Nordisk Diabetes Research and Development Center, Seattle, WA 98191, USA.
| |
Collapse
|
32
|
Sharples EJ, Mittal SM, Friend PJ. Challenges in pancreas transplantation. Acta Diabetol 2016; 53:871-878. [PMID: 27283012 DOI: 10.1007/s00592-016-0865-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 02/22/2016] [Indexed: 01/26/2023]
Abstract
Whole-organ pancreas transplantation, either alone or combined with a kidney transplant, is the only definitive treatment for many patients with type 1 diabetes that restores normal glucose homoeostasis and insulin independence. Pancreas transplantation delays, or potentially prevents, secondary diabetes complications and is associated with improvement in patient survival when compared with either patients remaining on the waiting list or those receiving kidney transplant alone. Pancreas transplantation is safe and effective, with 1-year patient survival >97 % and graft survival rates of 85 % at 1 year and 76 % at 5 years in recent UK data. This review focuses on some current areas of interest in pancreas transplantation.
Collapse
Affiliation(s)
- E J Sharples
- Oxford Transplant Centre, Churchill Hospital, Oxford University Hospitals, Old Road, Headington, Oxford, OX3 7LE, UK.
| | - S M Mittal
- Oxford Transplant Centre, Churchill Hospital, Oxford University Hospitals, Old Road, Headington, Oxford, OX3 7LE, UK
| | - P J Friend
- Nuffield Department of Surgery, Oxford University, Oxford, UK
| |
Collapse
|
33
|
Rudolph EN, Dunn TB, Mauer D, Noreen H, Sutherland DER, Kandaswamy R, Finger EB. HLA-A, -B, -C, -DR, and -DQ Matching in Pancreas Transplantation: Effect on Graft Rejection and Survival. Am J Transplant 2016; 16:2401-12. [PMID: 26814363 DOI: 10.1111/ajt.13734] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 12/23/2015] [Accepted: 01/09/2016] [Indexed: 01/25/2023]
Abstract
To enhance selection of appropriate deceased donors for pancreas transplants, we sought to determine whether HLA matching improved posttransplantation outcomes. In this single-center study of 1219 pancreas transplants, we correlated posttransplantation outcomes with HLA-A, -B, -C, -DR, and -DQ matches and mismatches. Rejection was linearly correlated with the number of mismatches. The individual number of HLA mismatches reached significance at four or more with a 2.3- to 2.9-fold increase in rejection. The effect was most predominant with HLA-B (1.8-fold with one mismatch and 2.0-fold with two mismatches) and -DR (1.9-fold with two mismatches) loci, whereas HLA-A, -C, and -DQ matches or mismatches did not independently predict acute rejection. The affect was strongest in solitary pancreas transplants, with little impact for simultaneous pancreas and kidney (SPK). In contrast, HLA matching did not affect graft or patient survival rates but was associated with a reduced risk of opportunistic infection. Avoidance of acute rejection saved an estimated $32 000 for solitary pancreas recipients and $52 000 for SPK recipients in hospital costs. Our data do not support the use of HLA matching for predicting pancreas graft survival but do support its significance for the reduction of acute rejection, particularly for solitary pancreas recipients.
Collapse
Affiliation(s)
- E N Rudolph
- Department of Surgery, Division of Transplantation, University of Minnesota, Minneapolis, MN
| | - T B Dunn
- Department of Surgery, Division of Transplantation, University of Minnesota, Minneapolis, MN
| | - D Mauer
- Immunology/Histocompatibility Laboratory, University of Minnesota, Minneapolis, MN
| | - H Noreen
- Immunology/Histocompatibility Laboratory, University of Minnesota, Minneapolis, MN
| | - D E R Sutherland
- Department of Surgery, Division of Transplantation, University of Minnesota, Minneapolis, MN
| | - R Kandaswamy
- Department of Surgery, Division of Transplantation, University of Minnesota, Minneapolis, MN
| | - E B Finger
- Department of Surgery, Division of Transplantation, University of Minnesota, Minneapolis, MN
| |
Collapse
|
34
|
Lauria MW, Ribeiro-Oliveira A. Diabetes and other endocrine-metabolic abnormalities in the long-term follow-up of pancreas transplantation. Clin Diabetes Endocrinol 2016; 2:14. [PMID: 28702248 PMCID: PMC5471933 DOI: 10.1186/s40842-016-0032-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 07/03/2016] [Indexed: 12/12/2022] Open
Abstract
Pancreas transplantation (PTX) has been demonstrated to restore long-term glucose homeostasis beyond what can be achieved by intensive insulin therapy or islet transplants. Moreover, PTX has been shown to decrease the progression of the chronic complications of diabetes. However, PTX patients require chronic use of immunosuppressive drugs with potential side effects. The long-term follow-up of PTX patients demands special care regarding metabolic deviations, infectious complications, and chronic rejection. Diabetes and other endocrine metabolic abnormalities following transplantation are common and can increase morbidity and mortality. Previous recipient-related and donor-related factors, as well as other aspects inherent to the transplant, act together in the pathogenesis of those abnormalities. Early recognition of these disturbances is the key to timely treatment; however, adequate tools to achieve this goal are often lacking. In a way, the type of PTX procedure, whether simultaneous pancreas kidney or not, seems to differentially influence the evolution of endocrine and metabolic abnormalities. Further studies are needed to define the best approach for PTX patients. This review will focus on the most common endocrine metabolic disorders seen in the long-term management of PTX: diabetes mellitus, hyperlipidemia, and bone loss. The authors here cover each one of these endocrine topics by showing the evaluation as well as proper management in the follow-up after PTX.
Collapse
Affiliation(s)
- Marcio W Lauria
- Department of Internal Medicine (Endocrinology section and Transplantation unit), Federal University of Minas Gerais, Rua Alfredo Balena, 190, 30130-100 Belo Horizonte, MG Brazil
| | - Antonio Ribeiro-Oliveira
- Department of Internal Medicine (Endocrinology section and Transplantation unit), Federal University of Minas Gerais, Rua Alfredo Balena, 190, 30130-100 Belo Horizonte, MG Brazil
| |
Collapse
|
35
|
Abstract
PURPOSE OF REVIEW The review analyzes the current biomarkers used in monitoring pancreas transplant, from the simple and time-tested, to more sophisticated, including markers of allo- and autoimmunity, that are likely to play a larger role in future studies. RECENT FINDINGS Evaluation of alloimmunity includes serum levels of donor-specific antibody, and, ultimately, pancreas transplant biopsies with C4d staining. Our center has focused on markers of autoimmunity, including assessment of autoantibodies and autoreactive T cells. We have found that conversion of autoantibodies (including GAD65, IA-2, and ZnT8), or the development of a new positive autoantibody, particularly ZnT8, are associated with type 1 diabetes (T1D) recurrence in the pancreas transplant. Autoreactive T cells have also been identified in the peripheral blood, pancreas transplant and peripancreas transplant-lymph nodes, that have the potential to mediate human β/islet cell destruction in vivo. SUMMARY The monitoring of pancreas transplant biomarkers, particularly those associated with autoimmunity, has led to new insights into the pathogenesis of T1D. Progress in the elucidation of mechanisms of autoimmunity may lead to novel therapeutic approaches to both T1D recurrence of the pancreas transplant and perhaps also new onset T1D.
Collapse
|
36
|
Kitchens WH, Turgeon NA. Immunosuppression in Pancreas Transplantation: What Has Changed in 20 Years? CURRENT TRANSPLANTATION REPORTS 2016. [DOI: 10.1007/s40472-016-0098-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
37
|
Burke GW, Vendrame F, Virdi SK, Ciancio G, Chen L, Ruiz P, Messinger S, Reijonen HK, Pugliese A. Lessons From Pancreas Transplantation in Type 1 Diabetes: Recurrence of Islet Autoimmunity. Curr Diab Rep 2015; 15:121. [PMID: 26547222 DOI: 10.1007/s11892-015-0691-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Type 1 diabetes recurrence (T1DR) affecting pancreas transplants was first reported in recipients of living-related pancreas grafts from twins or HLA identical siblings; given HLA identity, recipients received no or minimal immunosuppression. This observation provided critical evidence that type 1 diabetes (T1D) is an autoimmune disease. However, T1DR is traditionally considered very rare in immunosuppressed recipients of pancreas grafts from organ donors, representing the majority of recipients, and immunological graft failures are ascribed to chronic rejection. We have been performing simultaneous pancreas-kidney (SPK) transplants for over 25 years and find that 6-8 % of our recipients develop T1DR, with symptoms usually becoming manifest on extended follow-up. T1DR is typically characterized by (1) variable degree of insulitis and loss of insulin staining, on pancreas transplant biopsy (with most often absent), minimal to moderate and rarely severe pancreas, and/or kidney transplant rejection; (2) the conversion of T1D-associated autoantibodies (to the autoantigens GAD65, IA-2, and ZnT8), preceding hyperglycemia by a variable length of time; and (3) the presence of autoreactive T cells in the peripheral blood, pancreas transplant, and/or peripancreatic transplant lymph nodes. There is no therapeutic regimen that so far has controlled the progression of islet autoimmunity, even when additional immunosuppression was added to the ongoing chronic regimens; we hope that further studies and, in particular, in-depth analysis of pancreas transplant biopsies with recurrent diabetes will help identify more effective therapeutic approaches.
Collapse
Affiliation(s)
- George W Burke
- Miami Transplant Institute, 1801 NW 9th Ave, Highland Professional Building, Miami, FL, 33136, USA.
- Department of Surgery, Division of Transplantation, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Francesco Vendrame
- Department of Medicine, Division of Endocrinology and Metabolism, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Sahil K Virdi
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - G Ciancio
- Miami Transplant Institute, 1801 NW 9th Ave, Highland Professional Building, Miami, FL, 33136, USA
- Department of Surgery, Division of Transplantation, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Linda Chen
- Miami Transplant Institute, 1801 NW 9th Ave, Highland Professional Building, Miami, FL, 33136, USA
- Department of Surgery, Division of Transplantation, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Phillip Ruiz
- Miami Transplant Institute, 1801 NW 9th Ave, Highland Professional Building, Miami, FL, 33136, USA
- Department of Surgery, Division of Transplantation, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Shari Messinger
- Department of Epidemiology and Public Health Sciences, Division of Biostatistics, University of Miami Miller School of Medicine, Miami, FL, USA
| | | | - Alberto Pugliese
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Medicine, Division of Endocrinology and Metabolism, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|