1
|
Wang Q, Huang YX, Liu L, Zhao XH, Sun Y, Mao X, Li SW. Pancreatic islet transplantation: current advances and challenges. Front Immunol 2024; 15:1391504. [PMID: 38887292 PMCID: PMC11180903 DOI: 10.3389/fimmu.2024.1391504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 05/20/2024] [Indexed: 06/20/2024] Open
Abstract
Diabetes is a prevalent chronic disease that traditionally requires severe reliance on medication for treatment. Oral medication and exogenous insulin can only temporarily maintain blood glucose levels and do not cure the disease. Most patients need life-long injections of exogenous insulin. In recent years, advances in islet transplantation have significantly advanced the treatment of diabetes, allowing patients to discontinue exogenous insulin and avoid complications.Long-term follow-up results from recent reports on islet transplantation suggest that they provide significant therapeutic benefit although patients still require immunotherapy, suggesting the importance of future transplantation strategies. Although organ shortage remains the primary obstacle for the development of islet transplantation, new sources of islet cells, such as stem cells and porcine islet cells, have been proposed, and are gradually being incorporated into clinical research. Further research on new transplantation sites, such as the subcutaneous space and mesenteric fat, may eventually replace the traditional portal vein intra-islet cell infusion. Additionally, the immunological rejection reaction in islet transplantation will be resolved through the combined application of immunosuppressant agents, islet encapsulation technology, and the most promising mesenchymal stem cells/regulatory T cell and islet cell combined transplantation cell therapy. This review summarizes the progress achieved in islet transplantation, and discusses the research progress and potential solutions to the challenges faced.
Collapse
Affiliation(s)
- Qi Wang
- Department of Hepatobiliary and Pancreatic Surgery, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Yu-xi Huang
- Department of Hepatobiliary and Pancreatic Surgery, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Long Liu
- Department of Hepatobiliary and Pancreatic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiao-hong Zhao
- Department of Pharmacy, Taizhou Hospital, Zhejiang University, Taizhou, Zhejiang, China
| | - Yi Sun
- MRL Global Medical Affairs, MSD China, Shanghai, China
| | - Xinli Mao
- Department of Gastroenterology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
- Key Laboratory of Minimally Invasive Techniques and Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Shao-wei Li
- Department of Gastroenterology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
- Key Laboratory of Minimally Invasive Techniques and Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| |
Collapse
|
2
|
Garcia Valencia OA, Thongprayoon C, Jadlowiec CC, Mao SA, Leeaphorn N, Budhiraja P, Khoury N, Vaitla P, Suppadungsuk S, Cheungpasitporn W. Evaluating Global and Temporal Trends in Pancreas and Islet Cell Transplantation: Public Awareness and Engagement. Clin Pract 2024; 14:590-601. [PMID: 38666804 PMCID: PMC11049129 DOI: 10.3390/clinpract14020046] [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: 01/19/2024] [Revised: 03/24/2024] [Accepted: 03/27/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Pancreas transplantation is a crucial surgical intervention for managing diabetes, but it faces challenges such as its invasive nature, stringent patient selection criteria, organ scarcity, and centralized expertise. Despite the steadily increasing number of pancreas transplants in the United States, there is a need to understand global trends in interest to increase awareness of and participation in pancreas and islet cell transplantation. METHODS We analyzed Google Search trends for "Pancreas Transplantation" and "Islet Cell Transplantation" from 2004 to 14 November 2023, assessing variations in search interest over time and across geographical locations. The Augmented Dickey-Fuller (ADF) test was used to determine the stationarity of the trends (p < 0.05). RESULTS Search interest for "Pancreas Transplantation" varied from its 2004 baseline, with a general decline in peak interest over time. The lowest interest was in December 2010, with a slight increase by November 2023. Ecuador, Kuwait, and Saudi Arabia showed the highest search interest. "Islet Cell Transplantation" had its lowest interest in December 2016 and a more pronounced decline over time, with Poland, China, and South Korea having the highest search volumes. In the U.S., "Pancreas Transplantation" ranked 4th in interest, while "Islet Cell Transplantation" ranked 11th. The ADF test confirmed the stationarity of the search trends for both procedures. CONCLUSIONS "Pancreas Transplantation" and "Islet Cell Transplantation" showed initial peaks in search interest followed by a general downtrend. The stationary search trends suggest a lack of significant fluctuations or cyclical variations. These findings highlight the need for enhanced educational initiatives to increase the understanding and awareness of these critical transplant procedures among the public and professionals.
Collapse
Affiliation(s)
- Oscar A. Garcia Valencia
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA; (O.A.G.V.); (S.S.); (W.C.)
| | - Charat Thongprayoon
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA; (O.A.G.V.); (S.S.); (W.C.)
| | - Caroline C. Jadlowiec
- Division of Transplant Surgery, Department of Surgery, Mayo Clinic, Phoenix, AZ 85054, USA;
| | - Shennen A. Mao
- Division of Transplant Surgery, Department of Surgery, Mayo Clinic, Jacksonville, FL 32224, USA; (S.A.M.); (N.L.)
| | - Napat Leeaphorn
- Division of Transplant Surgery, Department of Surgery, Mayo Clinic, Jacksonville, FL 32224, USA; (S.A.M.); (N.L.)
| | - Pooja Budhiraja
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Phoenix, AZ 85054, USA;
| | - Nadeen Khoury
- Division of Nephrology, Henry Ford Hospital, Detroit, MI 48202, USA;
| | - Pradeep Vaitla
- Division of Nephrology, University of Mississippi Medical Center, Jackson, MS 39216, USA;
| | - Supawadee Suppadungsuk
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA; (O.A.G.V.); (S.S.); (W.C.)
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan 10540, Thailand
| | - Wisit Cheungpasitporn
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA; (O.A.G.V.); (S.S.); (W.C.)
| |
Collapse
|
3
|
Grosu-Bularda A, Hodea FV, Zamfirescu D, Stoian A, Teodoreanu RN, Lascăr I, Hariga CS. Exploring Costimulatory Blockade-Based Immunologic Strategies in Transplantation: Are They a Promising Immunomodulatory Approach for Organ and Vascularized Composite Allotransplantation? J Pers Med 2024; 14:322. [PMID: 38541064 PMCID: PMC10971463 DOI: 10.3390/jpm14030322] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 03/12/2024] [Accepted: 03/18/2024] [Indexed: 11/12/2024] Open
Abstract
The field of transplantation, including the specialized area of vascularized composite allotransplantation (VCA), has been transformed since the first hand transplant in 1998. The major challenge in VCA comes from the need for life-long immunosuppressive therapy due to its non-vital nature and a high rate of systemic complications. Ongoing research is focused on immunosuppressive therapeutic strategies to avoid toxicity and promote donor-specific tolerance. This includes studying the balance between tolerance and effector mechanisms in immune modulation, particularly the role of costimulatory signals in T lymphocyte activation. Costimulatory signals during T cell activation can have either stimulatory or inhibitory effects. Interfering with T cell activation through costimulation blockade strategies shows potential in avoiding rejection and prolonging the survival of transplanted organs. This review paper aims to summarize current data on the immunologic role of costimulatory blockade in the field of transplantation. It focuses on strategies that can be applied in vascularized composite allotransplantation, offering insights into novel methods for enhancing the success and safety of these procedures.
Collapse
Affiliation(s)
- Andreea Grosu-Bularda
- Department 11, Discipline Plastic and Reconstructive Surgery, Bucharest Clinical Emergency Hospital, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (R.N.T.); (I.L.); (C.S.H.)
- Clinic of Plastic Surgery, Aesthetic and Reconstructive Microsurgery, Emergency Clinical Hospital Bucharest, 050474 Bucharest, Romania
| | - Florin-Vlad Hodea
- Department 11, Discipline Plastic and Reconstructive Surgery, Bucharest Clinical Emergency Hospital, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (R.N.T.); (I.L.); (C.S.H.)
- Clinic of Plastic Surgery, Aesthetic and Reconstructive Microsurgery, Emergency Clinical Hospital Bucharest, 050474 Bucharest, Romania
| | | | | | - Răzvan Nicolae Teodoreanu
- Department 11, Discipline Plastic and Reconstructive Surgery, Bucharest Clinical Emergency Hospital, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (R.N.T.); (I.L.); (C.S.H.)
- Clinic of Plastic Surgery, Aesthetic and Reconstructive Microsurgery, Emergency Clinical Hospital Bucharest, 050474 Bucharest, Romania
| | - Ioan Lascăr
- Department 11, Discipline Plastic and Reconstructive Surgery, Bucharest Clinical Emergency Hospital, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (R.N.T.); (I.L.); (C.S.H.)
- Clinic of Plastic Surgery, Aesthetic and Reconstructive Microsurgery, Emergency Clinical Hospital Bucharest, 050474 Bucharest, Romania
| | - Cristian Sorin Hariga
- Department 11, Discipline Plastic and Reconstructive Surgery, Bucharest Clinical Emergency Hospital, University of Medicine and Pharmacy Carol Davila, 050474 Bucharest, Romania; (A.G.-B.); (R.N.T.); (I.L.); (C.S.H.)
- Clinic of Plastic Surgery, Aesthetic and Reconstructive Microsurgery, Emergency Clinical Hospital Bucharest, 050474 Bucharest, Romania
| |
Collapse
|
4
|
Martin TM, Burke SJ, Wasserfall CH, Collier JJ. Islet beta-cells and intercellular adhesion molecule-1 (ICAM-1): Integrating immune responses that influence autoimmunity and graft rejection. Autoimmun Rev 2023; 22:103414. [PMID: 37619906 PMCID: PMC10543623 DOI: 10.1016/j.autrev.2023.103414] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 08/16/2023] [Accepted: 08/20/2023] [Indexed: 08/26/2023]
Abstract
Type 1 diabetes (T1D) develops due to autoimmune targeting of the pancreatic islet β-cells. Clinical symptoms arise from reduced insulin in circulation. The molecular events and interactions between discrete immune cell populations, infiltration of such leukocytes into pancreatic and islet tissue, and selective targeting of the islet β-cells during autoimmunity and graft rejection are not entirely understood. One protein central to antigen presentation, priming of immune cells, trafficking of leukocytes, and vital for leukocyte effector function is the intercellular adhesion molecule-1 (ICAM-1). The gene encoding ICAM-1 is transcriptionally regulated and rapidly responsive (i.e., within hours) to pro-inflammatory cytokines. ICAM-1 is a transmembrane protein that can be glycosylated; its presence on the cell surface provides co-stimulatory functions for immune cell activation and stabilization of cell-cell contacts. ICAM-1 interacts with the β2-integrins, CD11a/CD18 (LFA-1) and CD11b/CD18 (Mac-1), which are present on discrete immune cell populations. A whole-body ICAM-1 deletion protects NOD mice from diabetes onset, strongly implicating this protein in autoimmune responses. Since several different cell types express ICAM-1, its biology is fundamentally essential for various physiological and pathological outcomes. Herein, we review the role of ICAM-1 during both autoimmunity and islet graft rejection to understand the mechanism(s) leading to islet β-cell death and dysfunction that results in insufficient circulating quantities of insulin to control glucose homeostasis.
Collapse
Affiliation(s)
- Thomas M Martin
- Laboratory of Islet Biology and Inflammation, Pennington Biomedical Research Center, Baton Rouge, LA 70808, United States of America; Department of Biological Sciences, Louisiana State University, Baton Rouge, LA 70803, United States of America
| | - Susan J Burke
- Laboratory of Immunogenetics, Pennington Biomedical Research Center, Baton Rouge, LA 70808, United States of America
| | - Clive H Wasserfall
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, United States of America
| | - J Jason Collier
- Laboratory of Islet Biology and Inflammation, Pennington Biomedical Research Center, Baton Rouge, LA 70808, United States of America; Department of Biological Sciences, Louisiana State University, Baton Rouge, LA 70803, United States of America.
| |
Collapse
|
5
|
Wisel SA, Posselt AM, Szot GL, Nunez M, Santos-Parker K, Gardner JM, Worner G, Roll GR, Syed S, Kelly Y, Ward C, Tavakol M, Johnson K, Masharani U, Stock PG. A Multi-Modal Approach to Islet and Pancreas Transplantation With Calcineurin-Sparing Immunosuppression Maintains Long-Term Insulin Independence in Patients With Type I Diabetes. Transpl Int 2023; 36:11367. [PMID: 37359825 PMCID: PMC10285771 DOI: 10.3389/ti.2023.11367] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 05/24/2023] [Indexed: 06/28/2023]
Abstract
Long-term success in beta-cell replacement remains limited by the toxic effects of calcineurin inhibitors (CNI) on beta-cells and renal function. We report a multi-modal approach including islet and pancreas-after-islet (PAI) transplant utilizing calcineurin-sparing immunosuppression. Ten consecutive non-uremic patients with Type 1 diabetes underwent islet transplant with immunosuppression based on belatacept (BELA; n = 5) or efalizumab (EFA; n = 5). Following islet failure, patients were considered for repeat islet infusion and/or PAI transplant. 70% of patients (four EFA, three BELA) maintained insulin independence at 10 years post-islet transplant, including four patients receiving a single islet infusion and three patients undergoing PAI transplant. 60% remain insulin independent at mean follow-up of 13.3 ± 1.1 years, including one patient 9 years after discontinuing all immunosuppression for adverse events, suggesting operational tolerance. All patients who underwent repeat islet transplant experienced graft failure. Overall, patients demonstrated preserved renal function, with a mild decrease in GFR from 76.5 ± 23.1 mL/min to 50.2 ± 27.1 mL/min (p = 0.192). Patients undergoing PAI showed the greatest degree of renal impairment following initiation of CNI (56% ± 18.7% decrease in GFR). In our series, repeat islet transplant is ineffective at maintaining long-term insulin independence. PAI results in durable insulin independence but is associated with impaired renal function secondary to CNI dependence.
Collapse
Affiliation(s)
- Steven A. Wisel
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Andrew M. Posselt
- Division of Transplantation, University of California, San Francisco, San Francisco, CA, United States
| | - Gregory L. Szot
- Division of Transplantation, University of California, San Francisco, San Francisco, CA, United States
| | - Miguel Nunez
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Keli Santos-Parker
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - James M. Gardner
- Division of Transplantation, University of California, San Francisco, San Francisco, CA, United States
| | - Giulia Worner
- Division of Transplantation, University of California, San Francisco, San Francisco, CA, United States
| | - Garrett R. Roll
- Division of Transplantation, University of California, San Francisco, San Francisco, CA, United States
| | - Shareef Syed
- Division of Transplantation, University of California, San Francisco, San Francisco, CA, United States
| | - Yvonne Kelly
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Casey Ward
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Medhi Tavakol
- Division of Transplantation, University of California, San Francisco, San Francisco, CA, United States
| | - Kristina Johnson
- Division of Transplantation, University of California, San Francisco, San Francisco, CA, United States
| | - Umesh Masharani
- Division of Endocrinology, University of California, San Francisco, San Francisco, CA, United States
| | - Peter G. Stock
- Division of Transplantation, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
6
|
Duneton C, Winterberg PD, Ford ML. Activation and regulation of alloreactive T cell immunity in solid organ transplantation. Nat Rev Nephrol 2022; 18:663-676. [PMID: 35902775 PMCID: PMC9968399 DOI: 10.1038/s41581-022-00600-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/23/2022] [Indexed: 01/18/2023]
Abstract
Transplantation is the only curative treatment for patients with kidney failure but it poses unique immunological challenges that must be overcome to prevent allograft rejection and ensure long-term graft survival. Alloreactive T cells are important contributors to graft rejection, and a clearer understanding of the mechanisms by which these cells recognize donor antigens - through direct, indirect or semi-direct pathways - will facilitate their therapeutic targeting. Post-T cell priming rejection responses can also be modified by targeting pathways that regulate T cell trafficking, survival cytokines or innate immune activation. Moreover, the quantity and quality of donor-reactive memory T cells crucially shape alloimmune responses. Of note, many fundamental concepts in transplant immunology have been derived from models of infection. However, the programmed differentiation of allograft-specific T cell responses is probably distinct from that of pathogen-elicited responses, owing to the dearth of pathogen-derived innate immune activation in the transplantation setting. Understanding the fundamental (and potentially unique) immunological pathways that lead to allograft rejection is therefore a prerequisite for the rational development of therapeutics that promote transplantation tolerance.
Collapse
Affiliation(s)
- Charlotte Duneton
- Paediatric Nephrology, Robert Debré Hospital, Paris, France
- Emory Transplant Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Pamela D Winterberg
- Paediatric Nephrology, Emory University Department of Paediatrics and Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Mandy L Ford
- Emory Transplant Center, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
7
|
Soltani S, Mansouri K, Emami Aleagha MS, Moasefi N, Yavari N, Shakouri SK, Notararigo S, Shojaeian A, Pociot F, Yarani R. Extracellular Vesicle Therapy for Type 1 Diabetes. Front Immunol 2022; 13:865782. [PMID: 35464488 PMCID: PMC9024141 DOI: 10.3389/fimmu.2022.865782] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 03/17/2022] [Indexed: 01/02/2023] Open
Abstract
Type 1 diabetes (T1D) is a chronic disorder characterized by immune-mediated destruction of pancreatic insulin-producing β-cells. The primary treatment for T1D is multiple daily insulin injections to control blood sugar levels. Cell-free delivery packets with therapeutic properties, extracellular vesicles (EVs), mainly from stem cells, have recently gained considerable attention for disease treatments. EVs provide a great potential to treat T1D ascribed to their regenerative, anti-inflammatory, and immunomodulatory effects. Here, we summarize the latest EV applications for T1D treatment and highlight opportunities for further investigation.
Collapse
Affiliation(s)
- Setareh Soltani
- Clinical Research Development Center, Taleghani and Imam Ali Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Kamran Mansouri
- Medical Biology Research Center, Health Technology Institute, University of Medical Sciences, Kermanshah, Iran
| | - Mohammad Sajad Emami Aleagha
- Medical Technology Research Center (MTRC), School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Narges Moasefi
- Medical Technology Research Center (MTRC), School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Niloofar Yavari
- Department of Cellular and Molecular Medicine, The Panum Institute, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Seyed Kazem Shakouri
- Physical Medicine and Rehabilitation Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sara Notararigo
- Instituto de Investigación Sanitaria de Santiago (IDIS), Complejo Hospitalario Universitario de Santiago (CHUS), Servicio Gallego de Salud (SERGAS), Santiago de Compostela, Spain
| | - Ali Shojaeian
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Flemming Pociot
- Translational Type 1 Diabetes Research, Department of Clinical, Research, Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | - Reza Yarani
- Translational Type 1 Diabetes Research, Department of Clinical, Research, Steno Diabetes Center Copenhagen, Gentofte, Denmark
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, Stanford University School of Medicine, Palo Alto, CA, United States
- *Correspondence: Reza Yarani, ;
| |
Collapse
|
8
|
Shu J, Wei W, Zhang L. Identification of Molecular Signatures and Candidate Drugs in Vascular Dementia by Bioinformatics Analyses. Front Mol Neurosci 2022; 15:751044. [PMID: 35221911 PMCID: PMC8873373 DOI: 10.3389/fnmol.2022.751044] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 01/17/2022] [Indexed: 01/30/2023] Open
Abstract
Vascular dementia (VaD) is considered to be the second most common form of dementia after Alzheimer’s disease, and no specific drugs have been approved for VaD treatment. We aimed to identify shared transcriptomic signatures between the frontal cortex and temporal cortex in VaD by bioinformatics analyses. Gene ontology and pathway enrichment analyses, protein–protein interaction (PPI) and hub gene identification, hub gene–transcription factor interaction, hub gene–microRNA interaction, and hub gene–drug interaction analyses were performed. We identified 159 overlapping differentially expressed genes (DEGs) between the frontal cortex and temporal cortex that were enriched mainly in inflammation and innate immunity, synapse pruning, regeneration, positive regulation of angiogenesis, response to nutrient levels, and positive regulation of the digestive system process. We identified 10 hub genes in the PPI network (GNG13, CD163, C1QA, TLR2, SST, C1QB, ITGB2, CCR5, CRH, and TAC1), four central regulatory transcription factors (FOXC1, CREB1, GATA2, and HINFP), and four microRNAs (miR-27a-3p, miR-146a-5p, miR-335-5p, and miR-129-2-3p). Hub gene–drug interaction analysis found four drugs (maraviroc, cenicriviroc, PF-04634817, and efalizumab) that could be potential drugs for VaD treatment. Together, our results may contribute to understanding the underlying mechanisms in VaD and provide potential targets and drugs for therapeutic intervention.
Collapse
|
9
|
Liu W, Flanders JA, Wang LH, Liu Q, Bowers DT, Wang K, Chiu A, Wang X, Ernst AU, Shariati K, Caserto JS, Parker B, Gao D, Plesser MD, Grunnet LG, Rescan C, Carletto RP, Winkel L, Melero-Martin JM, Ma M. A Safe, Fibrosis-Mitigating, and Scalable Encapsulation Device Supports Long-Term Function of Insulin-Producing Cells. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2104899. [PMID: 34897997 PMCID: PMC8881301 DOI: 10.1002/smll.202104899] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/12/2021] [Indexed: 06/12/2023]
Abstract
Encapsulation and transplantation of insulin-producing cells offer a promising curative treatment for type 1 diabetes (T1D) without immunosuppression. However, biomaterials used to encapsulate cells often elicit foreign body responses, leading to cellular overgrowth and deposition of fibrotic tissue, which in turn diminishes mass transfer to and from transplanted cells. Meanwhile, the encapsulation device must be safe, scalable, and ideally retrievable to meet clinical requirements. Here, a durable and safe nanofibrous device coated with a thin and uniform, fibrosis-mitigating, zwitterionically modified alginate hydrogel for encapsulation of islets and stem cell-derived beta (SC-β) cells is reported. The device with a configuration that has cells encapsulated within the cylindrical wall, allowing scale-up in both radial and longitudinal directions without sacrificing mass transfer, is designed. Due to its facile mass transfer and low level of fibrotic reactions, the device supports long-term cell engraftment, correcting diabetes in C57BL6/J mice with rat islets for up to 399 days and SCID-beige mice with human SC-β cells for up to 238 days. The scalability and retrievability in dogs are further demonstrated. These results suggest the potential of this new device for cell therapies to treat T1D and other diseases.
Collapse
Affiliation(s)
- Wanjun Liu
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
- Key Laboratory of Textile Science & Technology, Ministry of Education, College of Textiles, Donghua University, Shanghai 201620, China
| | - James A. Flanders
- Department of Clinical Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Long-Hai Wang
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Qingsheng Liu
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Daniel T. Bowers
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Kai Wang
- Department of Cardiac Surgery, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Alan Chiu
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Xi Wang
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Alexander U. Ernst
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Kaavian Shariati
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Julia S. Caserto
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
- Robert Frederick Smith School of Chemical & Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Benjamin Parker
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Daqian Gao
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Mitchell D. Plesser
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Lars G. Grunnet
- Stem Cell Delivery & Pharmacology, Novo Nordisk A/S, 2760 Måløv, Denmark
| | - Claude Rescan
- Stem Cell Delivery & Pharmacology, Novo Nordisk A/S, 2760 Måløv, Denmark
| | | | - Louise Winkel
- Stem Cell Delivery & Pharmacology, Novo Nordisk A/S, 2760 Måløv, Denmark
| | - Juan M. Melero-Martin
- Department of Cardiac Surgery, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Minglin Ma
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
10
|
Louis K, Macedo C, Metes D. Targeting T Follicular Helper Cells to Control Humoral Allogeneic Immunity. Transplantation 2021; 105:e168-e180. [PMID: 33909968 PMCID: PMC8484368 DOI: 10.1097/tp.0000000000003776] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Humoral allogeneic immunity driven by anti-HLA donor-specific antibodies and antibody-mediated rejection (AMR) significantly impede prolonged survival of organ allografts after transplantation. Although the importance of T follicular helper (TFH) cells in controlling antibody responses has been long established, their role in directing donor-specific antibody generation leading to AMR was only recently appreciated in the clinical setting of organ transplantation. In this review, we provide a comprehensive summary of the current knowledge on the biology of human TFH cells as well as their circulating counterparts and describe their pivotal role in driving humoral alloimmunity. In addition, we discuss the intrinsic effects of current induction therapies and maintenance immunosuppressive drugs as well as of biotherapies on TFH cells and provide future directions and novel opportunities of biotherapeutic targeting of TFH cells that have the potential of bringing the prophylactic and curative treatments of AMR toward personalized and precision medicine.
Collapse
Affiliation(s)
- Kevin Louis
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
- Human Immunology and Immunopathology, Inserm UMR 976, Université de Paris, Paris, France
- Kidney Transplant Department, Saint Louis Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Camila Macedo
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Diana Metes
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
11
|
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
|
12
|
Liu J, Ting JP, Al-Azzam S, Ding Y, Afshar S. Therapeutic Advances in Diabetes, Autoimmune, and Neurological Diseases. Int J Mol Sci 2021; 22:ijms22062805. [PMID: 33802091 PMCID: PMC8001105 DOI: 10.3390/ijms22062805] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/02/2021] [Accepted: 03/06/2021] [Indexed: 02/08/2023] Open
Abstract
Since 2015, 170 small molecules, 60 antibody-based entities, 12 peptides, and 15 gene- or cell-therapies have been approved by FDA for diverse disease indications. Recent advancement in medicine is facilitated by identification of new targets and mechanisms of actions, advancement in discovery and development platforms, and the emergence of novel technologies. Early disease detection, precision intervention, and personalized treatments have revolutionized patient care in the last decade. In this review, we provide a comprehensive overview of current and emerging therapeutic modalities developed in the recent years. We focus on nine diseases in three major therapeutics areas, diabetes, autoimmune, and neurological disorders. The pathogenesis of each disease at physiological and molecular levels is discussed and recently approved drugs as well as drugs in the clinic are presented.
Collapse
Affiliation(s)
- Jinsha Liu
- Protein Engineering, Lilly Biotechnology Center, Eli Lilly and Company, San Diego, CA 92121, USA; (J.L.); (J.P.T.); (Y.D.)
| | - Joey Paolo Ting
- Protein Engineering, Lilly Biotechnology Center, Eli Lilly and Company, San Diego, CA 92121, USA; (J.L.); (J.P.T.); (Y.D.)
| | - Shams Al-Azzam
- Professional Scientific Services, Eurofins Lancaster Laboratories, Lancaster, PA 17605, USA;
| | - Yun Ding
- Protein Engineering, Lilly Biotechnology Center, Eli Lilly and Company, San Diego, CA 92121, USA; (J.L.); (J.P.T.); (Y.D.)
| | - Sepideh Afshar
- Protein Engineering, Lilly Biotechnology Center, Eli Lilly and Company, San Diego, CA 92121, USA; (J.L.); (J.P.T.); (Y.D.)
- Correspondence:
| |
Collapse
|
13
|
Bachul PJ, Golab K, Basto L, Zangan S, Pyda JS, Perez-Gutierrez A, Borek P, Wang LJ, Tibudan M, Tran DK, Anteby R, Generette GS, Chrzanowski J, Fendler W, Perea L, Jayant K, Lucander A, Thomas C, Philipson L, Millis JM, Fung J, Witkowski P. Post-Hoc Analysis of a Randomized, Double Blind, Prospective Study at the University of Chicago: Additional Standardizations of Trial Protocol are Needed to Evaluate the Effect of a CXCR1/2 Inhibitor in Islet Allotransplantation. Cell Transplant 2021; 30:9636897211001774. [PMID: 33908301 PMCID: PMC8085379 DOI: 10.1177/09636897211001774] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 01/14/2021] [Accepted: 02/22/2021] [Indexed: 12/11/2022] Open
Abstract
A recent randomized, multicenter trial did not show benefit of a CXCR1/2 receptor inhibitor (Reparixin) when analysis included marginal islet mass (>3,000 IEQ/kg) for allotransplantation and when immunosuppression regimens were not standardized among participating centers. We present a post-hoc analysis of trial patients from our center at the University of Chicago who received an islet mass of over 5,000 IEQ/kg and a standardized immunosuppression regimen of anti-thymocyte globulin (ATG) for induction. Twelve islet allotransplantation (ITx) recipients were randomized (2:1) to receive Reparixin (N = 8) or placebo (N = 4) in accordance with the multicenter trial protocol. Pancreas and donor characteristics did not differ between Reparixin and placebo groups. Five (62.5%) patients who received Reparixin, compared to none in the placebo group, achieved insulin independence after only one islet infusion and remained insulin-free for over 2 years (P = 0.08). Following the first ITx with ATG induction, distinct cytokine, chemokine, and miR-375 release profiles were observed for both the Reparixin and placebo groups. After excluding procedures with complications, islet engraftment on post-operative day 75 after a single transplant was higher in the Reparixin group (n = 7) than in the placebo (n = 3) group (P = 0.03) when islet graft function was measured by the ratio of the area under the curve (AUC) for c-peptide to glucose in mixed meal tolerance test (MMTT). Additionally, the rate of engraftment was higher when determined via BETA-2 score instead of MMTT (P = 0.01). Our analysis suggests that Reparixin may have improved outcomes compared to placebo when sufficient islet mass is transplanted and when standardized immunosuppression with ATG is used for induction. However, further studies are warranted. Investigation of Reparixin and other novel agents under more standardized and optimized conditions would help exclude confounding factors and allow for a more definitive evaluation of their role in improving outcomes in islet transplantation. Clinical trial reg. no. NCT01817959, clinicaltrials.gov.
Collapse
Affiliation(s)
- Piotr J. Bachul
- Department of Surgery, The Transplantation Institute, University of Chicago, Chicago, IL, USA
| | - Karolina Golab
- Department of Surgery, The Transplantation Institute, University of Chicago, Chicago, IL, USA
| | - Lindsay Basto
- Department of Surgery, The Transplantation Institute, University of Chicago, Chicago, IL, USA
| | - Steven Zangan
- Department of Radiology, University of Chicago, Chicago, IL, USA
| | - Jordan S. Pyda
- Department of Surgery, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | | | - Peter Borek
- Department of Surgery, The Transplantation Institute, University of Chicago, Chicago, IL, USA
| | - Ling-Jia Wang
- Department of Surgery, The Transplantation Institute, University of Chicago, Chicago, IL, USA
| | - Martin Tibudan
- Department of Surgery, The Transplantation Institute, University of Chicago, Chicago, IL, USA
| | - Dong-Kha Tran
- Department of Surgery, The Transplantation Institute, University of Chicago, Chicago, IL, USA
| | - Roi Anteby
- Department of Surgery, The Transplantation Institute, University of Chicago, Chicago, IL, USA
| | - Gabriela S. Generette
- Department of Surgery, The Transplantation Institute, University of Chicago, Chicago, IL, USA
| | - Jędrzej Chrzanowski
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, Lodz, Poland
| | - Wojciech Fendler
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, Lodz, Poland
| | - Laurencia Perea
- Department of Surgery, The Transplantation Institute, University of Chicago, Chicago, IL, USA
| | - Kumar Jayant
- Department of Surgery, The Transplantation Institute, University of Chicago, Chicago, IL, USA
| | - Aaron Lucander
- Department of Surgery, The Transplantation Institute, University of Chicago, Chicago, IL, USA
| | - Celeste Thomas
- Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Louis Philipson
- Department of Medicine, University of Chicago, Chicago, IL, USA
| | - J. Michael Millis
- Department of Surgery, The Transplantation Institute, University of Chicago, Chicago, IL, USA
| | - John Fung
- Department of Surgery, The Transplantation Institute, University of Chicago, Chicago, IL, USA
| | - Piotr Witkowski
- Department of Surgery, The Transplantation Institute, University of Chicago, Chicago, IL, USA
| |
Collapse
|
14
|
Matsumoto S, Shimoda M. Current situation of clinical islet transplantation from allogeneic toward xenogeneic. J Diabetes 2020; 12:733-741. [PMID: 32246528 DOI: 10.1111/1753-0407.13041] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 03/25/2020] [Indexed: 12/30/2022] Open
Abstract
Currently, type 1 diabetes requires lifelong insulin injection and careful blood glucose control to prevent secondary complications, but islet transplantation could make a type 1 diabetic patient insulin independent. On the other hand, islet transplantation needs human donors and donor shortage is the most serious issue. To alleviate the donor shortage, non-heart-beating and living donors were used; in addition, the efficacy of islet isolation and transplantation has been improved. However, the donor shortage issue will not be solved as long as human donors are the only source. To solve the donor shortage issue, islet xenotransplantation using porcine islets was initiated in 1994. Islet xenotransplantation has a potential to cure many type 1 diabetic patients, although there is the risk of developing serious or novel infection. Therefore, the World Health Organization has been interested in xenotransplantation, and the International Xenotransplantation Association (IXA) has published consensus statements to initiate xenogeneic islet transplantation. Clinical islet xenotransplantation was conducted under the official regulation, and safety and efficacy data have been accumulated. Currently an efficient method to overcome xenorejection is an important research target. In addition to traditional immunosuppressive drugs and immune isolation methods, the gene modification with CRISPR and blastocyst complementation have been investigated with promising outcomes. Once the xenorejection issue is overcome, islet xenotransplantation should become a curative treatment for type 1 diabetic patients.
Collapse
Affiliation(s)
- Shinichi Matsumoto
- Islet Transplantation Project, National Institute for Global Health and Medicine, Tokyo, Japan
| | - Masayuki Shimoda
- Islet Transplantation Project, National Institute for Global Health and Medicine, Tokyo, Japan
| |
Collapse
|
15
|
Brovkina O, Dashinimaev E. Advances and complications of regenerative medicine in diabetes therapy. PeerJ 2020; 8:e9746. [PMID: 33194345 PMCID: PMC7485501 DOI: 10.7717/peerj.9746] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 07/27/2020] [Indexed: 12/23/2022] Open
Abstract
The rapid development of technologies in regenerative medicine indicates clearly that their common application is not a matter of if, but of when. However, the regeneration of beta-cells for diabetes patients remains a complex challenge due to the plurality of related problems. Indeed, the generation of beta-cells masses expressing marker genes is only a first step, with maintaining permanent insulin secretion, their protection from the immune system and avoiding pathological modifications in the genome being the necessary next developments. The prospects of regenerative medicine in diabetes therapy were promoted by the emergence of promising results with embryonic stem cells (ESCs). Their pluripotency and proliferation in an undifferentiated state during culture have ensured the success of ESCs in regenerative medicine. The discovery of induced pluripotent stem cells (iPSCs) derived from the patients’ own mesenchymal cells has provided further hope for diabetes treatment. Nonetheless, the use of stem cells has significant limitations related to the pluripotent stage, such as the risk of development of teratomas. Thus, the direct conversion of mature cells into beta-cells could address this issue. Recent studies have shown the possibility of such transdifferentiation and have set trends for regeneration medicine, directed at minimizing genome modifications and invasive procedures. In this review, we will discuss the published results of beta-cell regeneration and the advantages and disadvantages illustrated by these experiments.
Collapse
Affiliation(s)
- Olga Brovkina
- Federal Research Clinical Center for Specialized Types of Health Care and Medical Technologies of Federal Medical and Biology Agency, Moscow, Russia
| | - Erdem Dashinimaev
- Koltzov Institute of Developmental Biology of Russian Academy of Sciences, Moscow, Russia.,Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
| |
Collapse
|
16
|
Ruhela A, Kasinathan GN, Rath SN, Sasikala M, Sharma CS. Electrospun freestanding hydrophobic fabric as a potential polymer semi-permeable membrane for islet encapsulation. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 118:111409. [PMID: 33255012 DOI: 10.1016/j.msec.2020.111409] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/04/2020] [Accepted: 08/11/2020] [Indexed: 01/13/2023]
Abstract
One of the significant problems associated with islet encapsulation for type 1 diabetes treatment is the loss of islet functionality or cell death after transplantation because of the unfavorable environment for the cells. In this work, we propose a simple strategy to fabricate electrospun membranes that will provide a favorable environment for proper islet function and also a desirable pore size to cease cellular infiltration, protecting the encapsulated islet from immune cells. By electrospinning the wettability of three different biocompatible polymers: cellulose acetate (CA), polyethersulfone (PES), and polytetrafluoroethylene (PTFE) was greatly modified. The contact angle of electrospun CA, PES, and PTFE increased to 136°, 126°, and 155° as compared to 55°, 71°, and 128° respectively as a thin film, making the electrospun membranes hydrophobic. Commercial porous membranes of PES and PTFE show a contact angle of 30° and 118°, respectively, confirming the hydrophobicity of electrospun membranes is due to the surface morphology induced by electrospinning. In- vivo results confirm that the induced hydrophobicity and surface morphology of electrospun membranes impede cell attachment, which would help in maintaining the 3D circular morphology of islet cell. More importantly, the pore size of 0.3-0.6 μm obtained due to the densely packed structure of nanofibers, will be able to restrict immune cells but would allow free movement of molecules like insulin and glucose. Therefore, electrospun polymer fibrous membranes as fabricated in this work, with hydrophobic and porous properties, make a strong case for successful islet encapsulation.
Collapse
Affiliation(s)
- Aakanksha Ruhela
- Creative & Advanced Research Based On Nanomaterials (CARBON) Laboratory, Department of Chemical Engineering, Indian Institute of Technology Hyderabad, Kandi 502285, Telangana, India
| | - Gokula Nathan Kasinathan
- Regenerative Medicine and Stem Cell Laboratory (RMS), Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi 502285, Telangana, India
| | - Subha N Rath
- Regenerative Medicine and Stem Cell Laboratory (RMS), Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi 502285, Telangana, India
| | - M Sasikala
- Asian Healthcare Foundation, Gachibowli, Hyderabad 500032, Telangana, India
| | - Chandra S Sharma
- Creative & Advanced Research Based On Nanomaterials (CARBON) Laboratory, Department of Chemical Engineering, Indian Institute of Technology Hyderabad, Kandi 502285, Telangana, India.
| |
Collapse
|
17
|
Berman A, Wszola M, Gorski L, Serwanska-Swietek M, Ostaszewska A, Lipinska A, Durlik M, Chmura A, Kwiatkowski A. Do We Need Insulin Independence After Islet Transplantation? Transplant Proc 2019; 51:2775-2780. [DOI: 10.1016/j.transproceed.2019.02.067] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 02/06/2019] [Indexed: 12/21/2022]
|
18
|
|
19
|
Knechtle SJ, Shaw JM, Hering BJ, Kraemer K, Madsen JC. Translational impact of NIH-funded nonhuman primate research in transplantation. Sci Transl Med 2019; 11:eaau0143. [PMID: 31292263 PMCID: PMC7197021 DOI: 10.1126/scitranslmed.aau0143] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 12/13/2018] [Indexed: 12/23/2022]
Abstract
The National Institutes of Health (NIH) has long supported using nonhuman primate (NHP) models for research on kidney, pancreatic islet, heart, and lung transplantation. The primary purpose of this research has been to develop new treatments for down-modulating or preventing deleterious immune responses after transplantation in human patients. Here, we discuss NIH-funded NHP studies of immune cell depletion, costimulation blockade, regulatory cell therapy, desensitization, and mixed hematopoietic chimerism that either preceded clinical trials or prevented the human application of therapies that were toxic or ineffective.
Collapse
Affiliation(s)
- Stuart J Knechtle
- Duke Transplant Center, Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA.
| | - Julia M Shaw
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Bernhard J Hering
- Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN 55455, USA
| | - Kristy Kraemer
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Joren C Madsen
- Center for Transplantation Sciences and Division of Cardiac Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
20
|
Veroux M, Bottino R, Santini R, Bertera S, Corona D, Zerbo D, Li Volti G, Ekser B, Puzzo L, Raffaele M, Lo Bianco S, Giaquinta A, Veroux P, Vanella L. Mesenteric lymph nodes as alternative site for pancreatic islet transplantation in a diabetic rat model. BMC Surg 2019; 18:126. [PMID: 31074398 PMCID: PMC7402566 DOI: 10.1186/s12893-018-0452-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 11/29/2018] [Indexed: 12/02/2022] Open
Abstract
Background Islet transplantation has progressively become a safe alternative to pancreas transplantation for the treatment of type 1 diabetes. However, the long-term results of islet transplantation could be significantly increased by improving the quality of the islet isolation technique even exploring alternative islet transplantation sites to reduce the number of islets required to mitigate hyperglycemia. The goal of the study was to test the lymph node as a suitable anatomical location for islet engraftment in a rodent model. Methods Forty Lewis rats, 6–8 weeks old, body weight 250–300 g, have been used as islet donors and recipients in syngeneic islet transplantation experiments. Ten rats were rendered diabetic by one injection of 65 mg/Kg of streptozotocin. After pancreas retrieval from non diabetic donors, islet were isolated and transplanted in the mesenteric lymph nodes of 7 diabetic rats. Rats were followed for 30 days after islet transplantation. Results A total of 7 islet transplantations in mesenteric lymph nodes have been performed. Two rats died 24 and 36 h after transplantation due to complications. No transplanted rat acquired normal glucose blood levels and insulin independence after the transplantation. However, the mean blood levels of glycemia were significantly lower in transplanted rats compared with diabetic rats (470.4 mg/dl vs 605 mg/dl, p 0.04). Interestingly, transplanted rats have a significant weight increase after transplantation compared to diabetic rats (mean value 295 g in transplanted rats vs 245 g in diabetic rats, p < 0.05), with an overall improvement of social activities and health. Immunohistochemical analysis of the 5 mesenteric lymph nodes of transplanted rats demonstrated the presence of living islets in one lymph node. Conclusions Although islet engraftment in lymph nodes is possible, islet transplantation in lymph nodes in rats resulted in few improvements of glucose parameters.
Collapse
Affiliation(s)
- Massimiliano Veroux
- Vascular Surgery and Organ Transplant Unit, Department of Medical and Surgical Sciences, University Hospital of Catania, Via Santa Sofia, 84 95123, Catania, Italy.
| | - Rita Bottino
- Institute of Cellular Therapeutics, Allegheny-Singer Research Institute, Allegheny Health Network, Pittsburgh, PA, USA
| | - Roberta Santini
- Vascular Surgery and Organ Transplant Unit, Department of Medical and Surgical Sciences, University Hospital of Catania, Via Santa Sofia, 84 95123, Catania, Italy
| | - Suzanne Bertera
- Institute of Cellular Therapeutics, Allegheny-Singer Research Institute, Allegheny Health Network, Pittsburgh, PA, USA
| | - Daniela Corona
- Vascular Surgery and Organ Transplant Unit, Department of Medical and Surgical Sciences, University Hospital of Catania, Via Santa Sofia, 84 95123, Catania, Italy
| | - Domenico Zerbo
- Vascular Surgery and Organ Transplant Unit, Department of Medical and Surgical Sciences, University Hospital of Catania, Via Santa Sofia, 84 95123, Catania, Italy
| | - Giovanni Li Volti
- Department of Biomedical and Biotechnological Sciences, Section of Medical Biochemistry, University of Catania, Catania, Italy
| | - Burcin Ekser
- Transplant Division, Department of Surgery, Indiana University School of Medicine, Indianapolis, USA
| | - Lidia Puzzo
- Section of Anatomic Pathology, Department od Medical and Surgical Sciences, and Advanced Technologies, University Hospital of Catania, Catania, Italy
| | - Marco Raffaele
- Department of Drug Science, Biochemistry Section, University of Catania, Catania, Italy
| | | | - Alessia Giaquinta
- Vascular Surgery and Organ Transplant Unit, Department of Medical and Surgical Sciences, University Hospital of Catania, Via Santa Sofia, 84 95123, Catania, Italy
| | - Pierfrancesco Veroux
- Vascular Surgery and Organ Transplant Unit, Department of Medical and Surgical Sciences, University Hospital of Catania, Via Santa Sofia, 84 95123, Catania, Italy
| | - Luca Vanella
- Department of Drug Science, Biochemistry Section, University of Catania, Catania, Italy
| |
Collapse
|
21
|
Rickels MR, Robertson RP. Pancreatic Islet Transplantation in Humans: Recent Progress and Future Directions. Endocr Rev 2019; 40:631-668. [PMID: 30541144 PMCID: PMC6424003 DOI: 10.1210/er.2018-00154] [Citation(s) in RCA: 178] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 10/26/2018] [Indexed: 12/11/2022]
Abstract
Pancreatic islet transplantation has become an established approach to β-cell replacement therapy for the treatment of insulin-deficient diabetes. Recent progress in techniques for islet isolation, islet culture, and peritransplant management of the islet transplant recipient has resulted in substantial improvements in metabolic and safety outcomes for patients. For patients requiring total or subtotal pancreatectomy for benign disease of the pancreas, isolation of islets from the diseased pancreas with intrahepatic transplantation of autologous islets can prevent or ameliorate postsurgical diabetes, and for patients previously experiencing painful recurrent acute or chronic pancreatitis, quality of life is substantially improved. For patients with type 1 diabetes or insulin-deficient forms of pancreatogenic (type 3c) diabetes, isolation of islets from a deceased donor pancreas with intrahepatic transplantation of allogeneic islets can ameliorate problematic hypoglycemia, stabilize glycemic lability, and maintain on-target glycemic control, consequently with improved quality of life, and often without the requirement for insulin therapy. Because the metabolic benefits are dependent on the numbers of islets transplanted that survive engraftment, recipients of autoislets are limited to receive the number of islets isolated from their own pancreas, whereas recipients of alloislets may receive islets isolated from more than one donor pancreas. The development of alternative sources of islet cells for transplantation, whether from autologous, allogeneic, or xenogeneic tissues, is an active area of investigation that promises to expand access and indications for islet transplantation in the future treatment of diabetes.
Collapse
Affiliation(s)
- Michael R Rickels
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - R Paul Robertson
- Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, Washington
- Division of Endocrinology, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
- Pacific Northwest Diabetes Research Institute, Seattle, Washington
| |
Collapse
|
22
|
|
23
|
Anazawa T, Okajima H, Masui T, Uemoto S. Current state and future evolution of pancreatic islet transplantation. Ann Gastroenterol Surg 2019; 3:34-42. [PMID: 30697608 PMCID: PMC6345654 DOI: 10.1002/ags3.12214] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 08/12/2018] [Accepted: 08/27/2018] [Indexed: 12/17/2022] Open
Abstract
Pancreatic islet transplantation provides an effective treatment option for patients with type 1 diabetes (T1D) with intractable impaired awareness of hypoglycemia and severe hypoglycemic events. Currently, the primary goal of islet transplantation should be excellent glycemic control without severe hypoglycemia, rather than insulin independence. Islet transplant recipients were less likely to achieve insulin independence, whereas solid pancreas transplant recipients substantially had greater procedure-related morbidity. Excellent therapeutic effects of islet transplantation as a result of accurate blood glucose level-reactive insulin secretion, which cannot be reproduced by current drug therapy, have been confirmed. Recent improvement of islet transplantation outcome has been achieved by refinement of the pancreatic islet isolation technique, improvement of islet engraftment method, and introduction of effective immunosuppressive therapy. A disadvantage of islet transplantation is that donors are essential, and donor shortage has become a hindrance to its development. With the development of alternative transplantation sites and new cell sources, including porcine islet cells and embryonic stem/induced pluripotent stem (ES/iPS)-derived β cells, "On-demand" and "Unlimited" cell therapy for T1D can be established.
Collapse
Affiliation(s)
- Takayuki Anazawa
- Division of Hepato‐Biliary‐Pancreatic Surgery and TransplantationDepartment of SurgeryGraduate School of MedicineUniversity of KyotoKyotoJapan
| | - Hideaki Okajima
- Division of Hepato‐Biliary‐Pancreatic Surgery and TransplantationDepartment of SurgeryGraduate School of MedicineUniversity of KyotoKyotoJapan
| | - Toshihiko Masui
- Division of Hepato‐Biliary‐Pancreatic Surgery and TransplantationDepartment of SurgeryGraduate School of MedicineUniversity of KyotoKyotoJapan
| | - Shinji Uemoto
- Division of Hepato‐Biliary‐Pancreatic Surgery and TransplantationDepartment of SurgeryGraduate School of MedicineUniversity of KyotoKyotoJapan
| |
Collapse
|
24
|
Gan J, Wang Y, Zhou X. Stem cell transplantation for the treatment of patients with type 1 diabetes mellitus: A meta-analysis. Exp Ther Med 2018; 16:4479-4492. [PMID: 30542397 PMCID: PMC6257425 DOI: 10.3892/etm.2018.6769] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 08/31/2018] [Indexed: 12/28/2022] Open
Abstract
The efficacy of stem cell (SC) transplantation in patients with type 1 diabetes mellitus (T1DM) has remained to be fully elucidated. In the present study, a systematic review and meta-analysis was performed to determine the clinical outcomes. Electronic databases, including PubMed, MEDLINE, WanFang and the Cochrane Library were screened for relevant studies published until January 13, 2018. The references of retrieved papers, systematic reviews and trial registries were manually screened for additional papers. Two authors were involved in screening the titles in order to select eligible studies, extract data and assess the risk of bias. Studies were pooled using a random-effects model as well as the Begg's funnel plot and subgroup analysis was performed using Stata 14.0 software. A total of 47 studies were retrieved for detailed evaluation, of which 22 met the inclusion criteria. No substantial publication bias was identified. The meta-analysis revealed that SC therapy increased C-peptide levels when compared with placebo treatment in randomized-controlled trials [RCT; standardized mean difference (SMD), 0.93; 95% confidence interval (CI) 0.23-1.63] and self-controlled trials (SMD, 0.66; 95% CI, -0.22 to 1.54). An analysis demonstrated that SC therapy was more efficient at reducing the glycated hemoglobin level compared with the control group in RCTs (SMD, 0.56; 95% CI; 0.06-1.06; and SMD, 1.63; 95% CI, 0.92-2.34, respectively). The graphs demonstrated that SC transplantation resulted in a reduction of insulin requirement. Furthermore, subgroup analyses revealed that patient age, medical history and the SC injection dose may be sources of the heterogeneity observed. The greatest benefit of SC transplantation was seen in patients aged ≥18 years or a medical history of <3 months. In addition, the SC injection dose of ≥107 IU/kg/day was more effective than <107 IU/kg/day when the cellular composition included mesenchymal SCs and hematopoietic SCs. In conclusion, SC therapy represents an efficient option for patients with T1DM. This systematic review was registered at the International prospective register of systematic reviews (no. 42018093930).
Collapse
Affiliation(s)
- Jiadi Gan
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Department of Clinical Medicine, The First Clinical Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yingjin Wang
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
- Department of Clinical Medicine, The Second Clinical Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xiaodong Zhou
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
25
|
Anti-Inflammatory Strategies in Intrahepatic Islet Transplantation: A Comparative Study in Preclinical Models. Transplantation 2018; 102:240-248. [PMID: 28902069 DOI: 10.1097/tp.0000000000001925] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND The identification of pathway(s) playing a pivotal role in peritransplant detrimental inflammatory events represents the crucial step toward a better management and outcome of pancreatic islet transplanted patients. Recently, we selected the CXCR1/2 inhibition as a relevant strategy in enhancing pancreatic islet survival after transplantation. METHODS Here, the most clinically used anti-inflammatory compounds (IL1-receptor antagonist, steroids, and TNF-α inhibitor) alone or in combination with a CXCR1/2 inhibitor were evaluated in their ability to improve engraftment or delay graft rejection. To rule out bias related to transplantation site, we used well-established preclinical syngeneic (250 C57BL/6 equivalent islets in C57BL/6) and allogeneic (400 Balb/c equivalent islets in C57BL6) intrahepatic islet transplantation platforms. RESULTS In mice, we confirmed that targeting the CXCR1/2 pathway is crucial in preserving islet function and improving engraftment. In the allogeneic setting, CXCR1/2 inhibitor alone could reduce the overall recruitment of transplant-induced leukocytes and significantly prolong the time to graft rejection both as a single agent and in combination with immunosuppression. No other anti-inflammatory compounds tested (IL1-receptor antagonist, steroids, and TNF-α inhibitor) alone or in combination with CXCR1/2 inhibitor improve islet engraftment and significantly delay graft rejection in the presence of MMF + FK-506 immunosuppressive treatment. CONCLUSIONS These findings indicate that only the CXCR1/2-mediated axis plays a crucial role in controlling the islet damage and should be a target for intervention to improve the efficiency of islet transplantation.
Collapse
|
26
|
Anti-LFA-1 induces CD8 T-cell dependent allograft tolerance and augments suppressor phenotype CD8 cells. Cell Immunol 2018; 332:101-110. [PMID: 30103941 DOI: 10.1016/j.cellimm.2018.08.003] [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: 05/25/2018] [Revised: 07/30/2018] [Accepted: 08/06/2018] [Indexed: 11/22/2022]
Abstract
The induction of tolerance to transplanted organs is a major objective in transplantation immunology research. Lymphocyte function-associated antigen-1 (LFA-1) interactions have been identified as a key component of the T-cell activation process that may be interrupted to lead to allograft tolerance. In mice, αLFA-1 mAb is a potent monotherapy that leads to the induction of donor-specific transferable tolerance. By interrogating important adaptive and innate immunity pathways, we demonstrate that the induction of tolerance relies on CD8+T-cells. We further demonstrate that αLFA-1 induced tolerance is associated with CD8+CD28-T-cells with a suppressor phenotype, and that while CD8 cells are present, the effector T-cell response is abrogated. A recent publication has shown that CD8+CD28- cells are not diminished by cyclosporine or rapamycin, therefore CD8+CD28- cells represent a clinically relevant population. To our knowledge, this is the first time that a mechanism for αLFA-1 induced tolerance has been described.
Collapse
|
27
|
|
28
|
Odorico J, Markmann J, Melton D, Greenstein J, Hwa A, Nostro C, Rezania A, Oberholzer J, Pipeleers D, Yang L, Cowan C, Huangfu D, Egli D, Ben-David U, Vallier L, Grey ST, Tang Q, Roep B, Ricordi C, Naji A, Orlando G, Anderson DG, Poznansky M, Ludwig B, Tomei A, Greiner DL, Graham M, Carpenter M, Migliaccio G, D'Amour K, Hering B, Piemonti L, Berney T, Rickels M, Kay T, Adams A. Report of the Key Opinion Leaders Meeting on Stem Cell-derived Beta Cells. Transplantation 2018; 102:1223-1229. [PMID: 29781950 PMCID: PMC6775764 DOI: 10.1097/tp.0000000000002217] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Beta cell replacement has the potential to restore euglycemia in patients with insulin-dependent diabetes. Although great progress has been made in establishing allogeneic islet transplantation from deceased donors as the standard of care for those with the most labile diabetes, it is also clear that the deceased donor organ supply cannot possibly treat all those who could benefit from restoration of a normal beta cell mass, especially if immunosuppression were not required. Against this background, the International Pancreas and Islet Transplant Association in collaboration with the Harvard Stem Cell Institute, the Juvenile Diabetes Research Foundation (JDRF), and the Helmsley Foundation held a 2-day Key Opinion Leaders Meeting in Boston in 2016 to bring together experts in generating and transplanting beta cells derived from stem cells. The following summary highlights current technology, recent significant breakthroughs, unmet needs and roadblocks to stem cell-derived beta cell therapies, with the aim of spurring future preclinical collaborative investigations and progress toward the clinical application of stem cell-derived beta cells.
Collapse
Affiliation(s)
- Jon Odorico
- Division of Transplantation, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - James Markmann
- Department of Surgery, Massachusetts General Hospital, Boston, MA
| | - Douglas Melton
- Harvard Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Boston MA
| | | | - Albert Hwa
- Joslin Diabetes Center, Harvard Medical School, Boston, MA
| | - Cristina Nostro
- Department of Physiology, University of Toronto, University of Toronto, Toronto Canada
| | | | - Jose Oberholzer
- Department of Surgery, University of Illinois at Chicago, Chicago, IL
| | - Daniel Pipeleers
- Center for Beta Cell Therapy in Diabetes, Vrije Universiteit Brussel, Brussels, Belgium
| | | | - Chad Cowan
- Harvard Stem Cell Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Danwei Huangfu
- Developmental Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Dieter Egli
- Columbia Stem Cell Initiative, Columbia University, New York, NY
| | - Uri Ben-David
- Broad Institute of MIT and Harvard, Cancer Program, Golub Lab, Cambridge MA
| | - Ludovic Vallier
- Department of Surgery, University of Cambridge, Cambridge, United Kingdom
| | - Shane T Grey
- Department of Medicine, University of Sydney, Sydney, Australia
| | - Qizhi Tang
- Department of Surgery, UCSF Medical Center, San Francisco, CA
| | - Bart Roep
- National Diabetes Center of Excellence, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Ali Naji
- Department of Surgery, University of Pennsylvania, Philadelphia, PA
| | - Giuseppe Orlando
- Center on Diabetes, Obesity, and Metabolism, Wake Forest School of Medicine, Winston-Salem, NC
| | - Daniel G Anderson
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA
| | - Mark Poznansky
- Department of Medicine, Vaccine and Immunotherapy Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Barbara Ludwig
- Department of Endocrinology and Diabetes, University Hospital Dresden, Dresden, Germany
| | - Alice Tomei
- Department of Surgery, University of Miami, Miami, FL
| | - Dale L Greiner
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Melanie Graham
- Department of Surgery, University of Minnesota, Minneapolis, MN
| | | | | | | | - Bernhard Hering
- Department of Surgery, University of Minnesota, Minneapolis, MN
| | - Lorenzo Piemonti
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan Italy
| | - Thierry Berney
- Department of Surgery, Geneva University, Geneva, Switzerland
| | - Mike Rickels
- Department of Surgery, University of Pennsylvania, Philadelphia, PA
| | - Thomas Kay
- Department of Medicine, St. Vincent's Institute, Melbourne, Australia
| | - Ann Adams
- Department of Surgery, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
29
|
Wszola M, Berman A, Ostaszewska A, Gorski L, Serwanska-Swietek M, Gozdowska J, Bednarska K, Krajewska M, Lipinska A, Chmura A, Kwiatkowski A. Islets Allotransplantation Into Gastric Submucosa in a Patient with Portal Hypertension: 4-year Follow-up. Transplant Proc 2018; 50:1910-1913. [PMID: 30056927 DOI: 10.1016/j.transproceed.2018.02.170] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Accepted: 02/06/2018] [Indexed: 12/15/2022]
Abstract
BACKGROUND Islets transplantation is an established treatment method for patients suffering from brittle diabetes with hypoglycemia unawareness. The standard implantation technique is through the portal vein into the liver. In case of liver diseases or portal hypertension, finding an extra-hepatic site is recommended. There have been attempts to perform islets transplantations into muscles and into the gastric submucosa. OBJECTIVE The aim of this study is to show a 4-year follow-up of allotransplantation into gastric submucosa in a case of portal hypertension observed during the procedure of islets infusion. PATIENTS AND METHODS A 36-year-old woman with complicated diabetes for over 30 years was selected to receive simultaneous islets and kidney transplantation. The patient underwent an unsuccessful simultaneous pancreas and kidney transplantation 2 years earlier in another transplantation center. The patient's daily insulin requirement was 60 IU, which corresponded to 1.15 IU/kg of body weight. The HbA1c level was 7.4%. C-peptide levels, both fasting and stimulated, were 0.01 ng/mL. On December 7, 2013, the patient received transplanted kidney and islets procured from the same donor. Only 124,000 islets equivalents (IEQ) were isolated (2400 IEQ/kg body weight). Islets were suspended in 300 mL of Ringer's solution along with albumin, antibiotics, and heparin. After infusing 100 mL of the islets suspension into the portal vein, pressure in portal vein increased from 5 mm Hg to 23 mm Hg. Despite stopping the infusion, pressure did not drop after 30 minutes. The decision was made to transplant the reminder of the islets (200 mL) into the gastric wall. RESULTS No complications were observed after the procedure. Serum creatinine level was 1.6 mg/dL on day 10 and 1.5 mg/dL 4 years after the transplantation. Fasting C-peptide levels were 1.7, 0.65, 0.55, 0.69, 0.68, and 0.2 ng/mL at 1, 3, 6, 12, 18, and 36 months after the transplantation, respectively. HbA1c levels were 5.2, 6.4, 4.7, 5.2, and 5.9% at 3, 6, 12, 18, and 36 months, respectively. The patient's insulin requirement dropped to 15 U/day immediately after transplantation and equaled 20 and 27 U/day at 18 and 48 months after the simultaneous islet and kidney transplantation, respectively. CONCLUSION Allotransplantation of islets into the gastric wall may be a safe alternative in cases of contraindications for transplantation into the portal vein.
Collapse
Affiliation(s)
- M Wszola
- Foundation of Research and Science Development, Otwock, Poland.
| | - A Berman
- Foundation of Research and Science Development, Otwock, Poland
| | - A Ostaszewska
- Department of General and Transplantation Surgery, Medical University of Warsaw, Warsaw, Poland
| | - L Gorski
- Department of General and Transplantation Surgery, Medical University of Warsaw, Warsaw, Poland
| | - M Serwanska-Swietek
- Foundation of Research and Science Development, Otwock, Poland; Department of General and Transplantation Surgery, Medical University of Warsaw, Warsaw, Poland
| | - J Gozdowska
- Department of Transplantation Medicine and Nephrology, Transplantation Institute, Medical University of Warsaw, Warsaw, Poland
| | - K Bednarska
- Foundation of Research and Science Development, Otwock, Poland
| | - M Krajewska
- Department of Immunology, Transplantology and Internal Diseases, Medical University of Warsaw, Warsaw, Poland
| | - A Lipinska
- Department of Internal Medicine and Cardiology, Medical University of Warsaw, Warsaw, Poland
| | - A Chmura
- Department of General and Transplantation Surgery, Medical University of Warsaw, Warsaw, Poland
| | - A Kwiatkowski
- Foundation of Research and Science Development, Otwock, Poland; Department of General and Transplantation Surgery, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
30
|
Burrack AL, Landry LG, Siebert J, Coulombe M, Gill RG, Nakayama M. Simultaneous Recognition of Allogeneic MHC and Cognate Autoantigen by Autoreactive T Cells in Transplant Rejection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 200:1504-1512. [PMID: 29311365 PMCID: PMC5809255 DOI: 10.4049/jimmunol.1700856] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 12/10/2017] [Indexed: 12/15/2022]
Abstract
The autoimmune condition is a primary obstacle to inducing tolerance in type 1 diabetes patients receiving allogeneic pancreas transplants. It is unknown how autoreactive T cells that recognize self-MHC molecules contribute to MHC-disparate allograft rejection. In this report, we show the presence and accumulation of dual-reactive, that is autoreactive and alloreactive, T cells in C3H islet allografts that were transplanted into autoimmune diabetic NOD mice. Using high-throughput sequencing, we discovered that T cells prevalent in allografts share identical TCRs with autoreactive T cells present in pancreatic islets. T cells expressing TCRs that are enriched in allograft lesions recognized C3H MHC molecules, and five of six cell lines expressing these TCRs were also reactive to NOD islet cells. These results reveal the presence of autoreactive T cells that mediate cross-reactive alloreactivity, and indicate a requirement for regulating such dual-reactive T cells in tissue replacement therapies given to autoimmune individuals.
Collapse
Affiliation(s)
- Adam L Burrack
- Department of Surgery, University of Colorado School of Medicine, Aurora, CO 80045
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045
| | - Laurie G Landry
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora, CO 80045; and
| | | | - Marilyne Coulombe
- Department of Surgery, University of Colorado School of Medicine, Aurora, CO 80045
| | - Ronald G Gill
- Department of Surgery, University of Colorado School of Medicine, Aurora, CO 80045
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045
| | - Maki Nakayama
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045;
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora, CO 80045; and
| |
Collapse
|
31
|
Abstract
PURPOSE OF REVIEW Transplantation of allogenic pancreatic islets is a minimally invasive treatment option to control severe hypoglycemia and dependence on exogenous insulin among type 1 diabetes (T1D) patients. This overview summarizes the current issues and progress in islet transplantation outcomes and research. RECENT FINDINGS Several clinical trials from North America and other countries have documented the safety and efficacy of clinical islet transplantation for T1D patients with impaired hypoglycemia awareness. A recently completed phase 3 clinical trial allows centres in the United States to apply for a Food and Drug Administration Biologics License for the procedure. Introduction of anti-inflammatory drugs along with T-cell depleting induction therapy has significantly improved long-term function of transplanted islets. Research into islet biomarkers, immunosuppression, extrahepatic transplant sites and potential alternative beta cell sources is driving further progress. SUMMARY Allogeneic islet transplantation has vastly improved over the past two decades. Success in restoration of glycemic control and hypoglycemic awareness after islet transplantation has been further highlighted by clinical trials. However, lack of effective strategies to maintain long-term islet function and insufficient sources of donor tissue still impose limitations to the widespread use of islet transplantation. In the United States, wide adoption of this technology still awaits regulatory approval and, importantly, a financial mechanism to support the use of this technology.
Collapse
|
32
|
Current outcomes in islet versus solid organ pancreas transplant for β-cell replacement in type 1 diabetes. Curr Opin Organ Transplant 2017; 21:399-404. [PMID: 27258578 DOI: 10.1097/mot.0000000000000332] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW With continued optimization of islet isolation and immunosuppression protocols, the medium-term rates of insulin independence following islet transplantation have improved significantly. This review evaluates the most up-to-date outcomes data for both solid organ pancreas and islet transplantation to develop an algorithm for selection of β-cell replacement in type 1 diabetes patients. RECENT FINDINGS Solid organ pancreas and islet transplantation have both displayed improved rates of 5-year insulin independence, largely attributable to improvements in immunosuppressive regimens. The medium-term rates of insulin independence following islet transplantation in highly selected type 1 nonuremic diabetic recipients is beginning to approach the success rates observed following solitary pancreas transplantation. SUMMARY Although pancreas transplantation has historically been favored for β-cell replacement, current outcomes following islet transplantation justify the use of this minimally invasive therapy in carefully selected patients. Pancreas transplant remains the procedure of choice for β-cell replacement in uremic patients. Islet transplantation should be considered in nonuremic patients with low BMI and low insulin requirements, patients lacking the cardiovascular reserve to undergo open abdominal surgery, or patients who elect to forego the risks of a major operation in exchange for an increased risk of islet graft failure.
Collapse
|
33
|
Tissue adhesive FK506-loaded polymeric nanoparticles for multi-layered nano-shielding of pancreatic islets to enhance xenograft survival in a diabetic mouse model. Biomaterials 2017; 154:182-196. [PMID: 29128846 DOI: 10.1016/j.biomaterials.2017.10.049] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 10/25/2017] [Accepted: 10/30/2017] [Indexed: 12/15/2022]
Abstract
This study aims to develop a novel surface modification technology to prolong the survival time of pancreatic islets in a xenogenic transplantation model, using 3,4-dihydroxyphenethylamine (DOPA) conjugated poly(lactide-co-glycolide)-poly(ethylene glycol) (PLGA-PEG) nanoparticles (DOPA-NPs) carrying immunosuppressant FK506 (FK506/DOPA-NPs). The functionalized DOPA-NPs formed a versatile coating layer for antigen camouflage without interfering the viability and functionality of islets. The coating layer effectively preserved the morphology and viability of islets in a co-culture condition with xenogenic lymphocytes for 7 days. Interestingly, the mean survival time of islets coated with FK506/DOPA-NPs was significantly higher as compared with that of islets coated with DOPA-NPs (without FK506) and control. This study demonstrated that the combination of surface camouflage and localized low dose of immunosuppressant could be an effective approach in prolonging the survival of transplanted islets. This newly developed platform might be useful for immobilizing various types of small molecules on therapeutic cells and biomaterial surface to improve the therapeutic efficacy in cell therapy and regenerative medicine.
Collapse
|
34
|
Wisel SA, Gardner JM, Roll GR, Harbell J, Freise CE, Feng S, Kang SM, Hirose R, Kaufman DB, Posselt A, Stock PG. Pancreas-After-Islet Transplantation in Nonuremic Type 1 Diabetes: A Strategy for Restoring Durable Insulin Independence. Am J Transplant 2017; 17:2444-2450. [PMID: 28489277 PMCID: PMC5573612 DOI: 10.1111/ajt.14344] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Revised: 04/21/2017] [Accepted: 04/22/2017] [Indexed: 01/25/2023]
Abstract
Islet transplantation offers a minimally invasive approach for β cell replacement in diabetic patients with hypoglycemic unawareness. Attempts at insulin independence may require multiple islet reinfusions from distinct donors, increasing the risk of allogeneic sensitization. Currently, solid organ pancreas transplant is the only remaining surgical option following failed islet transplantation in the United States; however, the immunologic impact of repeated exposure to donor antigens on subsequent pancreas transplantation is unclear. We describe a case series of seven patients undergoing solid organ pancreas transplant following islet graft failure with long-term follow-up of pancreatic graft survival and renal function. Despite highly variable panel reactive antibody levels prior to pancreas transplant (mean 27 ± 35%), all seven patients achieved stable and durable insulin independence with a mean follow-up of 6.7 years. Mean hemoglobin A1c values improved significantly from postislet, prepancreas levels (mean 8.1 ± 1.5%) to postpancreas levels (mean 5.3 ± 0.1%; p = 0.0022). Three patients experienced acute rejection episodes that were successfully managed with thymoglobulin and methylprednisolone, and none of these preuremic type 1 diabetic recipients developed stage 4 or 5 chronic kidney disease postoperatively. These results support pancreas-after-islet transplantation with aggressive immunosuppression and protocol biopsies as a viable strategy to restore insulin independence after islet graft failure.
Collapse
Affiliation(s)
- SA Wisel
- Department of Surgery, University of California, San Francisco; San Francisco, CA, United States
| | - JM Gardner
- Department of Surgery, University of California, San Francisco; San Francisco, CA, United States
| | - GR Roll
- Department of Surgery, University of California, San Francisco; San Francisco, CA, United States
| | - J Harbell
- Department of Surgery, University of California, San Francisco; San Francisco, CA, United States
| | - CE Freise
- Department of Surgery, University of California, San Francisco; San Francisco, CA, United States
| | - S Feng
- Department of Surgery, University of California, San Francisco; San Francisco, CA, United States
| | - SM Kang
- Department of Surgery, University of California, San Francisco; San Francisco, CA, United States
| | - R Hirose
- Department of Surgery, University of California, San Francisco; San Francisco, CA, United States
| | - DB Kaufman
- Department of Surgery, University of Wisconsin; Madison, WI, United States
| | - A Posselt
- Department of Surgery, University of California, San Francisco; San Francisco, CA, United States
| | - PG Stock
- Department of Surgery, University of California, San Francisco; San Francisco, CA, United States
| |
Collapse
|
35
|
Chang R, Faleo G, Russ HA, Parent AV, Elledge SK, Bernards DA, Allen JL, Villanueva K, Hebrok M, Tang Q, Desai TA. Nanoporous Immunoprotective Device for Stem-Cell-Derived β-Cell Replacement Therapy. ACS NANO 2017; 11:7747-7757. [PMID: 28763191 PMCID: PMC5667644 DOI: 10.1021/acsnano.7b01239] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Encapsulation of human embryonic stem-cell-differentiated beta cell clusters (hES-βC) holds great promise for cell replacement therapy for the treatment of diabetics without the need for chronic systemic immune suppression. Here, we demonstrate a nanoporous immunoprotective polymer thin film cell encapsulation device that can exclude immune molecules while allowing exchange of oxygen and nutrients necessary for in vitro and in vivo stem cell viability and function. Biocompatibility studies show the device promotes neovascular formation with limited foreign body response in vivo. The device also successfully prevented teratoma escape into the peritoneal cavity of mice. Long-term animal studies demonstrate evidence of engraftment, viability, and function of cells encapsulated in the device after 6 months. Finally, in vivo study confirms that the device was able to effectively immuno-isolate cells from the host immune system.
Collapse
Affiliation(s)
- Ryan Chang
- UCSF-UC Berkeley Joint PhD Program in Bioengineering, San Francisco, California 94143, United States
| | - Gaetano Faleo
- Department of Surgery, University of California, San Francisco, San Francisco, California 94143, United States
| | - Holger A. Russ
- Diabetes Center, University of California, San Francisco, San Francisco, California 94143, United States
| | - Audrey V. Parent
- Diabetes Center, University of California, San Francisco, San Francisco, California 94143, United States
| | - Susanna K. Elledge
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California 94143, United States
| | - Daniel A. Bernards
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California 94143, United States
| | - Jessica L. Allen
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California 94143, United States
| | - Karina Villanueva
- Diabetes Center, University of California, San Francisco, San Francisco, California 94143, United States
| | - Matthias Hebrok
- Diabetes Center, University of California, San Francisco, San Francisco, California 94143, United States
| | - Qizhi Tang
- Department of Surgery, University of California, San Francisco, San Francisco, California 94143, United States
- Diabetes Center, University of California, San Francisco, San Francisco, California 94143, United States
| | - Tejal A. Desai
- UCSF-UC Berkeley Joint PhD Program in Bioengineering, San Francisco, California 94143, United States
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California 94143, United States
| |
Collapse
|
36
|
Samy KP, Anderson DA, Lo DJ, Mulvihill MS, Song M, Farris AB, Parker BS, MacDonald AL, Lu C, Springer TA, Kachlany SC, Reimann KA, How T, Leopardi FV, Franke KS, Williams KD, Collins BH, Kirk AD. Selective Targeting of High-Affinity LFA-1 Does Not Augment Costimulation Blockade in a Nonhuman Primate Renal Transplantation Model. Am J Transplant 2017; 17:1193-1203. [PMID: 27888551 PMCID: PMC5409867 DOI: 10.1111/ajt.14141] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2016] [Revised: 11/01/2016] [Accepted: 11/08/2016] [Indexed: 01/25/2023]
Abstract
Costimulation blockade (CoB) via belatacept is a lower-morbidity alternative to calcineurin inhibitor (CNI)-based immunosuppression. However, it has higher rates of early acute rejection. These early rejections are mediated in part by memory T cells, which have reduced dependence on the pathway targeted by belatacept and increased adhesion molecule expression. One such molecule is leukocyte function antigen (LFA)-1. LFA-1 exists in two forms: a commonly expressed, low-affinity form and a transient, high-affinity form, expressed only during activation. We have shown that antibodies reactive with LFA-1 regardless of its configuration are effective in eliminating memory T cells but at the cost of impaired protective immunity. Here we test two novel agents, leukotoxin A and AL-579, each of which targets the high-affinity form of LFA-1, to determine whether this more precise targeting prevents belatacept-resistant rejection. Despite evidence of ex vivo and in vivo ligand-specific activity, neither agent when combined with belatacept proved superior to belatacept monotherapy. Leukotoxin A approached a ceiling of toxicity before efficacy, while AL-579 failed to significantly alter the peripheral immune response. These data, and prior studies, suggest that LFA-1 blockade may not be a suitable adjuvant agent for CoB-resistant rejection.
Collapse
Affiliation(s)
- KP Samy
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710
| | - DA Anderson
- Emory Transplant Center, Emory University School of Medicine, Atlanta, GA 30322
| | - DJ Lo
- Emory Transplant Center, Emory University School of Medicine, Atlanta, GA 30322
| | - MS Mulvihill
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710
| | - M Song
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710
| | - AB Farris
- Department of Pathology & Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322
| | - BS Parker
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710
| | - AL MacDonald
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710
| | - C Lu
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115
| | - TA Springer
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115
| | - SC Kachlany
- Rutgers University, School of Medicine, Newark, NJ 07103,Actinobac Biomed, Inc., Kendall Park, NJ 08824
| | - KA Reimann
- Mass-Biologics, University of Massachusetts Medical School, Boston, MA 02126
| | - T How
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710
| | - FV Leopardi
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710
| | - KS Franke
- Division of Laboratory Animal Resources, Duke University, Durham, NC 27710
| | - KD Williams
- Division of Laboratory Animal Resources, Duke University, Durham, NC 27710
| | - BH Collins
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710
| | - AD Kirk
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710,Emory Transplant Center, Emory University School of Medicine, Atlanta, GA 30322
| |
Collapse
|
37
|
Govender L, Wyss JC, Kumar R, Pascual M, Golshayan D. IL-2-Mediated In Vivo Expansion of Regulatory T Cells Combined with CD154-CD40 Co-Stimulation Blockade but Not CTLA-4 Ig Prolongs Allograft Survival in Naive and Sensitized Mice. Front Immunol 2017; 8:421. [PMID: 28484450 PMCID: PMC5399033 DOI: 10.3389/fimmu.2017.00421] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Accepted: 03/24/2017] [Indexed: 01/03/2023] Open
Abstract
In recent years, regulatory T cells (Treg)-based immunotherapy has emerged as a promising strategy to promote operational tolerance after solid organ transplantation (SOT). However, a main hurdle for the therapeutic use of Treg in transplantation is their low frequency, particularly in non-lymphopenic hosts. We aimed to expand Treg directly in vivo and determine their efficacy in promoting donor-specific tolerance, using a stringent experimental model. Administration of the IL-2/JES6-1 immune complex at the time of transplantation resulted in significant expansion of donor-specific Treg, which suppressed alloreactive T cells. IL-2-mediated Treg expansion in combination with short-term CD154–CD40 co-stimulation blockade, but not CTLA-4 Ig or rapamycin, led to tolerance to MHC-mismatched skin grafts in non-lymphopenic mice, mainly by hindering alloreactive CD8+ effector T cells and the production of alloantibodies. Importantly, this treatment also allowed prolonged survival of allografts in the presence of either donor-specific or cross-reactive memory cells. However, late rejection occurred in sensitized hosts, partly mediated by activated B cells. Overall, these data illustrate the potential but also some important limitations of Treg-based therapy in clinical SOT as well as the importance of concomitant immunomodulatory strategies in particular in sensitized hosts.
Collapse
Affiliation(s)
- Lerisa Govender
- Transplantation Centre and Transplantation Immunopathology Laboratory, Service of Immunology and Allergy, Department of Medicine, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Jean-Christophe Wyss
- Transplantation Centre and Transplantation Immunopathology Laboratory, Service of Immunology and Allergy, Department of Medicine, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Rajesh Kumar
- Transplantation Centre and Transplantation Immunopathology Laboratory, Service of Immunology and Allergy, Department of Medicine, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Manuel Pascual
- Transplantation Centre and Transplantation Immunopathology Laboratory, Service of Immunology and Allergy, Department of Medicine, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Dela Golshayan
- Transplantation Centre and Transplantation Immunopathology Laboratory, Service of Immunology and Allergy, Department of Medicine, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
38
|
Kean LS, Turka LA, Blazar BR. Advances in targeting co-inhibitory and co-stimulatory pathways in transplantation settings: the Yin to the Yang of cancer immunotherapy. Immunol Rev 2017; 276:192-212. [PMID: 28258702 PMCID: PMC5338458 DOI: 10.1111/imr.12523] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In the past decade, the power of harnessing T-cell co-signaling pathways has become increasingly understood to have significant clinical importance. In cancer immunotherapy, the field has concentrated on two related modalities: First, targeting cancer antigens through highly activated chimeric antigen T cells (CAR-Ts) and second, re-animating endogenous quiescent T cells through checkpoint blockade. In each of these strategies, the therapeutic goal is to re-ignite T-cell immunity, in order to eradicate tumors. In transplantation, there is also great interest in targeting T-cell co-signaling, but with the opposite goal: in this field, we seek the Yin to cancer immunotherapy's Yang, and focus on manipulating T-cell co-signaling to induce tolerance rather than activation. In this review, we discuss the major T-cell signaling pathways that are being investigated for tolerance induction, detailing preclinical studies and the path to the clinic for many of these molecules. These include blockade of co-stimulation pathways and agonism of coinhibitory pathways, in order to achieve the delicate state of balance that is transplant tolerance: a state which guarantees lifelong transplant acceptance without ongoing immunosuppression, and with preservation of protective immune responses. In the context of the clinical translation of immune tolerance strategies, we discuss the significant challenge that is embodied by the fact that targeted pathway modulators may have opposing effects on tolerance based on their impact on effector vs regulatory T-cell biology. Achieving this delicate balance holds the key to the major challenge of transplantation: lifelong control of alloreactivity while maintaining an otherwise intact immune system.
Collapse
Affiliation(s)
- Leslie S Kean
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, WA, USA
- The Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - Laurence A Turka
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Immune Tolerance Network, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Bruce R Blazar
- Division of Blood and Marrow Transplantation, Department of Pediatrics and the Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
39
|
Itoh T, Nishinakamura H, Kumano K, Takahashi H, Kodama S. The Spleen Is an Ideal Site for Inducing Transplanted Islet Graft Expansion in Mice. PLoS One 2017; 12:e0170899. [PMID: 28135283 PMCID: PMC5279780 DOI: 10.1371/journal.pone.0170899] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 01/12/2017] [Indexed: 12/20/2022] Open
Abstract
Alternative islet transplantation sites have the potential to reduce the marginal number of islets required to ameliorate hyperglycemia in recipients with diabetes. Previously, we reported that T cell leukemia homeobox 1 (Tlx1)+ stem cells in the spleen effectively regenerated into insulin-producing cells in the pancreas of non-obese diabetic mice with end-stage disease. Thus, we investigated the spleen as a potential alternative islet transplantation site. Streptozotocin-induced diabetic C57BL/6 mice received syngeneic islets into the portal vein (PV), beneath the kidney capsule (KC), or into the spleen (SP). The marginal number of islets by PV, KC, or SP was 200, 100, and 50, respectively. Some plasma inflammatory cytokine levels in the SP group were significantly lower than those of the PV group after receiving a marginal number of islets, indicating reduced inflammation in the SP group. Insulin contents were increased 280 days after islet transplantation compared with those immediately following transplantation (p<0.05). Additionally, Tlx1-related genes, including Rrm2b and Pla2g2d, were up-regulated, which indicates that islet grafts expanded in the spleen. The spleen is an ideal candidate for an alternative islet transplantation site because of the resulting reduced inflammation and expansion of the islet graft.
Collapse
Affiliation(s)
- Takeshi Itoh
- Department of Regenerative Medicine & Transplantation, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
- Center for Regenerative Medicine, Fukuoka University Hospital, Fukuoka, Japan
| | - Hitomi Nishinakamura
- Department of Regenerative Medicine & Transplantation, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Kenjiro Kumano
- Department of Regenerative Medicine & Transplantation, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
- Center for Regenerative Medicine, Fukuoka University Hospital, Fukuoka, Japan
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hiroyuki Takahashi
- Department of Regenerative Medicine & Transplantation, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
- Center for Regenerative Medicine, Fukuoka University Hospital, Fukuoka, Japan
- Department of Gastroenterological Surgery, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Shohta Kodama
- Department of Regenerative Medicine & Transplantation, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
- Center for Regenerative Medicine, Fukuoka University Hospital, Fukuoka, Japan
| |
Collapse
|
40
|
Abstract
Even though type 2 diabetes rates plateaued, type 1 diabetes continues to increase. Pancreas transplantation is a treatment modality for patients who suffer hypoglycemic unawareness or complications from diabetes. Islet cell transplantation success rates have improved with modification and advances in isolation, transplantation, and new immunosuppression regimens. The new cell sources as well as delivery ways are explored and being tested in human trials.
Collapse
Affiliation(s)
- Betul Hatipoglu
- Endocrinology and Metabolism Institute, Cleveland Clinic, 9500 Euclid Avenue, F20, Cleveland, OH 44195, USA.
| |
Collapse
|
41
|
Abstract
Immunosuppression strategies that selectively inhibit effector T cells while preserving and even enhancing CD4FOXP3 regulatory T cells (Treg) permit immune self-regulation and may allow minimization of immunosuppression and associated toxicities. Many immunosuppressive drugs were developed before the identity and function of Treg were appreciated. A good understanding of the interactions between Treg and immunosuppressive agents will be valuable to the effective design of more tolerable immunosuppression regimens. This review will discuss preclinical and clinical evidence regarding the influence of current and emerging immunosuppressive drugs on Treg homeostasis, stability, and function as a guideline for the selection and development of Treg-friendly immunosuppressive regimens.
Collapse
Affiliation(s)
- Akiko Furukawa
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Steven A Wisel
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
| | - Qizhi Tang
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
42
|
Pellegrini S, Cantarelli E, Sordi V, Nano R, Piemonti L. The state of the art of islet transplantation and cell therapy in type 1 diabetes. Acta Diabetol 2016; 53:683-91. [PMID: 26923700 DOI: 10.1007/s00592-016-0847-z] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 02/06/2016] [Indexed: 12/17/2022]
Abstract
In patients with type 1 diabetes (T1D), pancreatic β cells are destroyed by a selective autoimmune attack and their replacement with functional insulin-producing cells is the only possible cure for this disease. The field of islet transplantation has evolved significantly from the breakthrough of the Edmonton Protocol in 2000, since significant advances in islet isolation and engraftment, together with improved immunosuppressive strategies, have been reported. The main limitations, however, remain the insufficient supply of human tissue and the need for lifelong immunosuppression therapy. Great effort is then invested in finding innovative sources of insulin-producing β cells. One old alternative with new recent perspectives is the use of non-human donor cells, in particular porcine β cells. Also the field of preexisting β cell expansion has advanced, with the development of new human β cell lines. Yet, large-scale production of human insulin-producing cells from stem cells is the most recent and promising alternative. In particular, the optimization of in vitro strategies to differentiate human embryonic stem cells into mature insulin-secreting β cells has made considerable progress and recently led to the first clinical trial of stem cell treatment for T1D. Finally, the discovery that it is possible to derive human induced pluripotent stem cells from somatic cells has raised the possibility that a sufficient amount of patient-specific β cells can be derived from patients through cell reprogramming and differentiation, suggesting that in the future there might be a cell therapy without immunosuppression.
Collapse
Affiliation(s)
- Silvia Pellegrini
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Elisa Cantarelli
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Valeria Sordi
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Rita Nano
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Lorenzo Piemonti
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
43
|
Ichihara Y, Utoh R, Yamada M, Shimizu T, Uchigata Y. Size effect of engineered islets prepared using microfabricated wells on islet cell function and arrangement. Heliyon 2016; 2:e00129. [PMID: 27441299 PMCID: PMC4946309 DOI: 10.1016/j.heliyon.2016.e00129] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 05/29/2016] [Accepted: 06/23/2016] [Indexed: 01/02/2023] Open
Abstract
Pancreatic islets are heterogeneous clusters mainly composed of α and β cells, and these clusters range in diameter from 50 to several hundred micrometers. Native small islets are known to have a higher insulin secretion ability in vitro and to provide better transplantation outcomes when compared with large islets. In this study, we prepared microengineered pseudo-islets from dispersed rat islet cells using precisely-fabricated agarose gel-based microwells with different diameters (100, 300, or 500 μm) to investigate the function and survival of islet cell aggregates with well-controlled sizes. We observed that dead cells were rarely present in the small pseudo-islets with an average diameter of ∼60 μm prepared using 100 μm microwells. In contrast, we observed more dead cells in the larger pseudo-islets prepared using 300 and 500 μm microwells. The relative amount of hypoxic cells was significantly low in the small pseudo-islets whereas a hypoxic condition was present in the core region of the larger pseudo-islets. In addition, we found that the small-sized pseudo-islets reconstituted the in vivo-tissue like arrangement of the α and β cells, and restored the high insulin secretory capacity in response to high glucose. These results clearly suggest that precise size control of pseudo-islets is essential for maintaining islet cell function and survival in vitro. The small-sized pseudo-islets may be advantageous for providing a better therapeutic approach for treating type 1 diabetes mellitus via islet reorganization and transplantation.
Collapse
Affiliation(s)
- Yumie Ichihara
- Diabetes Center, Tokyo Women's Medical University School of Medicine, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women’s Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan
| | - Rie Utoh
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women’s Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba 263-8522, Japan
- Corresponding author at: Research Fellow of the Japan Society for the Promotion of Science (JSPS). Department of Applied Chemistry and Biotechnology, Graduate School of Engineering, Chiba University, 1–33 Yayoi-cho, Inage-ku, Chiba 263–8522, Japan.Department of Applied Chemistry and BiotechnologyGraduate School of EngineeringChiba University1-33 Yayoi-choInage-kuChiba263-8522Japan
| | - Masumi Yamada
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba 263-8522, Japan
| | - Tatsuya Shimizu
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women’s Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan
| | - Yasuko Uchigata
- Diabetes Center, Tokyo Women's Medical University School of Medicine, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan
| |
Collapse
|
44
|
Madoff DC, Gaba RC, Weber CN, Clark TWI, Saad WE. Portal Venous Interventions: State of the Art. Radiology 2016; 278:333-53. [PMID: 26789601 DOI: 10.1148/radiol.2015141858] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In recent decades, there have been numerous advances in the management of liver cancer, cirrhosis, and diabetes mellitus. Although these diseases are wide ranging in their clinical manifestations, each can potentially be treated by exploiting the blood flow dynamics within the portal venous system, and in some cases, adding cellular therapies. To aid in the management of these disease states, minimally invasive transcatheter portal venous interventions have been developed to improve the safety of major hepatic resection, to reduce the untoward effects of sequelae from end-stage liver disease, and to minimize the requirement of exogenously administered insulin for patients with diabetes mellitus. This state of the art review therefore provides an overview of the most recent data and strategies for utilization of preoperative portal vein embolization, transjugular intrahepatic portosystemic shunt placement, balloon retrograde transvenous obliteration, and islet cell transplantation.
Collapse
Affiliation(s)
- David C Madoff
- From the Department of Radiology, Division of Interventional Radiology, New York-Presbyterian Hospital/Weill Cornell Medical Center, 525 E 68th St, P-518, New York, NY 10065 (D.C.M.); Department of Radiology, Interventional Radiology Section, University of Illinois Hospital, Chicago, Ill (R.C.G.); Department of Radiology, University of Pennsylvania School of Medicine, Penn Presbyterian Medical Center, Philadelphia, Pa (C.N.W., T.W.I.C.); and Department of Radiology, Division of Vascular and Interventional Radiology, University of Michigan Medical Center, Ann Arbor, Mich (W.E.S.)
| | - Ron C Gaba
- From the Department of Radiology, Division of Interventional Radiology, New York-Presbyterian Hospital/Weill Cornell Medical Center, 525 E 68th St, P-518, New York, NY 10065 (D.C.M.); Department of Radiology, Interventional Radiology Section, University of Illinois Hospital, Chicago, Ill (R.C.G.); Department of Radiology, University of Pennsylvania School of Medicine, Penn Presbyterian Medical Center, Philadelphia, Pa (C.N.W., T.W.I.C.); and Department of Radiology, Division of Vascular and Interventional Radiology, University of Michigan Medical Center, Ann Arbor, Mich (W.E.S.)
| | - Charles N Weber
- From the Department of Radiology, Division of Interventional Radiology, New York-Presbyterian Hospital/Weill Cornell Medical Center, 525 E 68th St, P-518, New York, NY 10065 (D.C.M.); Department of Radiology, Interventional Radiology Section, University of Illinois Hospital, Chicago, Ill (R.C.G.); Department of Radiology, University of Pennsylvania School of Medicine, Penn Presbyterian Medical Center, Philadelphia, Pa (C.N.W., T.W.I.C.); and Department of Radiology, Division of Vascular and Interventional Radiology, University of Michigan Medical Center, Ann Arbor, Mich (W.E.S.)
| | - Timothy W I Clark
- From the Department of Radiology, Division of Interventional Radiology, New York-Presbyterian Hospital/Weill Cornell Medical Center, 525 E 68th St, P-518, New York, NY 10065 (D.C.M.); Department of Radiology, Interventional Radiology Section, University of Illinois Hospital, Chicago, Ill (R.C.G.); Department of Radiology, University of Pennsylvania School of Medicine, Penn Presbyterian Medical Center, Philadelphia, Pa (C.N.W., T.W.I.C.); and Department of Radiology, Division of Vascular and Interventional Radiology, University of Michigan Medical Center, Ann Arbor, Mich (W.E.S.)
| | - Wael E Saad
- From the Department of Radiology, Division of Interventional Radiology, New York-Presbyterian Hospital/Weill Cornell Medical Center, 525 E 68th St, P-518, New York, NY 10065 (D.C.M.); Department of Radiology, Interventional Radiology Section, University of Illinois Hospital, Chicago, Ill (R.C.G.); Department of Radiology, University of Pennsylvania School of Medicine, Penn Presbyterian Medical Center, Philadelphia, Pa (C.N.W., T.W.I.C.); and Department of Radiology, Division of Vascular and Interventional Radiology, University of Michigan Medical Center, Ann Arbor, Mich (W.E.S.)
| |
Collapse
|
45
|
Schuetz C, Markmann JF. Islet cell transplant: Update on current clinical trials. CURRENT TRANSPLANTATION REPORTS 2016; 3:254-263. [PMID: 28451515 DOI: 10.1007/s40472-016-0103-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In the last 15 years clinical islet transplantation has made the leap from experimental procedure to standard of care for a highly selective group of patients. Due to a risk-benefit calculation involving the required systemic immunosuppression the procedure is only considered in patients with type 1 diabetes, complicated by severe hypoglycemia or end stage renal disease. In this review we summarize current outcomes of the procedure and take a look at ongoing and future improvements and refinements of beta cell therapy.
Collapse
Affiliation(s)
- Christian Schuetz
- Islet transplantation laboratory, Division of Transplantation, Department of Surgery
| | - James F Markmann
- Islet transplantation laboratory, Division of Transplantation, Department of Surgery
| |
Collapse
|
46
|
Abstract
The ultimate outcome of alloreactivity versus tolerance following transplantation is potently influenced by the constellation of cosignaling molecules expressed by immune cells during priming with alloantigen, and the net sum of costimulatory and coinhibitory signals transmitted via ligation of these molecules. Intense investigation over the last two decades has yielded a detailed understanding of the kinetics, cellular distribution, and intracellular signaling networks of cosignaling molecules such as the CD28, TNF, and TIM families of receptors in alloimmunity. More recent work has better defined the cellular and molecular mechanisms by which engagement of cosignaling networks serve to either dampen or augment alloimmunity. These findings will likely aid in the rational development of novel immunomodulatory strategies to prolong graft survival and improve outcomes following transplantation.
Collapse
Affiliation(s)
- Mandy L Ford
- Emory Transplant Center and Department of Surgery, Emory University, Atlanta, GA 30322, USA.
| |
Collapse
|
47
|
Anderson DJ, Lo DJ, Leopardi F, Song M, Turgeon NA, Strobert EA, Jenkins JB, Wang R, Reimann KA, Larsen CP, Kirk AD. Anti-Leukocyte Function-Associated Antigen 1 Therapy in a Nonhuman Primate Renal Transplant Model of Costimulation Blockade-Resistant Rejection. Am J Transplant 2016; 16:1456-64. [PMID: 26602755 PMCID: PMC5066576 DOI: 10.1111/ajt.13628] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 10/20/2015] [Accepted: 11/02/2015] [Indexed: 01/25/2023]
Abstract
Costimulation blockade with the fusion protein belatacept provides a desirable side effect profile and improvement in renal function compared with calcineurin inhibition in renal transplantation. This comes at the cost of increased rates of early acute rejection. Blockade of the integrin molecule leukocyte function-associated antigen 1 (LFA-1) has been shown to be an effective adjuvant to costimulation blockade in a rigorous nonhuman primate (NHP) model of islet transplantation; therefore, we sought to test this combination in an NHP renal transplant model. Rhesus macaques received belatacept maintenance therapy with or without the addition of LFA-1 blockade, which was achieved using a murine-derived LFA-1-specific antibody TS1/22. Additional experiments were performed using chimeric rhesus IgG1 (TS1/22R1) or IgG4 (TS1/22R4) variants, each engineered to limit antibody clearance. Despite evidence of proper binding to the target molecule and impaired cellular egress from the intravascular space indicative of a therapeutic effect similar to prior islet studies, LFA-1 blockade failed to significantly prolong graft survival. Furthermore, evidence of impaired protective immunity against cytomegalovirus was observed. These data highlight the difficulties in translating treatment regimens between organ models and suggest that the primarily vascularized renal model is more robust with regard to belatacept-resistant rejection than the islet model.
Collapse
Affiliation(s)
| | - Denise J. Lo
- Emory Transplant Center, Emory University, Atlanta, GA
| | - F. Leopardi
- Department of Surgery, Duke University, Durham, NC
| | | | | | | | | | - Rijian Wang
- MassBiologics, University of Massachusetts Medical School, Boston, MA
| | - Keith A. Reimann
- MassBiologics, University of Massachusetts Medical School, Boston, MA
| | | | - Allan D. Kirk
- Emory Transplant Center, Emory University, Atlanta, GA
- Department of Surgery, Duke University, Durham, NC
| |
Collapse
|
48
|
Abstract
Antigen-experienced T cells, also known as memory T cells, are functionally and phenotypically distinct from naive T cells. Their enhanced expression of adhesion molecules and reduced requirement for co-stimulation enables them to mount potent and rapid recall responses to subsequent antigen encounters. Memory T cells generated in response to prior antigen exposures can cross-react with other nonidentical, but similar, antigens. This heterologous cross-reactivity not only enhances protective immune responses, but also engenders de novo alloimmunity. This latter characteristic is increasingly recognized as a potential barrier to allograft acceptance that is worthy of immunotherapeutic intervention, and several approaches have been investigated. Calcineurin inhibition effectively controls memory T-cell responses to allografts, but this benefit comes at the expense of increased infectious morbidity. Lymphocyte depletion eliminates allospecific T cells but spares memory T cells to some extent, such that patients do not completely lose protective immunity. Co-stimulation blockade is associated with reduced adverse-effect profiles and improved graft function relative to calcineurin inhibition, but lacks efficacy in controlling memory T-cell responses. Targeting the adhesion molecules that are upregulated on memory T cells might offer additional means to control co-stimulation-blockade-resistant memory T-cell responses.
Collapse
|
49
|
Alfaro J, Pérez D, Jiménez C, Serrano M, Martínez-Flores JÁ, Grau M, Sánchez-Zapardiel E, Paz-Artal E, Serrano A. Blockade of cell adhesion molecules enhances cell engraftment in a murine model of liver cell transplantation. Transpl Immunol 2016; 35:7-11. [PMID: 26875547 DOI: 10.1016/j.trim.2016.01.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Revised: 12/13/2015] [Accepted: 01/30/2016] [Indexed: 11/26/2022]
Abstract
AIM OLT is the best alternative for patients with end-stage liver diseases. However, as the need for organs surpasses donor availability, alternatives to OLT are required. LCT could be a useful option versus OLT in several patients even though its low cell-engraftment hampers its efficiency. Endothelial cell barrier is the main obstacle for the implantation of cells into the parenchyma. Our study has focused on the modification of the endothelial barrier with monoclonal antibodies against adhesion molecules in order to increase cell engraftment in a mouse model of liver cell transplantation. METHODS Anti-mouse CD54 and anti-mouse CD61 antibodies were administered intrasplenically to healthy mice within 60 min prior to stem cell transplantation. Animals were sacrificed either short term at 2h or middle term seven days after transplantation. Immunohistochemical techniques to detect alkaline phosphatase activity were used to identify the transplanted cells within the liver parenchyma. RESULTS Anti-CD54 and anti-CD61 administration increases vascular patency and cell engraftment. This represents a 32% and 45% increase, respectively, of engrafted cells compared to the control (p<0.05). CONCLUSION Modification of the vascular wall with monoclonal antibodies against endothelial adhesion molecules before cell transplantation enhances cell engraftment into the mouse liver.
Collapse
Affiliation(s)
- Javier Alfaro
- Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain.
| | - Dolores Pérez
- Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Carlos Jiménez
- Department of General Surgery and Liver Transplantation, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Manuel Serrano
- Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | | | - Montserrat Grau
- Department of General Surgery and Liver Transplantation, Hospital Universitario 12 de Octubre, Madrid, Spain
| | | | - Estela Paz-Artal
- Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain; Section of Immunology, Universidad San Pablo-CEU. Madrid, Spain
| | - Antonio Serrano
- Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain; Section of Immunology, Universidad San Pablo-CEU. Madrid, Spain
| |
Collapse
|
50
|
Abstract
Immunosuppressive agents are commonly used in the nephrologist's practice in the treatment of autoimmune and immune-mediated diseases and transplantation, and they are investigational in the treatment of AKI and ESRD. Drug development has been rapid over the past decades as mechanisms of the immune response have been better defined both by serendipity (the discovery of agents with immunosuppressive activity that led to greater understanding of the immune response) and through mechanistic study (the study of immune deficiencies and autoimmune diseases and the critical pathways or mutations that contribute to disease). Toxicities of early immunosuppressive agents, such as corticosteroids, azathioprine, and cyclophosphamide, stimulated intense investigation for agents with more specificity and less harmful effects. Because the mechanisms of the immune response were better delineated over the past 30 years, this specialty is now bestowed with a multitude of therapeutic options that have reduced rejection rates and improved graft survival in kidney transplantation, provided alternatives to cytotoxic therapy in immune-mediated diseases, and opened new opportunities for intervention in diseases both common (AKI) and rare (atypical hemolytic syndrome). Rather than summarizing clinical indications and clinical trials for all currently available immunosuppressive medications, the purpose of this review is to place these agents into mechanistic context together with a brief discussion of unique features of development and use that are of interest to the nephrologist.
Collapse
Affiliation(s)
- Alexander C Wiseman
- Division of Renal Diseases and Hypertension, Transplant Center, University of Colorado, Denver, Aurora, Colorado
| |
Collapse
|