1
|
Galdina V, Puga Yung GL, Seebach JD. Cytotoxic Responses Mediated by NK Cells and Cytotoxic T Lymphocytes in Xenotransplantation. Transpl Int 2025; 38:13867. [PMID: 40012743 PMCID: PMC11862997 DOI: 10.3389/ti.2025.13867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 01/09/2025] [Indexed: 02/28/2025]
Abstract
Xenotransplantation represents a potential solution to the shortage of organs for transplantation. The recent advancements in porcine genetic modification have addressed hyperacute and acute vascular rejection; however, challenges persist with regard to delayed xenograft rejection. Porcine endothelial cells (pECs) represent a crucial target in the context of xenograft rejection, which is mediated by cytotoxic lymphocytes. It is crucial to comprehend the manner in which human natural killer (NK) cells and cytotoxic CD8+ T lymphocytes (CTL) recognize and target pECs in order to develop efficacious prophylactic strategies against rejection. The objective of the present review is to synthesize the existing knowledge regarding the mechanisms and techniques employed to modulate xenogeneic responses mediated by human NK cells and CTL. We will elucidate recent methodological advancements, debate potential novel strategies, and emphasize the imperative necessity for further research and innovative approaches to enhance graft survival.
Collapse
|
2
|
Sucu S, Yankol Y, Fernandez LA, Ekser B. Liver Xenotransplantation: A Path to Clinical Reality. Transpl Int 2025; 37:14040. [PMID: 39829719 PMCID: PMC11738628 DOI: 10.3389/ti.2024.14040] [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: 11/07/2024] [Accepted: 12/10/2024] [Indexed: 01/22/2025]
Abstract
Liver xenotransplantation has emerged as a potential solution to the shortage of deceased human donor organs and is now becoming a reality due to recent developments in genetic engineering and immunosuppressive therapy. Early efforts using non-human primates and genetically modified pigs faced significant challenges such as thrombocytopenia and graft rejection. Understanding the mechanism behind those challenges and using novel genetically engineered pigs enabled researchers to overcome some of the hurdles, but more research is needed. However, new advances might allow pig liver xenotransplantation to potentially serve as a bridge to liver allotransplantation or allow native liver regeneration in the near future.
Collapse
Affiliation(s)
- Serkan Sucu
- Division of Transplant Surgery, Department of Surgery, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, United States
- Department of Surgery, Koc University School of Medicine, Istanbul, Türkiye
| | - Yucel Yankol
- Division of Transplant Surgery, Department of Surgery, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, United States
| | - Luis A. Fernandez
- Division of Transplant Surgery, Department of Surgery, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, United States
| | - Burcin Ekser
- Division of Transplant Surgery, Department of Surgery, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, United States
| |
Collapse
|
3
|
Eisenson DL, Iwase H, Chen W, Hisadome Y, Cui W, Santillan MR, Schulick AC, Gu D, Maxwell A, Koenig K, Sun Z, Warren D, Yamada K. Combined islet and kidney xenotransplantation for diabetic nephropathy: an update in ongoing research for a clinically relevant application of porcine islet transplantation. Front Immunol 2024; 15:1351717. [PMID: 38476227 PMCID: PMC10927755 DOI: 10.3389/fimmu.2024.1351717] [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: 12/06/2023] [Accepted: 02/08/2024] [Indexed: 03/14/2024] Open
Abstract
Combined islet and kidney xenotransplantation for the treatment of diabetic nephropathy represents a compelling and increasingly relevant therapeutic possibility for an ever-growing number of patients who would benefit from both durable renal replacement and cure of the underlying cause of their renal insufficiency: diabetes. Here we briefly review immune barriers to islet transplantation, highlight preclinical progress in the field, and summarize our experience with combined islet and kidney xenotransplantation, including both challenges with islet-kidney composite grafts as well as our recent success with sequential kidney followed by islet xenotransplantation in a pig-to-baboon model.
Collapse
Affiliation(s)
- Daniel L. Eisenson
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Hayato Iwase
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Weili Chen
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Yu Hisadome
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Wanxing Cui
- Cell Therapy and Manufacturing, Medstar Georgetown University Hospital, Washington DC, United States
| | - Michelle R. Santillan
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Alexander C. Schulick
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Du Gu
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Amanda Maxwell
- Research Animal Resources, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Kristy Koenig
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Zhaoli Sun
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Daniel Warren
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Kazuhiko Yamada
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
4
|
Schmalkuche K, Rother T, Besli S, Schwinzer R, Blasczyk R, Petersen B, Figueiredo C. Human PD-L1 overexpression decreases xenogeneic human T-cell immune responses towards porcine kidneys. Front Immunol 2024; 15:1279050. [PMID: 38352884 PMCID: PMC10861674 DOI: 10.3389/fimmu.2024.1279050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 01/11/2024] [Indexed: 02/16/2024] Open
Abstract
Xenotransplantation offers a promising alternative to circumvent the lack of donated human organs available for transplantation. Different attempts to improve the survival of xenografts led to the generation of transgenic pigs expressing various combinations of human protective genes or knocked out for specific antigens. Currently, testing the efficiency of porcine organs carrying different genetic modifications in preventing xenogeneic immune responses completely relies on in vitro assays, humanized mouse models, or non-human primate transplantation models. However, these tests are often associated with major concerns due to reproducibility and generation of insufficient data as well as they raise ethical, logistical, and economic issues. In this study, we investigated the feasibility of specifically assessing the strength of human T-cell responses towards the kidneys of wild-type (WT) or transgenic pigs overexpressing human programmed death-1 ligand 1 (hPD-L1) during ex vivo kidney perfusion (EVKP). Human T cells were shown to adhere to the endothelium and transmigrate into WT and hPD-L1 kidneys. However, transcript levels of TNF-a and IFN-y as well as cytotoxic molecules such as granzyme B and perforin secreted by human T cells were significantly decreased in the tissue of hPD-L1 kidneys in comparison to WT kidneys. These results were confirmed via in vitro assays using renal endothelial cells (ECs) isolated from WT and hPD-L1 transgenic pigs. Both CD4+ and CD8+ T cells showed significantly lower proliferation rates after exposure to hPD-L1 porcine renal ECs in comparison to WT ECs. In addition, the secretion of pro-inflammatory cytokines was significantly reduced in cultures using hPD-L1 ECs in comparison to WT ECs. Remarkably, hPD-L1 EC survival was significantly increased in cytotoxic assays. This study demonstrates the feasibility of evaluating the human response of specific immune subsets such as human T cells towards the whole xenograft during EVKP. This may represent a robust strategy to assess the potency of different genetic modifications to prevent xenogeneic immune responses and thereby predict the risk of immune rejection of new genetically engineered xenografts.
Collapse
Affiliation(s)
- Katharina Schmalkuche
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
- Transregional Collaborative Research Centre 127, Hannover Medical School, Hannover, Germany
| | - Tamina Rother
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Sevval Besli
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Reinhard Schwinzer
- Transregional Collaborative Research Centre 127, Hannover Medical School, Hannover, Germany
- Transplantation Laboratory, Clinic for General, Visceral and Transplantation-Surgery, Hannover Medical School, Hannover, Germany
| | - Rainer Blasczyk
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Björn Petersen
- Transregional Collaborative Research Centre 127, Hannover Medical School, Hannover, Germany
- Department of Biotechnology, Institute of Farm Animal Genetics, Friedrich-Loeffler-Institute, Federal Research Institute for Animal Health, Neustadt am Rübenberge, Germany
| | - Constanca Figueiredo
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
- Transregional Collaborative Research Centre 127, Hannover Medical School, Hannover, Germany
| |
Collapse
|
5
|
Jou S, Mendez SR, Feinman J, Mitrani LR, Fuster V, Mangiola M, Moazami N, Gidea C. Heart transplantation: advances in expanding the donor pool and xenotransplantation. Nat Rev Cardiol 2024; 21:25-36. [PMID: 37452122 DOI: 10.1038/s41569-023-00902-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/13/2023] [Indexed: 07/18/2023]
Abstract
Approximately 65 million adults globally have heart failure, and the prevalence is expected to increase substantially with ageing populations. Despite advances in pharmacological and device therapy of heart failure, long-term morbidity and mortality remain high. Many patients progress to advanced heart failure and develop persistently severe symptoms. Heart transplantation remains the gold-standard therapy to improve the quality of life, functional status and survival of these patients. However, there is a large imbalance between the supply of organs and the demand for heart transplants. Therefore, expanding the donor pool is essential to reduce mortality while on the waiting list and improve clinical outcomes in this patient population. A shift has occurred to consider the use of organs from donors with hepatitis C virus, HIV or SARS-CoV-2 infection. Other advances in this field have also expanded the donor pool, including opt-out donation policies, organ donation after circulatory death and xenotransplantation. We provide a comprehensive overview of these various novel strategies, provide objective data on their safety and efficacy, and discuss some of the unresolved issues and controversies of each approach.
Collapse
Affiliation(s)
- Stephanie Jou
- The Zena and Michael A. Wiener Cardiovascular Institute, The Mount Sinai Hospital, New York, NY, USA.
| | - Sean R Mendez
- The Zena and Michael A. Wiener Cardiovascular Institute, The Mount Sinai Hospital, New York, NY, USA
| | - Jason Feinman
- The Zena and Michael A. Wiener Cardiovascular Institute, The Mount Sinai Hospital, New York, NY, USA
| | - Lindsey R Mitrani
- The Zena and Michael A. Wiener Cardiovascular Institute, The Mount Sinai Hospital, New York, NY, USA
| | - Valentin Fuster
- The Zena and Michael A. Wiener Cardiovascular Institute, The Mount Sinai Hospital, New York, NY, USA
| | - Massimo Mangiola
- Transplant Institute, New York University Langone Health, New York, NY, USA
| | - Nader Moazami
- Department of Cardiothoracic Surgery, New York University Langone Health, New York, NY, USA
| | - Claudia Gidea
- The Zena and Michael A. Wiener Cardiovascular Institute, The Mount Sinai Hospital, New York, NY, USA
| |
Collapse
|
6
|
Zhou L, Hou Y, Pan X, Wang X, Jin H, Yang X, Wang K, Ding X, Wang K, Zhu M, Pan Y, Wang W, Lu L. Trichosanthin-derived peptide Tk-PQ attenuates immune rejection in mouse tracheal allotransplant model by suppressing PI3K-Akt and inducing type II immune polarization. Int Immunopharmacol 2023; 125:111081. [PMID: 37862724 DOI: 10.1016/j.intimp.2023.111081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 10/10/2023] [Accepted: 10/11/2023] [Indexed: 10/22/2023]
Abstract
Obliterative bronchiolitis (OB) is one of the main complications affecting long-term survival of post-lung transplantation patients. In this study, we evaluated the efficacy of Tk-PQ (a peptide derived from trichosanthin) in alleviating OB in a mouse ectopic tracheal transplant model. We found that post-transplantation treatment of Tk-PQ significant ameliorated OB symptoms including luminal occlusion, epithelial cells loss and fibrosis in the allograft. In addition, Tk-PQ promoted immune suppressive environment by inducing Th2 polarization and increasing Treg population which in turn led to elevated levels of anti-inflammatory cytokines IL-4, IL-10, IL-33 and decreased levels of pro-inflammatory IL-1β. Mechanistically, we used transcriptome analysis of splenic T cells from allografted mice to show that Tk-PQ treatment down-regulated the PI3K-Akt signaling pathway. Indeed, the immune suppression phenotypes of Tk-PQ was recapitulated by a PI3K inhibitor LY294002. Taken together, Tk-PQ regulates post-transplantation immuno-rejection by modulating the balance of T cell response via the PI3K-Akt pathway, making it a promising peptide based immune rejection suppressant for patients receiving allotransplant.
Collapse
Affiliation(s)
- Lin Zhou
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yafei Hou
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xufeng Pan
- Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Xue Wang
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Haizhen Jin
- Central Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Xiaohua Yang
- Central Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Kefan Wang
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xuping Ding
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Kai Wang
- Central Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Minfang Zhu
- Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Yan Pan
- Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Weimin Wang
- Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Liming Lu
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Central Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China.
| |
Collapse
|
7
|
Morticelli L, Rossdam C, Cajic S, Böthig D, Magdei M, Tuladhar SR, Petersen B, Fischer K, Rapp E, Korossis S, Haverich A, Schnieke A, Niemann H, Buettner FFR, Hilfiker A. Genetic knockout of porcine GGTA1 or CMAH/GGTA1 is associated with the emergence of neo-glycans. Xenotransplantation 2023; 30:e12804. [PMID: 37148126 DOI: 10.1111/xen.12804] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 03/28/2023] [Accepted: 04/25/2023] [Indexed: 05/07/2023]
Abstract
BACKGROUND Pig-derived tissues could overcome the shortage of human donor organs in transplantation. However, the glycans with terminal α-Gal and Neu5Gc, which are synthesized by enzymes, encoded by the genes GGTA1 and CMAH, are known to play a major role in immunogenicity of porcine tissue, ultimately leading to xenograft rejection. METHODS The N-glycome and glycosphingolipidome of native and decellularized porcine pericardia from wildtype (WT), GGTA1-KO and GGTA1/CMAH-KO pigs were analyzed by multiplexed capillary gel electrophoresis coupled to laser-induced fluorescence detection. RESULTS We identified biantennary and core-fucosylated N-glycans terminating with immunogenic α-Gal- and α-Gal-/Neu5Gc-epitopes on pericardium of WT pigs that were absent in GGTA1 and GGTA1/CMAH-KO pigs, respectively. Levels of N-glycans terminating with galactose bound in β(1-4)-linkage to N-acetylglucosamine and their derivatives elongated by Neu5Ac were increased in both KO groups. N-glycans capped with Neu5Gc were increased in GGTA1-KO pigs compared to WT, but were not detected in GGTA1/CMAH-KO pigs. Similarly, the ganglioside Neu5Gc-GM3 was found in WT and GGTA1-KO but not in GGTA1/CMAH-KO pigs. The applied detergent based decellularization efficiently removed GSL glycans. CONCLUSION Genetic deletion of GGTA1 or GGTA1/CMAH removes specific epitopes providing a more human-like glycosylation pattern, but at the same time changes distribution and levels of other porcine glycans that are potentially immunogenic.
Collapse
Affiliation(s)
- Lucrezia Morticelli
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Hannover, Germany
| | - Charlotte Rossdam
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Samanta Cajic
- Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
- glyXera GmbH, Magdeburg, Germany
| | - Dietmar Böthig
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Mikhail Magdei
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Hannover, Germany
| | - Sugat Ratna Tuladhar
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Björn Petersen
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, Mariensee/Neustadt am Ruebenberge, Germany
| | - Konrad Fischer
- Chair of Livestock Biotechnology, School of Life Sciences Weihenstephan, Technische Universität München, Freising, Germany
| | - Erdmann Rapp
- Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
- glyXera GmbH, Magdeburg, Germany
| | - Sotirios Korossis
- Cardiopulmonary Regenerative Engineering (CARE) Group, Centre for Biological Engineering (CBE), Wolfson School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Loughborough, UK
| | - Axel Haverich
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Hannover, Germany
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Angelika Schnieke
- Chair of Livestock Biotechnology, School of Life Sciences Weihenstephan, Technische Universität München, Freising, Germany
| | - Heiner Niemann
- Clinic for Gastroenterology, Hepatology & Endocrinology, Hannover Medical School (MHH), Hannover, Germany
| | - Falk F R Buettner
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Andres Hilfiker
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Hannover, Germany
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| |
Collapse
|
8
|
Ye D, Qu S, Yang Y, Wang Z, Wang Q, Liu W, Zhang F, Guan Q, Wang X, Zang J, Li X, Liu H, Yao R, Feng Z, Luan Z. Intrauterine desensitization enables long term survival of human oligodendrocyte progenitor cells without immunosuppression. iScience 2023; 26:106647. [PMID: 37168574 PMCID: PMC10165029 DOI: 10.1016/j.isci.2023.106647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 03/02/2023] [Accepted: 04/05/2023] [Indexed: 05/13/2023] Open
Abstract
Immune rejection can be reduced using immunosuppressants which are not viable for premature infants. However, desensitization can induce immune tolerance for premature infants because of underdeveloped immune system. The fetuses of Wistar rats at 15-17 days gestation were injected via hOPCs-1 into brain, muscles, and abdomen ex utero and then returned while the fetuses of control without injection. After 6 weeks of desensitization, the brain and muscles were transplanted with hOPCs-1, hNSCs-1, and hOPCs-2. After 10 and 34 weeks of desensitization, hOPCs-1 and hNSCs-1 in desensitized groups was higher than that in the control group while hOPCs-2 were rejected. Treg, CD4CD28, CD8CD28, and CD45RC between the desensitization and the control group differed significantly. Inflammatory cells in group with hOPCs-1 and hNSCs-1 was lower than that in the control group. hOPCs-1 can differentiate into myelin in desensitized groups. Wistar rats with desensitization developed immune tolerance to desensitized and transplanted cells.
Collapse
Affiliation(s)
- Dou Ye
- Department of Pediatrics, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing 100037, China
- Medical School of Chinese PLA, Beijing 100853, China
| | - Suqing Qu
- Department of Pediatrics, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing 100037, China
| | - Yinxiang Yang
- Department of Pediatrics, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing 100037, China
| | - Zhaoyan Wang
- Department of Pediatrics, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing 100037, China
| | - Qian Wang
- Department of Pediatrics, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing 100037, China
| | - Weipeng Liu
- Department of Pediatrics, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing 100037, China
| | - Fan Zhang
- Department of Pediatrics, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing 100037, China
- Medical School of Chinese PLA, Beijing 100853, China
| | - Qian Guan
- Department of Pediatrics, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing 100037, China
| | - Xiaohua Wang
- Department of Pediatrics, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing 100037, China
| | - Jing Zang
- Department of Pediatrics, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing 100037, China
| | - Xin Li
- Department of Pediatrics, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing 100037, China
| | - Hengtao Liu
- Jiaen Genetics Laboratory, Beijing Jiaen Hospital, Beijing 100191, China
| | - Ruiqin Yao
- Department of Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu Province 221004, China
| | - Zhichun Feng
- Faculty of Pediatrics, The Seventh Medical Centre, Chinese PLA General Hospital, 100700 Beijing, China
| | - Zuo Luan
- Department of Pediatrics, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing 100037, China
- Medical School of Chinese PLA, Beijing 100853, China
| |
Collapse
|
9
|
Dittfeld C, Welzel C, König U, Jannasch A, Alexiou K, Blum E, Bronder S, Sperling C, Maitz MF, Tugtekin SM. Hemocompatibility tuning of an innovative glutaraldehyde-free preparation strategy using riboflavin/UV crosslinking and electron irradiation of bovine pericardium for cardiac substitutes. BIOMATERIALS ADVANCES 2023; 147:213328. [PMID: 36764200 DOI: 10.1016/j.bioadv.2023.213328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 01/17/2023] [Accepted: 02/01/2023] [Indexed: 02/09/2023]
Abstract
Hemocompatibility tuning was adopted to explore and refine an innovative, GA-free preparation strategy combining decellularization, riboflavin/UV crosslinking, and low-energy electron irradiation (SULEEI) procedure. A SULEEI-protocol was established to avoid GA-dependent deterioration that results in insufficient long-term aortic valve bioprosthesis durability. Final SULEEI-pericardium, intermediate steps and GA-fixed reference pericardium were exposed in vitro to fresh human whole blood to elucidate effects of preparation parameters on coagulation and inflammation activation and tissue histology. The riboflavin/UV crosslinking step showed to be less efficient in inactivating extracellular matrix (ECM) protein activity than the GA fixation, leading to tissue-factor mediated blood clotting. Intensifying the riboflavin/UV crosslinking with elevated riboflavin concentration and dextran caused an enhanced activation of the complement system. Yet activation processes induced by the previous protocol steps were quenched with the final electron beam treatment step. An optimized SULEEI protocol was developed using an intense and extended, trypsin-containing decellularization step to inactivate tissue factor and a dextran-free, low riboflavin, high UV crosslinking step. The innovative and improved GA-free SULEEI-preparation protocol results in low coagulant and low inflammatory bovine pericardium for surgical application.
Collapse
Affiliation(s)
- Claudia Dittfeld
- Department of Cardiac Surgery, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Heart Centre Dresden, Germany.
| | - Cindy Welzel
- Department of Cardiac Surgery, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Heart Centre Dresden, Germany
| | - Ulla König
- Fraunhofer Institute for Organic Electronics, Electron Beam and Plasma Technology FEP, Dresden, Germany
| | - Anett Jannasch
- Department of Cardiac Surgery, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Heart Centre Dresden, Germany
| | - Konstantin Alexiou
- Department of Cardiac Surgery, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Heart Centre Dresden, Germany
| | - Ekaterina Blum
- Fraunhofer Institute for Organic Electronics, Electron Beam and Plasma Technology FEP, Dresden, Germany
| | - Saskia Bronder
- Fraunhofer Institute for Organic Electronics, Electron Beam and Plasma Technology FEP, Dresden, Germany
| | - Claudia Sperling
- Leibniz-Institut für Polymerforschung Dresden e.V., Institute Biofunctional Polymer Materials, Dresden, Germany
| | - Manfred F Maitz
- Leibniz-Institut für Polymerforschung Dresden e.V., Institute Biofunctional Polymer Materials, Dresden, Germany
| | - Sems-Malte Tugtekin
- Department of Cardiac Surgery, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Heart Centre Dresden, Germany
| |
Collapse
|
10
|
Eisenson DL, Hisadome Y, Santillan MR, Yamada K. Progress in islet xenotransplantation: Immunologic barriers, advances in gene editing, and tolerance induction strategies for xenogeneic islets in pig-to-primate transplantation. FRONTIERS IN TRANSPLANTATION 2022; 1:989811. [PMID: 38390384 PMCID: PMC10883655 DOI: 10.3389/frtra.2022.989811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/24/2024]
Abstract
Islet transplantation has emerged as a curative therapy for diabetes in select patients but remains rare due to shortage of suitable donor pancreases. Islet transplantation using porcine islets has long been proposed as a solution to this organ shortage. There have already been several small clinical trials using porcine islets in humans, but results have been mixed and further trials limited by calls for more rigorous pre-clinical data. Recent progress in heart and kidney xenograft transplant, including three studies of pig-to-human xenograft transplant, have recaptured popular imagination and renewed interest in clinical islet xenotransplantation. This review outlines immunologic barriers to islet transplantation, summarizes current strategies to overcome these barriers with a particular focus on approaches to induce tolerance, and describes an innovative strategy for treatment of diabetic nephropathy with composite islet-kidney transplantation.
Collapse
Affiliation(s)
- Daniel L Eisenson
- Department of Surgery, The Johns Hopkins Hospital, Baltimore, MD, United States
| | - Yu Hisadome
- Department of Surgery, The Johns Hopkins Hospital, Baltimore, MD, United States
| | | | - Kazuhiko Yamada
- Department of Surgery, The Johns Hopkins Hospital, Baltimore, MD, United States
| |
Collapse
|
11
|
Kervella D, Le Bas-Bernardet S, Bruneau S, Blancho G. Protection of transplants against antibody-mediated injuries: from xenotransplantation to allogeneic transplantation, mechanisms and therapeutic insights. Front Immunol 2022; 13:932242. [PMID: 35990687 PMCID: PMC9389360 DOI: 10.3389/fimmu.2022.932242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 07/07/2022] [Indexed: 11/17/2022] Open
Abstract
Long-term allograft survival in allotransplantation, especially in kidney and heart transplantation, is mainly limited by the occurrence of antibody-mediated rejection due to anti-Human Leukocyte Antigen antibodies. These types of rejection are difficult to handle and chronic endothelial damages are often irreversible. In the settings of ABO-incompatible transplantation and xenotransplantation, the presence of antibodies targeting graft antigens is not always associated with rejection. This resistance to antibodies toxicity seems to associate changes in endothelial cells phenotype and modification of the immune response. We describe here these mechanisms with a special focus on endothelial cells resistance to antibodies. Endothelial protection against anti-HLA antibodies has been described in vitro and in animal models, but do not seem to be a common feature in immunized allograft recipients. Complement regulation and anti-apoptotic molecules expression appear to be common features in all these settings. Lastly, pharmacological interventions that may promote endothelial cell protection against donor specific antibodies will be described.
Collapse
Affiliation(s)
- Delphine Kervella
- CHU Nantes, Nantes Université, Néphrologie et Immunologie Clinique, Institut Transplantation Urologie Néphrologie (ITUN), Nantes, France
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, ITUN, Nantes, France
| | - Stéphanie Le Bas-Bernardet
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, ITUN, Nantes, France
| | - Sarah Bruneau
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, ITUN, Nantes, France
| | - Gilles Blancho
- CHU Nantes, Nantes Université, Néphrologie et Immunologie Clinique, Institut Transplantation Urologie Néphrologie (ITUN), Nantes, France
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, ITUN, Nantes, France
- *Correspondence: Gilles Blancho,
| |
Collapse
|
12
|
Zhou Q, Li T, Wang K, Zhang Q, Geng Z, Deng S, Cheng C, Wang Y. Current status of xenotransplantation research and the strategies for preventing xenograft rejection. Front Immunol 2022; 13:928173. [PMID: 35967435 PMCID: PMC9367636 DOI: 10.3389/fimmu.2022.928173] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 07/07/2022] [Indexed: 12/13/2022] Open
Abstract
Transplantation is often the last resort for end-stage organ failures, e.g., kidney, liver, heart, lung, and pancreas. The shortage of donor organs is the main limiting factor for successful transplantation in humans. Except living donations, other alternatives are needed, e.g., xenotransplantation of pig organs. However, immune rejection remains the major challenge to overcome in xenotransplantation. There are three different xenogeneic types of rejections, based on the responses and mechanisms involved. It includes hyperacute rejection (HAR), delayed xenograft rejection (DXR) and chronic rejection. DXR, sometimes involves acute humoral xenograft rejection (AHR) and cellular xenograft rejection (CXR), which cannot be strictly distinguished from each other in pathological process. In this review, we comprehensively discussed the mechanism of these immunological rejections and summarized the strategies for preventing them, such as generation of gene knock out donors by different genome editing tools and the use of immunosuppressive regimens. We also addressed organ-specific barriers and challenges needed to pave the way for clinical xenotransplantation. Taken together, this information will benefit the current immunological research in the field of xenotransplantation.
Collapse
Affiliation(s)
- Qiao Zhou
- Department of Rheumatology and Immunology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Ting Li
- Department of Rheumatology, Wenjiang District People’s Hospital, Chengdu, China
| | - Kaiwen Wang
- School of Medicine, Faculty of Medicine and Health, The University of Leeds, Leeds, United Kingdom
| | - Qi Zhang
- School of Medicine, University of Electronics and Technology of China, Chengdu, China
| | - Zhuowen Geng
- School of Medicine, Faculty of Medicine and Health, The University of Leeds, Leeds, United Kingdom
| | - Shaoping Deng
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
- Institute of Organ Transplantation, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, Chengdu, China
| | - Chunming Cheng
- Department of Radiation Oncology, James Comprehensive Cancer Center and College of Medicine at The Ohio State University, Columbus, OH, United States
- *Correspondence: Chunming Cheng, ; Yi Wang,
| | - Yi Wang
- Department of Critical Care Medicine, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, Chengdu, China
- *Correspondence: Chunming Cheng, ; Yi Wang,
| |
Collapse
|
13
|
Toyama C, Maeda A, Kogata S, Yamamoto R, Masahata K, Ueno T, Kamiyama M, Tazuke Y, Eguchi H, Okuyama H, Miyagawa S. Suppression of xenogeneic innate immune response by a membrane‑type human surfactant protein‑A. Exp Ther Med 2022; 24:590. [DOI: 10.3892/etm.2022.11527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 06/13/2022] [Indexed: 11/05/2022] Open
Affiliation(s)
- Chiyoshi Toyama
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565‑0871, Japan
| | - Akira Maeda
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565‑0871, Japan
| | - Shuhei Kogata
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565‑0871, Japan
| | - Riho Yamamoto
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565‑0871, Japan
| | - Kazunori Masahata
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565‑0871, Japan
| | - Takehisa Ueno
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565‑0871, Japan
| | - Masafumi Kamiyama
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565‑0871, Japan
| | - Yuko Tazuke
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565‑0871, Japan
| | - Hiroshi Eguchi
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565‑0871, Japan
| | - Hiroomi Okuyama
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565‑0871, Japan
| | - Shuji Miyagawa
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565‑0871, Japan
| |
Collapse
|
14
|
Kogata S, Lo PC, Maeda A, Okamatsu C, Sato K, Yamamoto R, Haneda T, Yoneyama T, Toyama C, Eguchi H, Masahata K, Kamiyama M, Okuyama H, Miyagawa S. Suppression of macrophage-mediated xenogeneic rejection by the ectopic expression of human CD177. Transpl Immunol 2022; 74:101663. [PMID: 35835297 DOI: 10.1016/j.trim.2022.101663] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 07/01/2022] [Accepted: 07/03/2022] [Indexed: 10/17/2022]
Abstract
Cellular xenogeneic rejection by the innate immune system is a major immunological obstruction that needs to be overcome for the successful clinical use of xenografts. Our focus has been on macrophage-mediated xenogeneic rejection, since suppressing macrophage function has considerable potential for practical applications in the area of xenotransplantation. We report herein on an investigation of the suppressive effect of human CD177 (hCD177) against macrophage-mediated xenogeneic rejection. Wild type swine aortic endothelial cell (SEC) and an SEC transfectant with hCD177 (SEC/hCD177) were co-cultured with macrophages, and the degree of cytotoxicity was evaluated by WST-8 assays, and phagocytosis was examined using Calcein-AM labeling methods. The expression of anti/pro-inflammatory cytokines was evaluated by RT-qPCR and the phosphorylation of SHP-1 on macrophages in co-culture was evaluated by Western blotting. The result of cytotoxicity assays indicated that hCD177 suppressed M1 macrophage-mediated xenogeneic rejection (vs. SEC, p < 0.0001). Similarly, the result of phagocytosis assays indicated that hCD177 suppressed it (vs. SEC, p < 0.05). In addition, hCD177 significantly suppressed the expression of IL-1β, a pro-inflammatory cytokine, in M1 macrophages (vs. SEC, p < 0.01). Luciferase assays using THP1-Lucia NF-kB also showed a significant difference in NF-kB activation (vs. SEC, p < 0.001). In addition, hCD177 was found to induce the phosphorylation of SHP-1 in M1 macrophages (vs. SEC, p < 0.05). These findings indicate that hCD177 suppresses M1 macrophage-mediated xenogeneic rejection, at least in part via in the phosphorylation of SHP-1.
Collapse
Affiliation(s)
- Shuhei Kogata
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan; Division of Pediatric Surgery, Department of Surgery, Kindai University Faculty of Medicine, Osaka, Japan
| | - Pei-Chi Lo
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Akira Maeda
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan.
| | - Chizu Okamatsu
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kazuki Sato
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Riho Yamamoto
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tomoko Haneda
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tomohisa Yoneyama
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Chiyoshi Toyama
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hiroshi Eguchi
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kazunori Masahata
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Masafumi Kamiyama
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hiroomi Okuyama
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Shuji Miyagawa
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan; Meiji University International Institute for Bio-Resource Research, Kanagawa, Japan
| |
Collapse
|
15
|
Ruan J, Zhang X, Zhao S, Xie S. Advances in CRISPR-Based Functional Genomics and Nucleic Acid Detection in Pigs. Front Genet 2022; 13:891098. [PMID: 35711930 PMCID: PMC9195075 DOI: 10.3389/fgene.2022.891098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 05/16/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- Jinxue Ruan
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, China
| | - Xuying Zhang
- Institute for Animal Breeding and Genetics, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Shuhong Zhao
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, China.,Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
| | - Shengsong Xie
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
16
|
Interdisciplinary Methods for Zoonotic Tissue Acellularization for Natural Heart Valve Substitute of Biomimetic Materials. MATERIALS 2022; 15:ma15072594. [PMID: 35407927 PMCID: PMC9000896 DOI: 10.3390/ma15072594] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/14/2022] [Accepted: 03/29/2022] [Indexed: 02/04/2023]
Abstract
The goal of this work was to create a bioactive tissue-based scaffold using multi-disciplinary engineering materials and tissue engineering techniques. Materials & methods: Physical techniques such as direct laser interference lithography and proton radiation were selected as alternative methods of enzymatic and chemical decellularization to remove cells from a tissue without degradation of the extracellular matrix nor its protein structure. This study was an attempt to prepare a functional scaffold for cell culture from tissue of animal origin using new physical methods that have not been considered before. The work was carried out under full control of the histological and molecular analysis. Results & conclusions: The most important finding was that the physical methods used to obtain the decellularized tissue scaffold differed in the efficiency of cell removal from the tissue in favour of the laser method. Both the laser method and the proton method exhibited a destructive effect on tissue structure and the genetic material in cell nuclei. This effect was visible on histology images as blurred areas within the cell nucleus. The finite element 3D simulation of decellularization process of the three-layer tissue of animal origin sample reflected well the mechanical response of tissue described by hyperelastic material models and provided results comparable to the experimental ones.
Collapse
|
17
|
Senage T, Paul A, Le Tourneau T, Fellah-Hebia I, Vadori M, Bashir S, Galiñanes M, Bottio T, Gerosa G, Evangelista A, Badano LP, Nassi A, Costa C, Cesare G, Manji RA, Cueff de Monchy C, Piriou N, Capoulade R, Serfaty JM, Guimbretière G, Dantan E, Ruiz-Majoral A, Coste du Fou G, Leviatan Ben-Arye S, Govani L, Yehuda S, Bachar Abramovitch S, Amon R, Reuven EM, Atiya-Nasagi Y, Yu H, Iop L, Casós K, Kuguel SG, Blasco-Lucas A, Permanyer E, Sbraga F, Llatjós R, Moreno-Gonzalez G, Sánchez-Martínez M, Breimer ME, Holgersson J, Teneberg S, Pascual-Gilabert M, Nonell-Canals A, Takeuchi Y, Chen X, Mañez R, Roussel JC, Soulillou JP, Cozzi E, Padler-Karavani V. The role of antibody responses against glycans in bioprosthetic heart valve calcification and deterioration. Nat Med 2022; 28:283-294. [PMID: 35177855 PMCID: PMC8863575 DOI: 10.1038/s41591-022-01682-w] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 01/06/2022] [Indexed: 12/11/2022]
Abstract
Bioprosthetic heart valves (BHVs) are commonly used to replace severely diseased heart valves but their susceptibility to structural valve degeneration (SVD) limits their use in young patients. We hypothesized that antibodies against immunogenic glycans present on BHVs, particularly antibodies against the xenoantigens galactose-α1,3-galactose (αGal) and N-glycolylneuraminic acid (Neu5Gc), could mediate their deterioration through calcification. We established a large longitudinal prospective international cohort of patients (n = 1668, 34 ± 43 months of follow-up (0.1-182); 4,998 blood samples) to investigate the hemodynamics and immune responses associated with BHVs up to 15 years after aortic valve replacement. Early signs of SVD appeared in <5% of BHV recipients within 2 years. The levels of both anti-αGal and anti-Neu5Gc IgGs significantly increased one month after BHV implantation. The levels of these IgGs declined thereafter but anti-αGal IgG levels declined significantly faster in control patients compared to BHV recipients. Neu5Gc, anti-Neu5Gc IgG and complement deposition were found in calcified BHVs at much higher levels than in calcified native aortic valves. Moreover, in mice, anti-Neu5Gc antibodies were unable to promote calcium deposition on subcutaneously implanted BHV tissue engineered to lack αGal and Neu5Gc antigens. These results indicate that BHVs manufactured using donor tissues deficient in αGal and Neu5Gc could be less prone to immune-mediated deterioration and have improved durability.
Collapse
Affiliation(s)
- Thomas Senage
- Institut du Thorax, Institut National de la Santé et de la Recherche Médicale UMR1087, University Hospital, Nantes, France
- Institut National de la Santé et de la Recherche Médicale UMR 1246-SPHERE, Nantes University, Tours University, Nantes, France
| | - Anu Paul
- Department of Cell Research and Immunology, Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Thierry Le Tourneau
- Institut du Thorax, Institut National de la Santé et de la Recherche Médicale UMR1087, University Hospital, Nantes, France
| | - Imen Fellah-Hebia
- Institut du Thorax, Institut National de la Santé et de la Recherche Médicale UMR1087, University Hospital, Nantes, France
| | - Marta Vadori
- Consortium for Research in Organ Transplantation, Ospedale Giustinianeo, Padova, Italy
| | - Salam Bashir
- Department of Cell Research and Immunology, Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Manuel Galiñanes
- Department of Cardiac Surgery and Reparative Therapy of the Heart, Vall d'Hebron Research Institute, University Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Tomaso Bottio
- Cardiovascular Regenerative Medicine Group, Department of Cardiac, Thoracic and Vascular Surgery, University of Padova, Padova, Italy
| | - Gino Gerosa
- Department of Cardiac, Vascular and Thoracic Sciences and Public Health University of Padova, L.I.F.E.L.A.B. Program Veneto Region, Padova, Italy
| | - Arturo Evangelista
- Department of Cardiology, Vall d'Hebron Research Institut, Hospital Vall d'Hebron, Barcelona, Spain
| | - Luigi P Badano
- Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
- Department of Cardiology, Neural and Metabolic Sciences, Istituto Auxologico Italiano, Istituto di Ricovero e Cura a Carattere Scientifico, San Luca Hospital, Milan, Italy
| | - Alberto Nassi
- Transplantation Immunology Unit, Padova University Hospital, Padova, Italy
| | - Cristina Costa
- Infectious Diseases and Transplantation Division, Institut d'Investigació Biomèdica de Bellvitge, L'Hospitalet de Llobregat, Barcelona, Spain
| | | | - Rizwan A Manji
- Department of Surgery, Max Rady College of Medicine, University of Manitoba Cardiac Sciences Program, St Boniface Hospital, Winnipeg, Manitoba, Canada
| | - Caroline Cueff de Monchy
- Institut du Thorax, Institut National de la Santé et de la Recherche Médicale UMR1087, University Hospital, Nantes, France
| | - Nicolas Piriou
- Institut du Thorax, Institut National de la Santé et de la Recherche Médicale UMR1087, University Hospital, Nantes, France
| | - Romain Capoulade
- Institut du Thorax, Institut National de la Santé et de la Recherche Médicale UMR1087, University Hospital, Nantes, France
| | - Jean-Michel Serfaty
- Institut du Thorax, Institut National de la Santé et de la Recherche Médicale UMR1087, University Hospital, Nantes, France
| | - Guillaume Guimbretière
- Institut du Thorax, Institut National de la Santé et de la Recherche Médicale UMR1087, University Hospital, Nantes, France
| | - Etienne Dantan
- Institut National de la Santé et de la Recherche Médicale UMR 1246-SPHERE, Nantes University, Tours University, Nantes, France
| | - Alejandro Ruiz-Majoral
- Department of Cardiology, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Guénola Coste du Fou
- Institut du Thorax, Institut National de la Santé et de la Recherche Médicale UMR1087, University Hospital, Nantes, France
| | - Shani Leviatan Ben-Arye
- Department of Cell Research and Immunology, Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Liana Govani
- Department of Cell Research and Immunology, Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Sharon Yehuda
- Department of Cell Research and Immunology, Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Shirley Bachar Abramovitch
- Department of Cell Research and Immunology, Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Ron Amon
- Department of Cell Research and Immunology, Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Eliran Moshe Reuven
- Department of Cell Research and Immunology, Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Yafit Atiya-Nasagi
- Department of Cell Research and Immunology, Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
- Israel Institute for Biological Research, Ness Ziona, Israel
| | - Hai Yu
- Department of Chemistry, University of California, Davis, Davis, CA, USA
| | - Laura Iop
- Cardiovascular Regenerative Medicine Group, Department of Cardiac, Thoracic and Vascular Surgery, University of Padova, Padova, Italy
- Department of Cardiac, Vascular and Thoracic Sciences and Public Health University of Padova, L.I.F.E.L.A.B. Program Veneto Region, Padova, Italy
- Department of Cardiac Thoracic and Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Kelly Casós
- Department of Cardiac Surgery and Reparative Therapy of the Heart, Vall d'Hebron Research Institute, University Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
- Infectious Diseases and Transplantation Division, Institut d'Investigació Biomèdica de Bellvitge, L'Hospitalet de Llobregat, Barcelona, Spain
- Department of Cardiovascular Disease at the Vall d'Hebron Institut Research, University Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Sebastián G Kuguel
- Infectious Diseases and Transplantation Division, Institut d'Investigació Biomèdica de Bellvitge, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Arnau Blasco-Lucas
- Department of Cardiac Surgery and Reparative Therapy of the Heart, Vall d'Hebron Research Institute, University Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
- Cardiac Surgery Department, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Eduard Permanyer
- Department of Cardiac Surgery and Reparative Therapy of the Heart, Vall d'Hebron Research Institute, University Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Cardiac Surgery, Quironsalud Teknon Heart Institute, Barcelona, Spain
| | - Fabrizio Sbraga
- Cardiac Surgery Department, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Roger Llatjós
- Pathology Department, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Gabriel Moreno-Gonzalez
- Infectious Diseases and Transplantation Division, Institut d'Investigació Biomèdica de Bellvitge, L'Hospitalet de Llobregat, Barcelona, Spain
- Intensive Care Department, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain
| | | | - Michael E Breimer
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Jan Holgersson
- Institute of Biomedicine, Department of Laboratory Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Susann Teneberg
- Institute of Biomedicine, Department of Medical Biochemistry and Cell Biology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | | | | | - Yasuhiro Takeuchi
- Division of Infection and Immunity, University College London, London, UK
| | - Xi Chen
- Department of Chemistry, University of California, Davis, Davis, CA, USA
| | - Rafael Mañez
- Infectious Diseases and Transplantation Division, Institut d'Investigació Biomèdica de Bellvitge, L'Hospitalet de Llobregat, Barcelona, Spain.
- Intensive Care Department, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain.
| | - Jean-Christian Roussel
- Institut du Thorax, Institut National de la Santé et de la Recherche Médicale UMR1087, University Hospital, Nantes, France.
| | - Jean-Paul Soulillou
- Institut de Transplantation-Urologie-Néphrologie, Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche 1064, Centre Hospitalier Universitaire de Nantes, Nantes, France.
| | - Emanuele Cozzi
- Transplantation Immunology Unit, Padova University Hospital, Padova, Italy.
| | - Vered Padler-Karavani
- Department of Cell Research and Immunology, Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
18
|
Aghali A. Craniofacial Bone Tissue Engineering: Current Approaches and Potential Therapy. Cells 2021; 10:cells10112993. [PMID: 34831216 PMCID: PMC8616509 DOI: 10.3390/cells10112993] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/16/2021] [Accepted: 10/22/2021] [Indexed: 01/10/2023] Open
Abstract
Craniofacial bone defects can result from various disorders, including congenital malformations, tumor resection, infection, severe trauma, and accidents. Successfully regenerating cranial defects is an integral step to restore craniofacial function. However, challenges managing and controlling new bone tissue formation remain. Current advances in tissue engineering and regenerative medicine use innovative techniques to address these challenges. The use of biomaterials, stromal cells, and growth factors have demonstrated promising outcomes in vitro and in vivo. Natural and synthetic bone grafts combined with Mesenchymal Stromal Cells (MSCs) and growth factors have shown encouraging results in regenerating critical-size cranial defects. One of prevalent growth factors is Bone Morphogenetic Protein-2 (BMP-2). BMP-2 is defined as a gold standard growth factor that enhances new bone formation in vitro and in vivo. Recently, emerging evidence suggested that Megakaryocytes (MKs), induced by Thrombopoietin (TPO), show an increase in osteoblast proliferation in vitro and bone mass in vivo. Furthermore, a co-culture study shows mature MKs enhance MSC survival rate while maintaining their phenotype. Therefore, MKs can provide an insight as a potential therapy offering a safe and effective approach to regenerating critical-size cranial defects.
Collapse
Affiliation(s)
- Arbi Aghali
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA;
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47908, USA
| |
Collapse
|
19
|
Morticelli L, Magdei M, Tschalaki N, Petersen B, Haverich A, Hilfiker A. Generation of glycans depleted decellularized porcine pericardium, using digestive enzymatic supplements and enzymatic mixtures for food industry. Xenotransplantation 2021; 28:e12705. [PMID: 34227157 DOI: 10.1111/xen.12705] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 05/27/2021] [Accepted: 06/24/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND Xenogeneic pericardium has been used largely for various applications in cardiovascular surgery. Nevertheless, xenogeneic pericardial patches fail mainly due to their antigenic components. The xenoantigens identified as playing a major role in recipient immune response are the Galα1-3Gal (α-Gal) epitope, the non-human sialic acid N-glycolylneuraminic acid (Neu5Gc), and the porcine SDa antigen, associated with both proteins and lipids. The reduction in glycans from porcine pericardium might hinder or reduce the immunogenicity of xenogeneic scaffolds. METHODS Decellularized porcine pericardia were further treated at different time points and dilutions with digestive enzymatic supplements and enzymatic mixtures applied for food industry, for the removal of potentially immunogenic carbohydrates. Carbohydrates removal was investigated using up to 8 different lectin stains for the identification of N- and O-glycosylations, as well as glycolipids. Histoarchitectural changes in the ECM were assessed using Elastica van Gieson stain, whereas changes in mechanical properties were investigated via uniaxial tensile test and burst pressure test. RESULTS Tissues after enzymatic treatments showed a dramatic decrease in lectin stainings in comparison to tissues which were only decellularized. Histological assessment revealed cell-nuclei removal after decellularization. Some of the enzymatic treatments induced elastic lamellae disruption. Tissue strength decreased after enzymatic treatment; however, treated tissues showed values of burst pressure higher than physiological transvalvular pressures. CONCLUSIONS The application of these enzymatic treatments for tissue deglycosylation is totally novel, low cost, and appears to be very efficient for glycan removal. The immunogenic potential of treated tissues will be further investigated in subsequent studies, in vitro and in vivo.
Collapse
Affiliation(s)
- Lucrezia Morticelli
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Hannover, Germany.,Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Mikhail Magdei
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Hannover, Germany.,Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Negin Tschalaki
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Hannover, Germany.,Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Björn Petersen
- Department of Biotechnology, Institute of Farm Animal Genetics, Friedrich-Loeffler-Institute, Mariensee, Neustadt, Germany
| | - Axel Haverich
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Hannover, Germany.,Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Andres Hilfiker
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Hannover, Germany.,Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| |
Collapse
|
20
|
CRISPR/Cas Technology in Pig-to-Human Xenotransplantation Research. Int J Mol Sci 2021; 22:ijms22063196. [PMID: 33801123 PMCID: PMC8004187 DOI: 10.3390/ijms22063196] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/17/2021] [Accepted: 03/18/2021] [Indexed: 02/06/2023] Open
Abstract
CRISPR/Cas (clustered regularly interspaced short palindromic repeats linked to Cas nuclease) technology has revolutionized many aspects of genetic engineering research. Thanks to it, it became possible to study the functions and mechanisms of biology with greater precision, as well as to obtain genetically modified organisms, both prokaryotic and eukaryotic. The changes introduced by the CRISPR/Cas system are based on the repair paths of the single or double strand DNA breaks that cause insertions, deletions, or precise integrations of donor DNA. These changes are crucial for many fields of science, one of which is the use of animals (pigs) as a reservoir of tissues and organs for xenotransplantation into humans. Non-genetically modified animals cannot be used to save human life and health due to acute immunological reactions resulting from the phylogenetic distance of these two species. This review is intended to collect and summarize the advantages as well as achievements of the CRISPR/Cas system in pig-to-human xenotransplantation research. In addition, it demonstrates barriers and limitations that require careful evaluation before attempting to experiment with this technology.
Collapse
|
21
|
Bozso SJ, El-Andari R, Al-Adra D, Moon MC, Freed DH, Nagendran J, Nagendran J. A review of the immune response stimulated by xenogenic tissue heart valves. Scand J Immunol 2021; 93:e13018. [PMID: 33372305 DOI: 10.1111/sji.13018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/30/2020] [Accepted: 12/26/2020] [Indexed: 12/23/2022]
Abstract
Valvular heart disease continues to afflict millions of people around the world. In many cases, the only corrective treatment for valvular heart disease is valve replacement. Valve replacement options are currently limited, and the most common construct utilized are xenogenic tissue heart valves. The main limitation with the use of this valve type is the development of valvular deterioration. Valve deterioration results in intrinsic permanent changes in the valve structure, often leading to hemodynamic compromise and clinical symptoms of valve re-stenosis. A significant amount of research has been performed regarding the incidence of valve deterioration and determination of significant risk factors for its development. As a result, many believe that the underlying driver of valve deterioration is a chronic immune-mediated rejection process of the foreign xenogenic-derived tissue. The underlying mechanisms of how this occurs are an area of ongoing research and active debate. In this review, we provide an overview of the important components of the immune system and how they respond to xenografts. A review of the proposed mechanisms of xenogenic heart valve deterioration is provided including the immune response to xenografts. Finally, we discuss the role of strategies to combat valve degeneration such as preservation protocols, epitope modification and decellularization.
Collapse
Affiliation(s)
- Sabin J Bozso
- Department of Surgery, Division of Cardiac Surgery, University of Alberta, Edmonton, AB, Canada
| | - Ryaan El-Andari
- Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - David Al-Adra
- Department of Surgery, Division of Transplantation, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Michael C Moon
- Department of Surgery, Division of Cardiac Surgery, University of Alberta, Edmonton, AB, Canada
| | - Darren H Freed
- Department of Surgery, Division of Cardiac Surgery, University of Alberta, Edmonton, AB, Canada
| | - Jayan Nagendran
- Department of Surgery, Division of Cardiac Surgery, University of Alberta, Edmonton, AB, Canada
| | - Jeevan Nagendran
- Department of Surgery, Division of Cardiac Surgery, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
22
|
Huang CP, Yang CY, Shyr CR. Utilizing Xenogeneic Cells As a Therapeutic Agent for Treating Diseases. Cell Transplant 2021; 30:9636897211011995. [PMID: 33975464 PMCID: PMC8120531 DOI: 10.1177/09636897211011995] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/19/2021] [Accepted: 04/05/2021] [Indexed: 12/27/2022] Open
Abstract
The utilization of biologically produced cells to treat diseases is a revolutionary invention in modern medicine after chemically synthesized small molecule drugs and biochemically made protein drugs. Cells are basic units of life with diverse functions in mature and developing organs, which biological properties could be utilized as a promising therapeutic approach for currently intractable and incurable diseases. Xenogeneic cell therapy utilizing animal cells other than human for medicinal purpose has been studied as a new way of treating diseases. Xenogeneic cell therapy is considered as a potential regenerative approach to fulfill current unmet medical needs because xenogeneic cells could be isolated from different animal organs and expanded ex vivo as well as maintain the characteristics of original organs, providing a versatile and plenty cell source for cell-based therapeutics beside autologous and allogeneic sources. The swine species is considered the most suitable source because of the similarity with humans in size and physiology of many organs in addition to the economic and ethical reasons plus the possibility of genetic modification. This review discusses the old proposed uses of xenogeneic cells such as xenogeneic pancreatic islet cells, hepatocytes and neuronal cells as a living drug for the treatment of degenerative and organ failure diseases. Novel applications of xenogeneic mesenchymal stroma cells and urothelial cells are also discussed. There are formidable immunological barriers toward successful cellular xenotransplantation in clinic despite major progress in the development of novel immunosuppression regimens and genetically multimodified donor pigs. However, immunological barriers could be turn into immune boosters by using xenogeneic cells of specific tissue types as a novel immunotherapeutic agent to elicit bystander antitumor immunity due to rejection immune responses. Xenogeneic cells have the potential to become a safe and efficacious option for intractable diseases and hard-to-treat cancers, adding a new class of cellular medicine in our drug armamentarium.
Collapse
Affiliation(s)
- Chi-Ping Huang
- Department of Urology, School of Medicine, China Medical University and Hospital, Taichung, Taiwan
| | - Chi-Yu Yang
- Animal Technology Research Center/Division of Animal Technology, Agriculture Technology Research Institute, Miaoli, Taiwan
| | - Chih-Rong Shyr
- Sex Hormone Research Center, Department of Medical Laboratory Science and Biotechnology, China Medical University and Hospital, Taichung, Taiwan
| |
Collapse
|
23
|
Ming SL, Zeng L, Guo YK, Zhang S, Li GL, Ma YX, Zhai YY, Chang WR, Yang L, Wang J, Yang GY, Chu BB. The Human-Specific STING Agonist G10 Activates Type I Interferon and the NLRP3 Inflammasome in Porcine Cells. Front Immunol 2020; 11:575818. [PMID: 33072119 PMCID: PMC7543045 DOI: 10.3389/fimmu.2020.575818] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 08/28/2020] [Indexed: 01/18/2023] Open
Abstract
Pigs have anatomical and physiological characteristics comparable to those in humans and, therefore, are a favorable model for immune function research. Interferons (IFNs) and inflammasomes have essential roles in the innate immune system. Here, we report that G10, a human-specific agonist of stimulator of interferon genes (STING), activates both type I IFN and the canonical NLRP3 inflammasome in a STING-dependent manner in porcine cells. Without a priming signal, G10 alone transcriptionally stimulated Sp1-dependent p65 expression, thus triggering activation of the nuclear factor-κB (NF-κB) signaling pathway and thereby priming inflammasome activation. G10 was also found to induce potassium efflux- and NLRP3/ASC/Caspase-1-dependent secretion of IL-1β and IL-18. Pharmacological and genetic inhibition of NLRP3 inflammasomes increased G10-induced type I IFN expression, thereby preventing virus infection, suggesting negative regulation of the NLRP3 inflammasome in the IFN response in the context of STING-mediated innate immune activation. Overall, our findings reveal a new mechanism through which G10 activates the NLRP3 inflammasome in porcine cells and provide new insights into STING-mediated innate immunity in pigs compared with humans.
Collapse
Affiliation(s)
- Sheng-Li Ming
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Lei Zeng
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Yu-Kun Guo
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Shuang Zhang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Guo-Li Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Ying-Xian Ma
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Yun-Yun Zhai
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Wen-Ru Chang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Le Yang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Jiang Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Guo-Yu Yang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Bei-Bei Chu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| |
Collapse
|
24
|
Longoni A, Pennings I, Cuenca Lopera M, van Rijen MHP, Peperzak V, Rosenberg AJWP, Levato R, Gawlitta D. Endochondral Bone Regeneration by Non-autologous Mesenchymal Stem Cells. Front Bioeng Biotechnol 2020; 8:651. [PMID: 32733861 PMCID: PMC7363768 DOI: 10.3389/fbioe.2020.00651] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 05/27/2020] [Indexed: 12/31/2022] Open
Abstract
Mimicking endochondral bone formation is a promising strategy for bone regeneration. To become a successful therapy, the cell source is a crucial translational aspect. Typically, autologous cells are used. The use of non-autologous mesenchymal stromal cells (MSCs) represents an interesting alternative. Nevertheless, non-autologous, differentiated MSCs may trigger an undesired immune response, hampering bone regeneration. The aim of this study was to unravel the influence of the immune response on endochondral bone regeneration, when using xenogeneic (human) or allogeneic (Dark Agouti) MSCs. To this end, chondrogenically differentiated MSCs embedded in a collagen carrier were implanted in critical size femoral defects of immunocompetent Brown Norway rats. Control groups were included with syngeneic/autologous (Brown Norway) MSCs or a cell-free carrier. The amount of neo-bone formation was proportional to the degree of host-donor relatedness, as no full bridging of the defect was observed in the xenogeneic group whereas 2/8 and 7/7 bridges occurred in the allogeneic and the syngeneic group, respectively. One week post-implantation, the xenogeneic grafts were invaded by pro-inflammatory macrophages, T lymphocytes, which persisted after 12 weeks, and anti-human antibodies were developed. The immune response toward the allogeneic graft was comparable to the one evoked by the syngeneic implants, aside from an increased production of alloantibodies, which might be responsible for the more heterogeneous bone formation. Our results demonstrate for the first time the feasibility of using non-autologous MSC-derived chondrocytes to elicit endochondral bone regeneration in vivo. Nevertheless, the pronounced immune response and the limited bone formation observed in the xenogeneic group undermine the clinical relevance of this group. On the contrary, although further research on how to achieve robust bone formation with allogeneic cells is needed, they may represent an alternative to autologous transplantation.
Collapse
Affiliation(s)
- Alessia Longoni
- Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Regenerative Medicine Center Utrecht, Utrecht, Netherlands
| | - I Pennings
- Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Regenerative Medicine Center Utrecht, Utrecht, Netherlands
| | - Marta Cuenca Lopera
- Laboratory for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - M H P van Rijen
- Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Regenerative Medicine Center Utrecht, Utrecht, Netherlands.,Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Victor Peperzak
- Laboratory for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - A J W P Rosenberg
- Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Riccardo Levato
- Regenerative Medicine Center Utrecht, Utrecht, Netherlands.,Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Debby Gawlitta
- Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Regenerative Medicine Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
25
|
Evaluation of the CRISPR/Cas9 Genetic Constructs in Efficient Disruption of Porcine Genes for Xenotransplantation Purposes Along with an Assessment of the Off-Target Mutation Formation. Genes (Basel) 2020; 11:genes11060713. [PMID: 32604937 PMCID: PMC7349392 DOI: 10.3390/genes11060713] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 06/17/2020] [Accepted: 06/24/2020] [Indexed: 12/17/2022] Open
Abstract
The increasing life expectancy of humans has led to an increase in the number of patients with chronic diseases and organ failure. However, the imbalance between the supply and the demand for human organs is a serious problem in modern transplantology. One of many solutions to overcome this problem is the use of xenotransplantation. The domestic pig (Sus scrofa domestica) is currently considered as the most suitable for human organ procurement. However, there are discrepancies between pigs and humans that lead to the creation of immunological barriers preventing the direct xenograft. The introduction of appropriate modifications to the pig genome to prevent xenograft rejection is crucial in xenotransplantation studies. In this study, porcine GGTA1, CMAH, β4GalNT2, vWF, ASGR1 genes were selected to introduce genetic modifications. The evaluation of three selected gRNAs within each gene was obtained, which enabled the selection of the best site for efficient introduction of changes. Modifications were examined after nucleofection of porcine primary kidney fibroblasts with CRISPR/Cas9 system genetic constructs, followed by the tracking of indels by decomposition (TIDE) analysis. In addition, off-target analysis was carried out for selected best gRNAs using the TIDE tool, which is new in the research conducted so far and shows the utility of this tool in these studies.
Collapse
|
26
|
He J, Li Z, Yu T, Wang W, Tao M, Wang S, Ma Y, Fan J, Tian X, Wang X, Javed R, Ao Q. In vitro and in vivo biocompatibility study on acellular sheep periosteum for guided bone regeneration. Biomed Mater 2020; 15:015013. [DOI: 10.1088/1748-605x/ab597f] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
27
|
Lu T, Yang B, Wang R, Qin C. Xenotransplantation: Current Status in Preclinical Research. Front Immunol 2020; 10:3060. [PMID: 32038617 PMCID: PMC6989439 DOI: 10.3389/fimmu.2019.03060] [Citation(s) in RCA: 130] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 12/16/2019] [Indexed: 12/13/2022] Open
Abstract
The increasing life expectancy of humans has led to a growing numbers of patients with chronic diseases and end-stage organ failure. Transplantation is an effective approach for the treatment of end-stage organ failure; however, the imbalance between organ supply and the demand for human organs is a bottleneck for clinical transplantation. Therefore, xenotransplantation might be a promising alternative approach to bridge the gap between the supply and demand of organs, tissues, and cells; however, immunological barriers are limiting factors in clinical xenotransplantation. Thanks to advances in gene-editing tools and immunosuppressive therapy as well as the prolonged xenograft survival time in pig-to-non-human primate models, clinical xenotransplantation has become more viable. In this review, we focus on the evolution and current status of xenotransplantation research, including our current understanding of the immunological mechanisms involved in xenograft rejection, genetically modified pigs used for xenotransplantation, and progress that has been made in developing pig-to-pig-to-non-human primate models. Three main types of rejection can occur after xenotransplantation, which we discuss in detail: (1) hyperacute xenograft rejection, (2) acute humoral xenograft rejection, and (3) acute cellular rejection. Furthermore, in studies on immunological rejection, genetically modified pigs have been generated to bridge cross-species molecular incompatibilities; in the last decade, most advances made in the field of xenotransplantation have resulted from the production of genetically engineered pigs; accordingly, we summarize the genetically modified pigs that are currently available for xenotransplantation. Next, we summarize the longest survival time of solid organs in preclinical models in recent years, including heart, liver, kidney, and lung xenotransplantation. Overall, we conclude that recent achievements and the accumulation of experience in xenotransplantation mean that the first-in-human clinical trial could be possible in the near future. Furthermore, we hope that xenotransplantation and various approaches will be able to collectively solve the problem of human organ shortage.
Collapse
Affiliation(s)
- Tianyu Lu
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China.,NHC Key Laboratory of Human Disease Comparative Medicine, The Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Beijing, China
| | - Bochao Yang
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China.,NHC Key Laboratory of Human Disease Comparative Medicine, The Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Beijing, China
| | - Ruolin Wang
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China.,NHC Key Laboratory of Human Disease Comparative Medicine, The Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Beijing, China
| | - Chuan Qin
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China.,NHC Key Laboratory of Human Disease Comparative Medicine, The Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Beijing, China
| |
Collapse
|
28
|
Chen L, Yang Z, Wang Y, Du L, Li Y, Zhang N, Gao W, Peng R, Zhu F, Wang L, Li C, Li J, Wang F, Sun Q, Zhang D. Single xenotransplant of rat brown adipose tissue prolonged the ovarian lifespan of aging mice by improving follicle survival. Aging Cell 2019; 18:e13024. [PMID: 31389140 PMCID: PMC6826128 DOI: 10.1111/acel.13024] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 06/27/2019] [Accepted: 07/14/2019] [Indexed: 02/06/2023] Open
Abstract
Prolonging the ovarian lifespan is attractive and challenging. An optimal clinical strategy must be safe, long-acting, simple, and economical. Allotransplantation of brown adipose tissue (BAT), which is most abundant and robust in infants, has been utilized to treat various mouse models of human disease. Could we use BAT to prolong the ovarian lifespan of aging mice? Could we try BAT xenotransplantation to alleviate the clinical need for allogeneic BAT due to the lack of voluntary infant donors? In the current study, we found that a single rat-to-mouse (RTM) BAT xenotransplantation did not cause systemic immune rejection but did significantly increase the fertility of mice and was effective for more than 5 months (equivalent to 10 years in humans). Next, we did a series of analysis including follicle counting; AMH level; estrous cycle; mTOR activity; GDF9, BMP15, LHR, Sirt1, and Cyp19a level; ROS and annexin V level; IL6 and adiponectin level; biochemical blood indices; body temperature; transcriptome; and DNA methylation studies. From these, we proposed that rat BAT xenotransplantation rescued multiple indices indicative of follicle and oocyte quality; rat BAT also improved the metabolism and general health of the aging mice; and transcriptional and epigenetic (DNA methylation) improvement in F0 mice could benefit F1 mice; and multiple KEGG pathways and GO classified biological processes the differentially expressed genes (DEGs) or differentially methylated regions (DMRs) involved were identical between F0 and F1. This study could be a helpful reference for clinical BAT xenotransplantation from close human relatives to the woman.
Collapse
Affiliation(s)
- Liang‐Jian Chen
- State Key Lab of Reproductive MedicineNanjing Medical UniversityNanjingChina
| | - Zhi‐Xia Yang
- State Key Lab of Reproductive MedicineNanjing Medical UniversityNanjingChina
| | - Yang Wang
- State Key Lab of Reproductive MedicineNanjing Medical UniversityNanjingChina
| | - Lei Du
- State Key Lab of Reproductive MedicineNanjing Medical UniversityNanjingChina
- Department of Center for Medical ExperimentsThird Xiang‐Ya Hospital of Central South UniversityChangshaChina
| | - Yan‐Ru Li
- State Key Lab of Reproductive MedicineNanjing Medical UniversityNanjingChina
| | - Na‐Na Zhang
- State Key Lab of Reproductive MedicineNanjing Medical UniversityNanjingChina
| | - Wen‐Yi Gao
- State Key Lab of Reproductive MedicineNanjing Medical UniversityNanjingChina
| | - Rui‐Rui Peng
- State Key Lab of Reproductive MedicineNanjing Medical UniversityNanjingChina
| | - Feng‐Yu Zhu
- State Key Lab of Reproductive MedicineNanjing Medical UniversityNanjingChina
| | - Li‐Li Wang
- State Key Lab of Reproductive MedicineNanjing Medical UniversityNanjingChina
| | - Cong‐Rong Li
- State Key Lab of Reproductive MedicineNanjing Medical UniversityNanjingChina
| | - Jian‐Min Li
- Animal Core FacilityNanjing Medical UniversityNanjingChina
| | - Fu‐Qiang Wang
- Analysis & Test CenterNanjing Medical UniversityNanjingChina
| | - Qing‐Yuan Sun
- State Key Lab of Stem Cell and Reproductive Biology, Institute of ZoologyChinese Academy of SciencesBeijingChina
| | - Dong Zhang
- State Key Lab of Reproductive MedicineNanjing Medical UniversityNanjingChina
- Animal Core FacilityNanjing Medical UniversityNanjingChina
| |
Collapse
|
29
|
Bracey DN, Jinnah AH, Willey JS, Seyler TM, Hutchinson ID, Whitlock PW, Smith TL, Danelson KA, Emory CL, Kerr BA. Investigating the Osteoinductive Potential of a Decellularized Xenograft Bone Substitute. Cells Tissues Organs 2019; 207:97-113. [PMID: 31655811 PMCID: PMC6935535 DOI: 10.1159/000503280] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 09/09/2019] [Indexed: 12/14/2022] Open
Abstract
Bone grafting is the second most common tissue transplantation procedure worldwide. One of the alternative methods for bone repair under investigation is a tissue-engineered bone substitute. An ideal property of tissue-engineered bone substitutes is osteoinductivity, defined as the ability to stimulate primitive cells to differentiate into a bone-forming lineage. In the current study, we use a decellularization and oxidation protocol to produce a porcine bone scaffold and examine whether it possesses osteoinductive potential and can be used to create a tissue-engineered bone microenvironment. The decellularization protocol was patented by our lab and consists of chemical decellularization and oxidation steps using combinations of deionized water, trypsin, antimicrobials, peracetic acid, and triton-X100. To test if the bone scaffold was a viable host, preosteoblasts were seeded and analyzed for markers of osteogenic differentiation. The osteoinductive potential was observed in vitro with similar osteogenic markers being expressed in preosteoblasts seeded on the scaffolds and demineralized bone matrix. To assess these properties in vivo, scaffolds with and without preosteoblasts preseeded were subcutaneously implanted in mice for 4 weeks. MicroCT scanning revealed 1.6-fold increased bone volume to total volume ratio and 1.4-fold increase in trabecular thickness in scaffolds after implantation. The histological analysis demonstrates new bone formation and blood vessel formation with pentachrome staining demonstrating osteogenesis and angiogenesis, respectively, within the scaffold. Furthermore, CD31+ staining confirmed the endothelial lining of the blood vessels. These results demonstrate that porcine bone maintains its osteoinductive properties after the application of a patented decellularization and oxidation protocol developed in our laboratory. Future work must be performed to definitively prove osteogenesis of human mesenchymal stem cells, biocompatibility in large animal models, and osteoinduction/osseointegration in a relevant clinical model in vivo. The ability to create a functional bone microenvironment using decellularized xenografts will impact regenerative medicine, orthopedic reconstruction, and could be used in the research of multiple diseases.
Collapse
Affiliation(s)
- Daniel N. Bracey
- Wake Forest Baptist Medical Center, Orthopaedic Surgery, Winston Salem, NC, USA
| | - Alexander H. Jinnah
- Wake Forest Baptist Medical Center, Orthopaedic Surgery, Winston Salem, NC, USA
| | - Jeffrey S. Willey
- Wake Forest Baptist Medical Center, Radiation Oncology, Winston Salem, NC, USA
| | | | | | | | - Thomas L. Smith
- Wake Forest Baptist Medical Center, Orthopaedic Surgery, Winston Salem, NC, USA
| | - Kerry A. Danelson
- Wake Forest Baptist Medical Center, Orthopaedic Surgery, Winston Salem, NC, USA
| | - Cynthia L. Emory
- Wake Forest Baptist Medical Center, Orthopaedic Surgery, Winston Salem, NC, USA
| | - Bethany A. Kerr
- Wake Forest Baptist Medical Center, Orthopaedic Surgery, Winston Salem, NC, USA
- Virginia Tech-Wake Forest University School for Bioengineering and Sciences, Winston Salem, NC, USA
- Wake Forest School of Medicine, Cancer Biology, Winston Salem, NC, USA
| |
Collapse
|
30
|
Wolf E, Kemter E, Klymiuk N, Reichart B. Genetically modified pigs as donors of cells, tissues, and organs for xenotransplantation. Anim Front 2019; 9:13-20. [PMID: 32002258 PMCID: PMC6951927 DOI: 10.1093/af/vfz014] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Eckhard Wolf
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Science, LMU Munich, Munich, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Elisabeth Kemter
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Science, LMU Munich, Munich, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Nikolai Klymiuk
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Science, LMU Munich, Munich, Germany
| | - Bruno Reichart
- Walter Brendel Center for Experimental Medicine, LMU Munich, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Munich, Germany
| |
Collapse
|
31
|
Lo PC, Maeda A, Kodama T, Takakura C, Yoneyama T, Sakai R, Noguchi Y, Matsuura R, Eguchi H, Matsunami K, Okuyama H, Miyagawa S. The novel immunosuppressant prenylated quinolinecarboxylic acid-18 (PQA-18) suppresses macrophage differentiation and cytotoxicity in xenotransplantation. Immunobiology 2019; 224:575-584. [PMID: 30967296 DOI: 10.1016/j.imbio.2019.04.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 03/28/2019] [Accepted: 04/01/2019] [Indexed: 12/12/2022]
Abstract
Innate immunity plays a major role in xenograft rejection. However, the majority of immunosuppressants focus on inhibiting acquired immunity and not innate immunity. Therefore, a novel immunosuppressant suitable for use in conjunction with xenografts continues to be needed. It has been reported that prenylated quinolinecarboxylic acid-18 (PQA-18), a p21-activated kinase 2 (PAK2) inhibitor, exerts an immunosuppressive function on T cells. Hence, the possibility exists that PQA-18 might be used in conjunction with xenografts, which prompted us to investigate the efficacy of PQA-18 on macrophages compared with Tofacitinib, a janus kinase (JAK) inhibitor. Initial experiments confirmed that PQA-18 is non-toxic to swine endothelial cells (SECs) and human monocytes. Both PQA-18 and Tofacitinib suppressed macrophage-mediated cytotoxicity in both the differentiation and effector phases. Both PQA-18 and tofacitinib suppressed the expression of HLA-ABC by macrophages. However, contrary to Tofacitinib, PQA-18 also significantly suppressed the expression of CD11b, HLA-DR and CD40 on macrophages. PQA-18 significantly suppressed CCR7 expression on day 3 and on day 6, but Tofacitinib-induced suppression only on day 6. In a mixed lymphocyte reaction (MLR) assay, PQA-18 was found to suppress Interleukin-2 (IL-2)-stimulated T cell proliferation to a lesser extent than Tofacitinib. However, PQA-18 suppressed xenogeneic-induced T cell proliferation more strongly than Tofacitinib on day 3 and the suppression was similar on day 7. In conclusion, PQA-18 has the potential to function as an immunosuppressant for xenotransplantation.
Collapse
Affiliation(s)
- Pei-Chi Lo
- Department of Surgery, Osaka University Graduate School of Medicine Japan
| | - Akira Maeda
- Department of Surgery, Osaka University Graduate School of Medicine Japan.
| | - Tasuku Kodama
- Department of Surgery, Osaka University Graduate School of Medicine Japan
| | - Chihiro Takakura
- Department of Surgery, Osaka University Graduate School of Medicine Japan
| | - Tomohisa Yoneyama
- Department of Surgery, Osaka University Graduate School of Medicine Japan
| | - Rieko Sakai
- Department of Surgery, Osaka University Graduate School of Medicine Japan
| | - Yuki Noguchi
- Department of Surgery, Osaka University Graduate School of Medicine Japan
| | - Rei Matsuura
- Department of Surgery, Osaka University Graduate School of Medicine Japan
| | - Hiroshi Eguchi
- Department of Surgery, Osaka University Graduate School of Medicine Japan
| | | | - Hiroomi Okuyama
- Department of Surgery, Osaka University Graduate School of Medicine Japan
| | - Shuji Miyagawa
- Department of Surgery, Osaka University Graduate School of Medicine Japan
| |
Collapse
|
32
|
Edri R, Gal I, Noor N, Harel T, Fleischer S, Adadi N, Green O, Shabat D, Heller L, Shapira A, Gat-Viks I, Peer D, Dvir T. Personalized Hydrogels for Engineering Diverse Fully Autologous Tissue Implants. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1803895. [PMID: 30406960 DOI: 10.1002/adma.201803895] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 08/27/2018] [Indexed: 05/22/2023]
Abstract
Despite incremental improvements in the field of tissue engineering, no technology is currently available for producing completely autologous implants where both the cells and the scaffolding material are generated from the patient, and thus do not provoke an immune response that may lead to implant rejection. Here, a new approach is introduced to efficiently engineer any tissue type, which its differentiation cues are known, from one small tissue biopsy. Pieces of omental tissues are extracted from patients and, while the cells are reprogrammed to become induced pluripotent stem cells, the extracellular matrix is processed into an immunologically matching, thermoresponsive hydrogel. Efficient cell differentiation within a large 3D hydrogel is reported, and, as a proof of concept, the generation of functional cardiac, cortical, spinal cord, and adipogenic tissue implants is demonstrated. This versatile bioengineering approach may assist to regenerate any tissue and organ with a minimal risk for immune rejection.
Collapse
Affiliation(s)
- Reuven Edri
- School for Molecular Cell Biology and Biotechnology, Tel Aviv University, Tel Aviv, 69978, Israel
- The Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Idan Gal
- School for Molecular Cell Biology and Biotechnology, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Nadav Noor
- Department of Materials Science and Engineering, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Tom Harel
- School for Molecular Cell Biology and Biotechnology, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Sharon Fleischer
- School for Molecular Cell Biology and Biotechnology, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Nofar Adadi
- Department of Materials Science and Engineering, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Ori Green
- School of Chemistry, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Doron Shabat
- School of Chemistry, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Lior Heller
- Department of Plastic Surgery, Assaf Harofeh MC, Beer Ya'akov, Zerifin, 70300, Israel
| | - Assaf Shapira
- School for Molecular Cell Biology and Biotechnology, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Irit Gat-Viks
- School for Molecular Cell Biology and Biotechnology, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Dan Peer
- School for Molecular Cell Biology and Biotechnology, Tel Aviv University, Tel Aviv, 69978, Israel
- The Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, 69978, Israel
- Department of Materials Science and Engineering, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Tal Dvir
- School for Molecular Cell Biology and Biotechnology, Tel Aviv University, Tel Aviv, 69978, Israel
- The Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, 69978, Israel
- Department of Materials Science and Engineering, Tel Aviv University, Tel Aviv, 69978, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 69978, Israel
- Sagol Center for Regenerative Biotechnology, Tel Aviv University, Tel Aviv, 69978, Israel
| |
Collapse
|
33
|
Bhattacharya R, Das P, Joardar SN, Biswas BK, Batabyal S, Das PK, Nandi SK. Novel decellularized animal conchal cartilage graft for application in human patient. J Tissue Eng Regen Med 2018; 13:46-57. [PMID: 30358120 DOI: 10.1002/term.2767] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 08/19/2018] [Accepted: 10/18/2018] [Indexed: 12/16/2022]
Abstract
Restoration of the external ear and nose in human patients, in either congenital deformity or acquired defects, is a challenge in reconstructive surgery. Optimization of the currently available materials is necessary for rhinoplasty and microtia correction to avoid intraoperative manoeuvring and early rejection. The aim of this study was to develop cross-linked decellularized caprine conchal cartilages as biocompatible, robust, and non-toxic matrix template. The characterization of the decellularized tissue encompasses in vitro lymphoproliferation assay, cytotoxicity test, agar gel precipitation test, in vivo immunocompatibility study, histology, and determination of pro-inflammatory cytokines in animal model. Decellularized cartilage was implanted in human volunteer at R. G. Kar Medical College and Hospital, Kolkata, India, and samples were assessed histologically by retrieving those after 4 months. The processed cartilages were implanted in rhinoplasty (nine) and microtia patients (six) keeping autogenous cartilage graft as control up to 18 months after surgery. Primary outcomes were viability and safety of the material, both in animal model and human pre-application in actual site. Secondary outcomes included self-assessed clinical findings on gross examination. This study is under the ethical approval no. RKC/14 dated January 27, 2012. The in vitro cellular reactivity was less in processed cartilage protein than control. Histology of retrieved tissues in animal model and human volunteer showed no adverse reactions. Production of IL-2, IL-6, and TNF-α cytokines was lower at 4 weeks. The rhinoplasty and microtia operation in clinical patients utilizing the processed cartilage showed satisfactory recovery with improved facial look. These low cost, easily available, biocompatible, safe xenocartilage biomatrices of caprine conchal cartilage origin are very flexible in shape and size, enabling them as potential bioimplant for repair of nasal and auricular structure without any rejection or diverse biomedical applications.
Collapse
Affiliation(s)
- Rupnarayan Bhattacharya
- Department of Plastic Surgery, R. G. Kar Medical College and Hospital, Kolkata, West Bengal, India
| | - Piyali Das
- Department of Biotechnology Project, Department of Veterinary Surgery and Radiology, West Bengal University of Animal and Fishery Sciences, Kolkata, West Bengal, India
| | - Siddhartha Narayan Joardar
- Department of Veterinary Microbiology, West Bengal University of Animal and Fishery Sciences, Kolkata, West Bengal, India
| | - Bikash Kanti Biswas
- Directorate of Research, Extension and Farms, West Bengal University of Animal and Fishery Sciences, Kolkata, West Bengal, India
| | - Subhasis Batabyal
- Department of Veterinary Biochemistry, West Bengal University of Animal and Fishery Sciences, Kolkata, West Bengal, India
| | - Pradip Kumar Das
- Department of Veterinary Physiology, West Bengal University of Animal and Fishery Sciences, Kolkata, West Bengal, India
| | - Samit Kumar Nandi
- Department of Veterinary Surgery and Radiology, West Bengal University of Animal and Fishery Sciences, Kolkata, West Bengal, India
| |
Collapse
|
34
|
Jin X, Hu M, Gong L, Li H, Wang Y, Ji M, Li H. Adoptive transfer of xenoantigen‑stimulated T cell receptor Vβ‑restricted human regulatory T cells prevents porcine islet xenograft rejection in humanized mice. Mol Med Rep 2018; 18:4457-4467. [PMID: 30221725 PMCID: PMC6172378 DOI: 10.3892/mmr.2018.9471] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 08/03/2018] [Indexed: 02/05/2023] Open
Abstract
Polyclonal expansion of human regulatory T cells (Tregs) prevents xenogeneic rejection by suppressing effector T cell responses in vitro and in vivo. However, a major limitation to using polyclonally expanded Tregs is that they may cause pan‑immunosuppressive effects. The present study was conducted to compare the ability of ex vivo expanded human xenoantigen‑stimulated Tregs (Xeno‑Treg) and polyclonal Tregs (Poly‑Treg) to protect islet xenografts from rejection in NOD‑SCID interleukin (IL)‑2 receptor (IL2r)γ‑/‑ mice. Human cluster of differentiation (CD)4+CD25+CD127lo Tregs, expanded either by stimulating with porcine peripheral blood mononuclear cells (PBMCs) or anti‑CD3/CD28 beads, were characterized by immune cell phenotyping, T cell receptor (TCR) Vβ CDR3 spectratyping and performing suppressive activity assays in vitro. The efficiency of adoptively transferred ex vivo human Tregs was evaluated in vivo using neonatal porcine islet cell clusters (NICC) transplanted into NOD‑SCID IL‑2rγ‑/‑ mice, which received human PBMCs with or without Xeno‑Treg or Poly‑Treg. Xeno‑Treg, which expressed increased levels of human leukocyte antigen‑DR and secreted higher levels of IL‑10, demonstrated enhanced suppressive capacity in a pig‑human mixed lymphocyte reaction. Spectratypes of TCR Vβ4, Vβ10, Vβ18 and Vβ20 in Xeno‑Treg showed restriction and expanded clones at sizes of 205, 441, 332 and 196 respectively, compared to those of Poly‑Treg. Reconstitution of mice with human PBMCs and Poly‑Treg resulted in NICC xenograft rejection at 63 days. Adoptive transfer with human PBMCs and Xeno‑Treg prolonged islet xenograft survival beyond 84 days, with grafts containing intact insulin‑secreting cells surrounded by a small number of human CD45+ cells. This study demonstrated that adoptive transfer of ex vivo expanded human Xeno‑Treg may potently prevent islet xenograft rejection in humanized NOD‑SCID IL2rγ‑/‑ mice compared with Poly‑Treg. These findings suggested that adoptive Treg therapy may be used for immunomodulation in islet xenotransplantation by minimizing systemic immunosuppression.
Collapse
Affiliation(s)
- Xi Jin
- Institute of Urology, Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Min Hu
- Centre for Transplant and Renal Research, Westmead Millennium Institute, University of Sydney, Westmead, NSW 2145, Australia
| | - Lina Gong
- Institute of Urology, Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Huifang Li
- Cellular Biology Laboratory, Core Facility of West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yan Wang
- Cellular Biology Laboratory, Core Facility of West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Ming Ji
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410083, P.R. China
| | - Hong Li
- Institute of Urology, Department of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
35
|
Guruswamy Damodaran R, Vermette P. Tissue and organ decellularization in regenerative medicine. Biotechnol Prog 2018; 34:1494-1505. [PMID: 30294883 DOI: 10.1002/btpr.2699] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 04/30/2018] [Indexed: 12/22/2022]
Abstract
The advancement and improvement in decellularization methods can be attributed to the increasing demand for tissues and organs for transplantation. Decellularized tissues and organs, which are free of cells and genetic materials while retaining the complex ultrastructure of the extracellular matrix (ECM), can serve as scaffolds to subsequently embed cells for transplantation. They have the potential to mimic the native physiology of the targeted anatomic site. ECM from different tissues and organs harvested from various sources have been applied. Many techniques are currently involved in the decellularization process, which come along with their own advantages and disadvantages. This review focuses on recent developments in decellularization methods, the importance and nature of detergents used for decellularization, as well as on the role of the ECM either as merely a physical support or as a scaffold in retaining and providing cues for cell survival, differentiation and homeostasis. In addition, application, status, and perspectives on commercialization of bioproducts derived from decellularized tissues and organs are addressed. © 2018 American Institute of Chemical Engineers Biotechnol. Prog., 34:1494-1505, 2018.
Collapse
Affiliation(s)
- Rajesh Guruswamy Damodaran
- Laboratoire de bio-ingénierie et de biophysique de l'Université de Sherbrooke, Department of Chemical and Biotechnological Engineering, Université de Sherbrooke, 2500 Boul. de l'Université, Sherbrooke, QC, J1K 2R1, Canada.,Pharmacology Institute of Sherbrooke, Faculté de médecine et des sciences de la santé, 3001 12ième Avenue Nord, Sherbrooke, Québec, J1H 5N4, Canada.,Research Centre on Aging, Institut universitaire de gériatrie de Sherbrooke, 1036 rue Belvédère Sud, Sherbrooke, Québec, J1H 4C4, Canada
| | - Patrick Vermette
- Laboratoire de bio-ingénierie et de biophysique de l'Université de Sherbrooke, Department of Chemical and Biotechnological Engineering, Université de Sherbrooke, 2500 Boul. de l'Université, Sherbrooke, QC, J1K 2R1, Canada.,Pharmacology Institute of Sherbrooke, Faculté de médecine et des sciences de la santé, 3001 12ième Avenue Nord, Sherbrooke, Québec, J1H 5N4, Canada.,Research Centre on Aging, Institut universitaire de gériatrie de Sherbrooke, 1036 rue Belvédère Sud, Sherbrooke, Québec, J1H 4C4, Canada
| |
Collapse
|
36
|
Black CK, Termanini KM, Aguirre O, Hawksworth JS, Sosin M. Solid organ transplantation in the 21 st century. ANNALS OF TRANSLATIONAL MEDICINE 2018; 6:409. [PMID: 30498736 PMCID: PMC6230860 DOI: 10.21037/atm.2018.09.68] [Citation(s) in RCA: 189] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 09/29/2018] [Indexed: 12/20/2022]
Abstract
Solid organ transplantation (SOT) has emerged from an experimental approach in the 20th century to now being an established and practical definitive treatment option for patients with end-organ dysfunction. The evolution of SOT has seen the field progress rapidly over the past few decades with incorporation of a variety of solid organs-liver, kidney, pancreas, heart, and lung-into the donor pool. New advancements in surgical technique have allowed for more efficient and refined multi-organ procurements with minimal complications and decreased ischemic injury events. Additionally, immunosuppression therapy has also seen advancements with the expansion of immunosuppressive protocols to dampen the host immune response and improve short and long-term graft survival. However, the field of SOT faces new barriers, most importantly the expanding demand for SOT that is outpacing the current supply. Allocation protocols have been developed in an attempt to address these concerns. Other avenues for SOT are also being explored to increase the donor pool, including split-liver donor transplants, islet cell implantation for pancreas transplants, and xenotransplantation. The future of SOT is bright with exciting new research being explored to overcome current obstacles.
Collapse
Affiliation(s)
- Cara K. Black
- Georgetown University School of Medicine, Washington, DC, USA
| | | | - Oswaldo Aguirre
- MedStar Georgetown University Hospital, MedStar Georgetown Transplant Institute, Washington, DC, USA
| | - Jason S. Hawksworth
- MedStar Georgetown University Hospital, MedStar Georgetown Transplant Institute, Washington, DC, USA
| | - Michael Sosin
- Hansjörg Wyss Department of Plastic Surgery, NYU Langone Health, New York, NY, USA
| |
Collapse
|
37
|
Abstract
PURPOSE OF REVIEW Porcine islets represent a potentially attractive beta-cell source for xenotransplantation into patients with type 1 diabetes, who are not eligible to islet allo-transplantation due to a lack of suitable human donor organs. Recent progress in genetic engineering/gene editing of donor pigs provides new opportunities to overcome rejection of xeno-islets, to improve their engraftment and insulin secretion capacity, and to reduce the risk for transmission of porcine endogenous retroviruses. This review summarizes the current issues and progress in islet xenotransplantation with special emphasis on genetically modified/gene edited donor pigs. RECENT FINDINGS Attempts to overcome acute rejection of xeno-islets, especially after intraportal transplantation into the liver, include the genetic elimination of specific carbohydrate antigens such as αGal, Neu5Gc, and Sd(a) for which humans and-in part-non-human primates have natural antibodies that bind to these targets leading to activation of complement and coagulation. A complementary approach is the expression of one or more human complement regulatory proteins (hCD46, hCD55, hCD59). Transgenic attempts to overcome cellular rejection of islet xenotransplants include the expression of proteins that inhibit co-stimulation of T cells. Expression of glucagon-like peptide-1 and M3 muscarinic receptors has been shown to increase the insulin secretion of virally transduced porcine islets in vitro and it will be interesting to see the effects of these modifications in transgenic pigs and islet products derived from them. Genome-wide inactivation of porcine endogenous retrovirus (PERV) integrants by mutating their pol genes using CRISPR/Cas9 is a recent approach to reduce the risk for PERV transmission by xeno-islets. Genetic engineering/gene editing of xeno-islet donor pigs facilitated major progress towards clinical islet xenotransplantation. The required set of genetic modifications will depend on the source of islets (fetal/neonatal vs. adult), the mode of delivery (encapsulated vs. free), and the transplantation site.
Collapse
Affiliation(s)
- Elisabeth Kemter
- Gene Center, and Center for Innovative Medical Models (CiMM), LMU Munich, Feodor-Lynen-Str. 25, 81377, Munich, Germany
| | - Joachim Denner
- Robert Koch Institute, Nordufer 20, 13353, Berlin, Germany
| | - Eckhard Wolf
- Gene Center, and Center for Innovative Medical Models (CiMM), LMU Munich, Feodor-Lynen-Str. 25, 81377, Munich, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
| |
Collapse
|
38
|
Paul A, Padler-Karavani V. Evolution of sialic acids: Implications in xenotransplant biology. Xenotransplantation 2018; 25:e12424. [PMID: 29932472 PMCID: PMC6756921 DOI: 10.1111/xen.12424] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 05/14/2018] [Indexed: 12/13/2022]
Abstract
All living cells are covered with a dense “sugar-coat” of carbohydrate chains (glycans) conjugated to proteins and lipids. The cell surface glycome is determined by a non-template driven process related to the collection of enzymes that assemble glycans in a sequential manner. In mammals, many of these glycans are topped with sialic acids (Sia), a large family of acidic sugars. The “Sialome” is highly diverse owing to various Sia types, linkage to underlying glycans, range of carriers, and complex spatial organization. Presented at the front of cells, Sia play a major role in immunity and recognition of “self” versus “non-self,” largely mediated by the siglecs family of Sia-binding host receptors. Albeit many mammalian pathogens have evolved to hijack this recognition system to avoid host immune attack, presenting a fascinating host-pathogen evolutionary arms race. Similarly, cancer cells exploit Sia for their own survival and propagation. As part of this ongoing fitness, humans lost the ability to synthesize the Sia type N-glycolylneuraminic acid (Neu5Gc), in contrast to other mammals. While this loss had provided an advantage against certain pathogens, humans are continuously exposed to Neu5Gc through mammalian-derived diet (eg, red meat), consequently generating a complex immune response against it. Circulating anti-Neu5Gc antibodies together with Neu5Gc on some human tissues mediate chronic inflammation “xenosialitis” that exacerbate various human diseases (eg, cancer and atherosclerosis). Similarly, Neu5Gc-containing xenografts are exposed to human anti-Neu5Gc antibodies with implications to sustainability. This review aimed to provide a glimpse into the evolution of Sia and their implications to xenotransplantation.
Collapse
Affiliation(s)
- Anu Paul
- Department of Cell Research and Immunology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Vered Padler-Karavani
- Department of Cell Research and Immunology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
39
|
Smith KE, Johnson RC, Papas KK. Update on cellular encapsulation. Xenotransplantation 2018; 25:e12399. [DOI: 10.1111/xen.12399] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Accepted: 03/27/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Kate E. Smith
- Department of Physiological Sciences; University of Arizona; Tucson AZ USA
- Department of Surgery; University of Arizona; Tucson AZ USA
| | | | | |
Collapse
|
40
|
Buermann A, Petkov S, Petersen B, Hein R, Lucas-Hahn A, Baars W, Brinkmann A, Niemann H, Schwinzer R. Pigs expressing the human inhibitory ligand PD-L1 (CD 274) provide a new source of xenogeneic cells and tissues with low immunogenic properties. Xenotransplantation 2018; 25:e12387. [DOI: 10.1111/xen.12387] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 12/27/2017] [Accepted: 01/12/2018] [Indexed: 01/01/2023]
Affiliation(s)
- Anna Buermann
- Transplant Laboratory; Department of General-, Visceral-, and Transplantation Surgery; Hannover Medical School; Hannover Germany
| | - Stoyan Petkov
- Institute of Farm Animal Genetics; Friedrich-Loeffler-Institut; Mariensee Germany
| | - Björn Petersen
- Institute of Farm Animal Genetics; Friedrich-Loeffler-Institut; Mariensee Germany
| | - Rabea Hein
- Transplant Laboratory; Department of General-, Visceral-, and Transplantation Surgery; Hannover Medical School; Hannover Germany
| | - Andrea Lucas-Hahn
- Institute of Farm Animal Genetics; Friedrich-Loeffler-Institut; Mariensee Germany
| | - Wiebke Baars
- Transplant Laboratory; Department of General-, Visceral-, and Transplantation Surgery; Hannover Medical School; Hannover Germany
| | - Antje Brinkmann
- Transplant Laboratory; Department of General-, Visceral-, and Transplantation Surgery; Hannover Medical School; Hannover Germany
| | - Heiner Niemann
- Institute of Farm Animal Genetics; Friedrich-Loeffler-Institut; Mariensee Germany
| | - Reinhard Schwinzer
- Transplant Laboratory; Department of General-, Visceral-, and Transplantation Surgery; Hannover Medical School; Hannover Germany
| |
Collapse
|
41
|
Shrock E, Güell M. CRISPR in Animals and Animal Models. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 152:95-114. [PMID: 29150007 DOI: 10.1016/bs.pmbts.2017.07.010] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
CRISPR-Cas9 has revolutionized the generation of transgenic animals. This system has demonstrated an unprecedented efficiency, multiplexability, and ease of use, thereby reducing the time and cost required for genome editing and enabling the production of animals with more extensive genetic modifications. It has also been shown to be applicable to a wide variety of animals, from early-branching metazoans to primates. Genome-wide screens in model organisms have been performed, accurate models of human diseases have been constructed, and potential therapies have been tested and validated in animal models. Several achievements in genetic modification of animals have been translated into products for the agricultural and pharmaceutical industries. Based on the remarkable progress to date, one may anticipate that in the future, CRISPR-Cas9 technology will enable additional far-reaching advances, including understanding the bases of diseases with complex genetic origins, engineering animals to produce organs for human transplantation, and genetically transforming entire populations of organisms to prevent the spread of disease.
Collapse
Affiliation(s)
- Ellen Shrock
- Biological and Biomedical Sciences, Harvard University, Boston, MA, United States; Harvard Medical School, Boston, MA, United States
| | - Marc Güell
- Harvard Medical School, Boston, MA, United States; Pompeu Fabra University, Barcelona, Spain.
| |
Collapse
|
42
|
Guo F, Hu M, Huang D, Zhao Y, Heng B, Guillemin G, Lim CK, Hawthorne WJ, Yi S. Human regulatory macrophages are potent in suppression of the xenoimmune response via indoleamine-2,3-dioxygenase-involved mechanism(s). Xenotransplantation 2017; 24. [PMID: 28771838 DOI: 10.1111/xen.12326] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Revised: 05/17/2017] [Accepted: 07/06/2017] [Indexed: 12/15/2022]
Abstract
BACKGROUND For xenotransplantation to truly succeed, we must develop immunomodulatory strategies to suppress the xenoimmune response but by minimizing immunosuppression over the long term. Regulatory macrophages (Mreg) have been shown to suppress polyclonal T-cell proliferation in vitro and prolong allograft survival in vivo. However, the question of whether they are capable of suppressing xenoimmune responses remains unknown. This study assessed the potential of human Mreg to be used as an effective immunomodulatory method in xenotransplantation. METHODS CD14+ monocytes selected from human peripheral blood mononuclear cells (PBMC) were cultured with macrophage colony-stimulating factor (M-CSF) for 7 days with IFN-γ added at day 6 for Mreg induction. Mreg phenotyping was performed by flow cytometric analysis, and the in vitro suppressive function was assessed by mixed lymphocyte reaction (MLR) using irradiated pig PBMC as the xenogeneic stimulator cells, human PBMC as responder cells, and autologous Mreg as suppressor cells. To assess mRNA expression of Mreg functional molecules indoleamine-2,3-dioxygenase (IDO), IL-10, inducible nitric oxide synthase (iNOS) and TGF-β were measured by real-time PCR. Supernatants were collected from the MLR cultures for IDO activity assay by high-performance liquid chromatography (HPLC). The effects of the IDO inhibitor 1-D/L-methyl-tryptophan (1-MT), iNOS inhibitor NG -monomethyl-l-arginine (L-NMMA), and anti-IFN-γ or anti-TGF-β monoclonal antibody (mAb) treatment on Mreg suppressive capacity were tested from the supernatants of the MLR assays. RESULTS We demonstrated that induced Mreg with a phenotype of CD14low CD16-/low CD80low CD83-/low CD86+/hi HLA-DR+/hi were capable of suppressing proliferating human PBMC, CD4+, and CD8+ T cells, even at a higher responder:Mreg ratio of 32:1 in a pig-human xenogeneic MLR. The strong suppressive potency of Mreg was further correlated with their upregulated IDO expression and activity. The IDO upregulation of Mreg was associated with an increased production of IFN-γ, an IDO stimulator, by xenoreactive responder cells in the xenogeneic MLR. While no effect on Mreg suppressive potency was detected by addition of the iNOS inhibitor L-NMMA or anti-TGF-β mAb into the MLR assays, inhibition of IDO activity by neutralizing IFN-γ or by IDO inhibitor 1-MT substantially impaired the capacity of Mreg to suppress the xenogeneic response, indicating the importance of upregulated IDO activity in Mreg-mediated suppression of the xenogeneic response in vitro. CONCLUSION This study demonstrates that human Mreg are capable of suppressing the xenoimmune response in vitro via IDO-involved mechanism(s), suggesting their potential role as an effective immunomodulatory tool in xenotransplantation.
Collapse
Affiliation(s)
- Fei Guo
- Centre for Transplant & Renal Research, Westmead Institute for Medical Research, University of Sydney, Westmead, NSW, Australia.,Cell Transplantation and Gene Therapy Institute of Central South University at the 3rd Xiangya Hospital, Changsha, Hunan, China
| | - Min Hu
- Centre for Transplant & Renal Research, Westmead Institute for Medical Research, University of Sydney, Westmead, NSW, Australia
| | - Dandan Huang
- Centre for Transplant & Renal Research, Westmead Institute for Medical Research, University of Sydney, Westmead, NSW, Australia
| | - Yuanfei Zhao
- Centre for Transplant & Renal Research, Westmead Institute for Medical Research, University of Sydney, Westmead, NSW, Australia
| | - Benjamin Heng
- Faculty of Medicine and Health Sciences, Macquarie University, North Ryde, NSW, Australia
| | - Gilles Guillemin
- Faculty of Medicine and Health Sciences, Macquarie University, North Ryde, NSW, Australia
| | - Chai K Lim
- Faculty of Medicine and Health Sciences, Macquarie University, North Ryde, NSW, Australia
| | - Wayne J Hawthorne
- Centre for Transplant & Renal Research, Westmead Institute for Medical Research, University of Sydney, Westmead, NSW, Australia
| | - Shounan Yi
- Centre for Transplant & Renal Research, Westmead Institute for Medical Research, University of Sydney, Westmead, NSW, Australia
| |
Collapse
|
43
|
Abstract
Type 1 diabetes (T1D) patients who receive pancreatic islet transplant experience significant improvement in their quality-of-life. This comes primarily through improved control of blood sugar levels, restored awareness of hypoglycemia, and prevention of serious and potentially life-threatening diabetes-associated complications, such as kidney failure, heart and vascular disease, stroke, nerve damage, and blindness. Therefore, beta cell replacement through transplantation of isolated islets is an important option in the treatment of T1D. However, lasting success of this promising therapy depends on durable survival and efficacy of the transplanted islets, which are directly influenced by the islet isolation procedures. Thus, isolating pancreatic islets with consistent and reliable quality is critical in the clinical application of islet transplantation.Quality of isolated islets is important in pre-clinical studies as well, as efforts to advance and improve clinical outcomes of islet transplant therapy have relied heavily on animal models ranging from rodents, to pigs, to nonhuman primates. As a result, pancreatic islets have been isolated from these and other species and used in a variety of in vitro or in vivo applications for this and other research purposes. Protocols for islet isolation have been somewhat similar across species, especially, in mammals. However, given the increasing evidence about the distinct structural and functional features of human and mouse islets, using similar methods of islet isolation may contribute to inconsistencies in the islet quality, immunogenicity, and experimental outcomes. This may also contribute to the discrepancies commonly observed between pre-clinical findings and clinical outcomes. Therefore, it is prudent to consider the particular features of pancreatic islets from different species when optimizing islet isolation protocols.In this chapter, we explore the structural and functional features of pancreatic islets from mice, pigs, nonhuman primates, and humans because of their prevalent use in nonclinical, preclinical, and clinical applications.
Collapse
|
44
|
Londono R, Dziki JL, Haljasmaa E, Turner NJ, Leifer CA, Badylak SF. The effect of cell debris within biologic scaffolds upon the macrophage response. J Biomed Mater Res A 2017; 105:2109-2118. [DOI: 10.1002/jbm.a.36055] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2016] [Revised: 02/08/2017] [Accepted: 03/02/2017] [Indexed: 12/16/2022]
Affiliation(s)
- Ricardo Londono
- Department of Surgery; McGowan Institute for Regenerative Medicine, University of Pittsburgh; Pittsburgh Pennsylvania
- School of Medicine; University of Pittsburgh; Pittsburgh Pennsylvania
| | - Jenna L. Dziki
- Department of Surgery; McGowan Institute for Regenerative Medicine, University of Pittsburgh; Pittsburgh Pennsylvania
- Department of Bioengineering; University of Pittsburgh; Pittsburgh Pennsylvania
| | - Eric Haljasmaa
- Department of Surgery; McGowan Institute for Regenerative Medicine, University of Pittsburgh; Pittsburgh Pennsylvania
- Department of Bioengineering; University of Pittsburgh; Pittsburgh Pennsylvania
| | - Neill J. Turner
- Department of Surgery; McGowan Institute for Regenerative Medicine, University of Pittsburgh; Pittsburgh Pennsylvania
| | - Cynthia A. Leifer
- Department of Microbiology and Immunology; Cornell University College of Veterinary Medicine; Ithaca New York
| | - Stephen F. Badylak
- Department of Surgery; McGowan Institute for Regenerative Medicine, University of Pittsburgh; Pittsburgh Pennsylvania
- School of Medicine; University of Pittsburgh; Pittsburgh Pennsylvania
- Department of Bioengineering; University of Pittsburgh; Pittsburgh Pennsylvania
- Department of Surgery; University of Pittsburgh; Pittsburgh Pennsylvania
| |
Collapse
|