1
|
Hirose T, Hotta K, Otsuka R, Seino KI. Mechanism and regulation of the complement activity in kidney xenotransplantation. Transplant Rev (Orlando) 2025; 39:100931. [PMID: 40233672 DOI: 10.1016/j.trre.2025.100931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 04/05/2025] [Accepted: 04/06/2025] [Indexed: 04/17/2025]
Abstract
Xenotransplantation is emerging as one of several potential solutions for addressing organ donor shortages, with significant progress bringing it closer to clinical application. However, challenges remain, particularly concerning complement system dysregulation caused by species differences, as well as xenoantigens and coagulopathy. Complement regulatory proteins expressed on endothelial cells of donor xenografts are less compatible with complement components in recipients. These difficulties contribute to hyperacute rejection, characterized by antibody-mediated complement activation that destroys the graft within 24 h. Moreover, because molecules are incompatible across different species, ischemia-reperfusion injury or infection can easily elicit complement activity via all three pathways, resulting in xenograft loss via complement-mediated vascular injury. Complement activity also stimulate innate and adaptive immune cells. To address this issue, genetic modifications in donor pigs and the development of novel medicines have been tested in preclinical models with promising results. Pigs modified to express human complement-regulating molecules such as CD46, CD55, and CD59 have shown longer kidney xenograft survivals over years in preclinical models with nonhuman primates, paving the way for clinical trials. Anti-complement component agents such as C1 esterase and C5 inhibitors have also been shown to increase xenograft survivals. This review examines the role of the complement system in kidney xenotransplantation, emphasizing new research and clinical trial advancements.
Collapse
Affiliation(s)
- Takayuki Hirose
- Department of Urology, Hokkaido University Hospital, Sapporo, Japan.
| | - Kiyohiko Hotta
- Department of Urology, Hokkaido University Hospital, Sapporo, Japan
| | - Ryo Otsuka
- Massachusetts General Hospital, Department of Surgery, Harvard Medical School, Boston, MA 02114, USA
| | - Ken-Ichiro Seino
- Division of Immunobiology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| |
Collapse
|
2
|
Hidalgo-Vicelis JL, Rivera-Contreras AR, Hernández-Téllez B, Piñón-Zárate G, Jarquín-Yáñez K, Fiordelisio-Coll T, Saniger-Blesa JM, González-Gómez GH, Falcón-Neri MA, Canales-Martínez MM, Castell-Rodríguez AE. Thermosensitive Porcine Myocardial Extracellular Matrix Hydrogel Coupled with Proanthocyanidins for Cardiac Tissue Engineering. Gels 2025; 11:53. [PMID: 39852024 PMCID: PMC11764510 DOI: 10.3390/gels11010053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/03/2025] [Accepted: 01/05/2025] [Indexed: 01/26/2025] Open
Abstract
Currently, there are no therapies that prevent the negative myocardial remodeling process that occurs after a heart attack. Injectable hydrogels are a treatment option because they may replace the damaged extracellular matrix and, in addition, can be administered minimally invasively. Reactive oxygen species generated by ischemia-reperfusion damage can limit the therapeutic efficacy of injectable hydrogels. In order to overcome this limitation, grape seed proanthocyanidins were incorporated as antioxidant compounds into a thermosensitive myocardial extracellular matrix hydrogel in this study. For the fabrication of the hydrogel, the extracellular matrix obtained by decellularization of porcine myocardium was solubilized through enzymatic digestion, and the proanthocyanidins were incorporated. After exposing this extracellular matrix solution to 37 °C, it self-assembled into a hydrogel with a porous structure. According to the physicochemical and biological evaluation, the coupling of proanthocyanidins in the hydrogel has a positive effect on the antioxidant capacity, gelation kinetics, in vitro degradation, and cardiomyocyte viability, indicating that the hydrogel coupled with this type of antioxidants represents a promising alternative for potential application in post-infarction myocardial regeneration. Furthermore, this study proposes the best concentrations of proanthocyanidins that resulted in the hydrogels for future studies in cardiac tissue engineering.
Collapse
Affiliation(s)
- José Luis Hidalgo-Vicelis
- Laboratory of Immunotherapy and Tissue Engineering, Department of Cellular and Tissue Biology, Faculty of Medicine, National Autonomous University of Mexico, Av. Universidad 3000, Copilco Universidad, Coyoacán, Ciudad de México 04510, Mexico; (J.L.H.-V.); (A.R.R.-C.); (B.H.-T.); (G.P.-Z.); (K.J.-Y.)
| | - Angélica Raquel Rivera-Contreras
- Laboratory of Immunotherapy and Tissue Engineering, Department of Cellular and Tissue Biology, Faculty of Medicine, National Autonomous University of Mexico, Av. Universidad 3000, Copilco Universidad, Coyoacán, Ciudad de México 04510, Mexico; (J.L.H.-V.); (A.R.R.-C.); (B.H.-T.); (G.P.-Z.); (K.J.-Y.)
| | - Beatriz Hernández-Téllez
- Laboratory of Immunotherapy and Tissue Engineering, Department of Cellular and Tissue Biology, Faculty of Medicine, National Autonomous University of Mexico, Av. Universidad 3000, Copilco Universidad, Coyoacán, Ciudad de México 04510, Mexico; (J.L.H.-V.); (A.R.R.-C.); (B.H.-T.); (G.P.-Z.); (K.J.-Y.)
| | - Gabriela Piñón-Zárate
- Laboratory of Immunotherapy and Tissue Engineering, Department of Cellular and Tissue Biology, Faculty of Medicine, National Autonomous University of Mexico, Av. Universidad 3000, Copilco Universidad, Coyoacán, Ciudad de México 04510, Mexico; (J.L.H.-V.); (A.R.R.-C.); (B.H.-T.); (G.P.-Z.); (K.J.-Y.)
| | - Katia Jarquín-Yáñez
- Laboratory of Immunotherapy and Tissue Engineering, Department of Cellular and Tissue Biology, Faculty of Medicine, National Autonomous University of Mexico, Av. Universidad 3000, Copilco Universidad, Coyoacán, Ciudad de México 04510, Mexico; (J.L.H.-V.); (A.R.R.-C.); (B.H.-T.); (G.P.-Z.); (K.J.-Y.)
| | - Tatiana Fiordelisio-Coll
- Laboratory of Comparative Neuroendocrinology, Department of Biology, Faculty of Sciences, National Autonomous University of Mexico, Av. Universidad 3000, Copilco Universidad, Coyoacán, Ciudad de México 04510, Mexico;
| | - José Manuel Saniger-Blesa
- Group of Nanostructured Supports, Department of Micro and Nanotechnologies, Institute of Applied Sciences and Technology, National Autonomous University of Mexico, Av. Universidad 3000, Copilco Universidad, Coyoacán, Ciudad de México 04510, Mexico;
| | - Gertrudis Hortensia González-Gómez
- Laboratory of Functional Biophysics, Department of Physics, Faculty of Sciences, National Autonomous University of Mexico, Av. Universidad 3000, Copilco Universidad, Coyoacán, Ciudad de México 04510, Mexico; (G.H.G.-G.); (M.A.F.-N.)
| | - María Alicia Falcón-Neri
- Laboratory of Functional Biophysics, Department of Physics, Faculty of Sciences, National Autonomous University of Mexico, Av. Universidad 3000, Copilco Universidad, Coyoacán, Ciudad de México 04510, Mexico; (G.H.G.-G.); (M.A.F.-N.)
| | - María Margarita Canales-Martínez
- Laboratory of Pharmacognosy, Unit of Biotechnology and Prototypes, Faculty of Higher Studies Iztacala, National Autonomous University of Mexico, Avenida de los Barrios 1, Los Reyes Iztacala, Tlalnepantla de Baz 54090, Estado de México, Mexico;
| | - Andrés Eliú Castell-Rodríguez
- Laboratory of Immunotherapy and Tissue Engineering, Department of Cellular and Tissue Biology, Faculty of Medicine, National Autonomous University of Mexico, Av. Universidad 3000, Copilco Universidad, Coyoacán, Ciudad de México 04510, Mexico; (J.L.H.-V.); (A.R.R.-C.); (B.H.-T.); (G.P.-Z.); (K.J.-Y.)
| |
Collapse
|
3
|
Ali A, Kurome M, Kessler B, Kemter E, Wolf E. What Genetic Modifications of Source Pigs Are Essential and Sufficient for Cell, Tissue, and Organ Xenotransplantation? Transpl Int 2024; 37:13681. [PMID: 39697899 PMCID: PMC11652200 DOI: 10.3389/ti.2024.13681] [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: 08/21/2024] [Accepted: 11/22/2024] [Indexed: 12/20/2024]
Abstract
Xenotransplantation of porcine organs has made remarkable progress towards clinical application. A key factor has been the generation of genetically multi-modified source pigs for xenotransplants, protected against immune rejection and coagulation dysregulation. While efficient gene editing tools and multi-cistronic expression cassettes facilitate sophisticated and complex genetic modifications with multiple gene knockouts and protective transgenes, an increasing number of independently segregating genetic units complicates the breeding of the source pigs. Therefore, an optimal combination of essential genetic modifications may be preferable to extensive editing of the source pigs. Here, we discuss the prioritization of genetic modifications to achieve long-term survival and function of xenotransplants and summarise the genotypes that have been most successful for xenogeneic heart, kidney, and islet transplantation. Specific emphasis is given to the choice of the breed/genetic background of the source pigs. Moreover, multimodal deep phenotyping of porcine organs after xenotransplantation into human decedents will be discussed as a strategy for selecting essential genetic modifications of the source pigs. In addition to germ-line gene editing, some of these modifications may also be induced during organ preservation/perfusion, as demonstrated recently by the successful knockdown of swine leukocyte antigens in porcine lungs during ex vivo perfusion.
Collapse
Affiliation(s)
- Asghar Ali
- Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany
- Center for Innovative Medical Models (CiMM), LMU Munich, Oberschleißheim, Germany
| | - Mayuko Kurome
- Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany
- Center for Innovative Medical Models (CiMM), LMU Munich, Oberschleißheim, Germany
| | - Barbara Kessler
- Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany
- Center for Innovative Medical Models (CiMM), LMU Munich, Oberschleißheim, Germany
| | - Elisabeth Kemter
- Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany
- Center for Innovative Medical Models (CiMM), LMU Munich, Oberschleißheim, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Eckhard Wolf
- Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany
- Center for Innovative Medical Models (CiMM), LMU Munich, Oberschleißheim, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| |
Collapse
|
4
|
Peterson L, Yacoub MH, Ayares D, Yamada K, Eisenson D, Griffith BP, Mohiuddin MM, Eyestone W, Venter JC, Smolenski RT, Rothblatt M. Physiological basis for xenotransplantation from genetically modified pigs to humans. Physiol Rev 2024; 104:1409-1459. [PMID: 38517040 PMCID: PMC11390123 DOI: 10.1152/physrev.00041.2023] [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: 10/26/2023] [Revised: 03/06/2024] [Accepted: 03/14/2024] [Indexed: 03/23/2024] Open
Abstract
The collective efforts of scientists over multiple decades have led to advancements in molecular and cellular biology-based technologies including genetic engineering and animal cloning that are now being harnessed to enhance the suitability of pig organs for xenotransplantation into humans. Using organs sourced from pigs with multiple gene deletions and human transgene insertions, investigators have overcome formidable immunological and physiological barriers in pig-to-nonhuman primate (NHP) xenotransplantation and achieved prolonged pig xenograft survival. These studies informed the design of Revivicor's (Revivicor Inc, Blacksburg, VA) genetically engineered pigs with 10 genetic modifications (10 GE) (including the inactivation of 4 endogenous porcine genes and insertion of 6 human transgenes), whose hearts and kidneys have now been studied in preclinical human xenotransplantation models with brain-dead recipients. Additionally, the first two clinical cases of pig-to-human heart xenotransplantation were recently performed with hearts from this 10 GE pig at the University of Maryland. Although this review focuses on xenotransplantation of hearts and kidneys, multiple organs, tissues, and cell types from genetically engineered pigs will provide much-needed therapeutic interventions in the future.
Collapse
Affiliation(s)
- Leigh Peterson
- United Therapeutics Corporation, Silver Spring, Maryland, United States
| | | | - David Ayares
- United Therapeutics Corporation, Silver Spring, Maryland, United States
| | - Kazuhiko Yamada
- Department of Surgery, Division of Transplantation, Johns Hopkins Medicine, Baltimore, Maryland, United States
| | - Daniel Eisenson
- Department of Surgery, Division of Transplantation, Johns Hopkins Medicine, Baltimore, Maryland, United States
| | - Bartley P Griffith
- University of Maryland Medical Center, Baltimore, Maryland, United States
| | | | - Willard Eyestone
- United Therapeutics Corporation, Silver Spring, Maryland, United States
| | - J Craig Venter
- J. Craig Venter Institute, Rockville, Maryland, United States
| | | | - Martine Rothblatt
- United Therapeutics Corporation, Silver Spring, Maryland, United States
| |
Collapse
|
5
|
Cooper DKC, Kobayashi T. Xenotransplantation experiments in brain-dead human subjects-A critical appraisal. Am J Transplant 2024; 24:520-525. [PMID: 38158188 DOI: 10.1016/j.ajt.2023.12.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 12/19/2023] [Accepted: 12/20/2023] [Indexed: 01/03/2024]
Abstract
Brain-dead human subjects (decedents) were recently introduced as a potential preclinical experimental model in xenotransplantation. Brain death is associated with major pathophysiological changes, eg, structural injury and cell infiltration in vital organs, and major hormonal, metabolic, inflammatory, and hemodynamic changes. In 2 of the 3 initial experiments, the design of the experiments resulted in little or no new information becoming available. In the third, the experiment was unfortunately unsuccessful as neither of the 2 pig kidneys transplanted into the decedent functioned adequately. Failure may well have been associated with the effects of brain death, but an immune/inflammatory response to the xenograft could not be excluded. Subsequently, 2 further pig kidney transplants and 2 pig heart transplants have been carried out in human decedents, but again the data obtained do not add much to what is already known. In view of the profound changes that take place during and after brain death, it may prove difficult to determine whether graft failure or dysfunction results from the effects of brain death or from an immune/inflammatory response to the xenograft. A major concern is that, if the results are confusing, they may impact decisions relating to the introduction of clinical xenotransplantation.
Collapse
Affiliation(s)
- David K C Cooper
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA.
| | - Takaaki Kobayashi
- Department of Renal Transplant Surgery, Aichi University School of Medicine, Nagakute, Japan
| |
Collapse
|
6
|
Cooper DKC, Mou L, Bottino R. A brief review of the current status of pig islet xenotransplantation. Front Immunol 2024; 15:1366530. [PMID: 38464515 PMCID: PMC10920266 DOI: 10.3389/fimmu.2024.1366530] [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: 01/06/2024] [Accepted: 02/07/2024] [Indexed: 03/12/2024] Open
Abstract
An estimated 1.5 million Americans suffer from Type I diabetes mellitus, and its incidence is increasing worldwide. Islet allotransplantation offers a treatment, but the availability of deceased human donor pancreases is limited. The transplantation of islets from gene-edited pigs, if successful, would resolve this problem. Pigs are now available in which the expression of the three known xenoantigens against which humans have natural (preformed) antibodies has been deleted, and in which several human 'protective' genes have been introduced. The transplantation of neonatal pig islets has some advantages over that of adult pig islets. Transplantation into the portal vein of the recipient results in loss of many islets from the instant blood-mediated inflammatory reaction (IBMIR) and so the search for an alternative site continues. The adaptive immune response can be largely suppressed by an immunosuppressive regimen based on blockade of the CD40/CD154 T cell co-stimulation pathway, whereas conventional therapy (e.g., based on tacrolimus) is less successful. We suggest that, despite the need for effective immunosuppressive therapy, the transplantation of 'free' islets will prove more successful than that of encapsulated islets. There are data to suggest that, in the absence of rejection, the function of pig islets, though less efficient than human islets, will be sufficient to maintain normoglycemia in diabetic recipients. Pig islets transplanted into immunosuppressed nonhuman primates have maintained normoglycemia for periods extending more than two years, illustrating the potential of this novel form of therapy.
Collapse
Affiliation(s)
- David K. C. Cooper
- Center for Transplantation Sciences, Massachusetts General Hospital/Harvard Medical School, Boston, MA, United States
| | - Lisha Mou
- Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, Guangdong, China
- MetaLife Center, Shenzhen Institute of Translational Medicine, Shenzhen, Guangdong, China
| | - Rita Bottino
- Imagine Islet Center, Imagine Pharma, Pittsburgh, PA, United States
| |
Collapse
|
7
|
Primous NR, Elvin PT, Carter KV, Andrade HL, La Fontaine J, Shibuya N, Biguetti CC. Bioengineered Skin for Diabetic Foot Ulcers: A Scoping Review. J Clin Med 2024; 13:1221. [PMID: 38592047 PMCID: PMC10932123 DOI: 10.3390/jcm13051221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 02/06/2024] [Accepted: 02/09/2024] [Indexed: 04/10/2024] Open
Abstract
Diabetic foot ulcers (DFUs) pose a significant threat to individuals with diabetes mellitus (DM), such as lower limb amputation and severe morbidity. Bioengineered skin substitutes (BSS) are alternatives to traditional interventions for treating DFUs, but their efficacy compared to standard wound care (SWC) or other treatment types, such as allografts, remains unknown. A scoping review of human studies was conducted to identify current approaches in the treatment of DFUs using BSS as compared with other treatment options. Systematic searches in PubMed, Cochrane Library, and Web of Science were conducted to identify comparative studies that enrolled 10 or more patients and evaluated wound healing outcomes (closure, time-to-healing, and area reduction). Database searches isolated articles published from 1 December 2012 to 1 December 2022 and were conducted in accordance with PRISMA-ScR guidelines. The literature search yielded 1312 articles, 24 of which were included for the qualitative analysis. Findings in these studies demonstrated that BSS outperformed SWC in all measured outcomes, suggesting that BSS may be a superior treatment for DFUs. Of the 24 articles, 8 articles compared human amniotic membrane allografts (hAMA) to BSS. Conflicting evidence was observed when comparing BSS and hAMA treatments, highlighting the need for future research.
Collapse
Affiliation(s)
- Nathaniel R. Primous
- Department of Podiatric Medicine, Surgery and Biomechanics, School of Podiatric Medicine, University of Texas Rio Grande Valley, Harlingen, TX 78550, USA; (N.R.P.); (P.T.E.); (H.L.A.); (J.L.F.); (N.S.)
| | - Peter T. Elvin
- Department of Podiatric Medicine, Surgery and Biomechanics, School of Podiatric Medicine, University of Texas Rio Grande Valley, Harlingen, TX 78550, USA; (N.R.P.); (P.T.E.); (H.L.A.); (J.L.F.); (N.S.)
- Department of Biomedical Engineering, University of Texas at Dallas, Dallas, TX 75080, USA
| | - Kathleen V. Carter
- Library, School of Medicine, University of Texas Rio Grande Valley, Harlingen, TX 78550, USA;
| | - Hagner L. Andrade
- Department of Podiatric Medicine, Surgery and Biomechanics, School of Podiatric Medicine, University of Texas Rio Grande Valley, Harlingen, TX 78550, USA; (N.R.P.); (P.T.E.); (H.L.A.); (J.L.F.); (N.S.)
| | - Javier La Fontaine
- Department of Podiatric Medicine, Surgery and Biomechanics, School of Podiatric Medicine, University of Texas Rio Grande Valley, Harlingen, TX 78550, USA; (N.R.P.); (P.T.E.); (H.L.A.); (J.L.F.); (N.S.)
| | - Naohiro Shibuya
- Department of Podiatric Medicine, Surgery and Biomechanics, School of Podiatric Medicine, University of Texas Rio Grande Valley, Harlingen, TX 78550, USA; (N.R.P.); (P.T.E.); (H.L.A.); (J.L.F.); (N.S.)
| | - Claudia C. Biguetti
- Department of Podiatric Medicine, Surgery and Biomechanics, School of Podiatric Medicine, University of Texas Rio Grande Valley, Harlingen, TX 78550, USA; (N.R.P.); (P.T.E.); (H.L.A.); (J.L.F.); (N.S.)
- Department of Biomedical Engineering, University of Texas at Dallas, Dallas, TX 75080, USA
| |
Collapse
|
8
|
Abstract
End-stage organ failure can result from various preexisting conditions and occurs in patients of all ages, and organ transplantation remains its only treatment. In recent years, extensive research has been done to explore the possibility of transplanting animal organs into humans, a process referred to as xenotransplantation. Due to their matching organ sizes and other anatomical and physiological similarities with humans, pigs are the preferred organ donor species. Organ rejection due to host immune response and possible interspecies infectious pathogen transmission have been the biggest hurdles to xenotransplantation's success. Use of genetically engineered pigs as tissue and organ donors for xenotransplantation has helped to address these hurdles. Although several preclinical trials have been conducted in nonhuman primates, some barriers still exist and demand further efforts. This review focuses on the recent advances and remaining challenges in organ and tissue xenotransplantation.
Collapse
Affiliation(s)
- Asghar Ali
- Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany; , ,
- Center for Innovative Medical Models (CiMM), LMU Munich, Oberschleißheim, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany
| | - Elisabeth Kemter
- Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany; , ,
- Center for Innovative Medical Models (CiMM), LMU Munich, Oberschleißheim, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany
| | - Eckhard Wolf
- Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany; , ,
- Center for Innovative Medical Models (CiMM), LMU Munich, Oberschleißheim, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany
| |
Collapse
|
9
|
Lim B, Jang MJ, Oh SM, No JG, Lee J, Kim SE, Ock SA, Yun IJ, Kim J, Chee HK, Kim WS, Kang HJ, Cho K, Oh KB, Kim JM. Comparative transcriptome analysis between long- and short-term survival after pig-to-monkey cardiac xenotransplantation reveals differential heart failure development. Anim Cells Syst (Seoul) 2023; 27:234-248. [PMID: 37808548 PMCID: PMC10552608 DOI: 10.1080/19768354.2023.2265150] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 07/27/2023] [Indexed: 10/10/2023] Open
Abstract
Cardiac xenotransplantation is the potential treatment for end-stage heart failure, but the allogenic organ supply needs to catch up to clinical demand. Therefore, genetically-modified porcine heart xenotransplantation could be a potential alternative. So far, pig-to-monkey heart xenografts have been studied using multi-transgenic pigs, indicating various survival periods. However, functional mechanisms based on survival period-related gene expression are unclear. This study aimed to identify the differential mechanisms between pig-to-monkey post-xenotransplantation long- and short-term survivals. Heterotopic abdominal transplantation was performed using a donor CD46-expressing GTKO pig and a recipient cynomolgus monkey. RNA-seq was performed using samples from POD60 XH from monkey and NH from age-matched pigs, D35 and D95. Gene-annotated DEGs for POD60 XH were compared with those for POD9 XH (Park et al. 2021). DEGs were identified by comparing gene expression levels in POD60 XH versus either D35 or D95 NH. 1,804 and 1,655 DEGs were identified in POD60 XH versus D35 NH and POD60 XH versus D95 NH, respectively. Overlapped 1,148 DEGs were annotated and compared with 1,348 DEGs for POD9 XH. Transcriptomic features for heart failure and inhibition of T cell activation were observed in both long (POD60)- and short (POD9)-term survived monkeys. Only short-term survived monkey showed heart remodeling and regeneration features, while long-term survived monkey indicated multi-organ failure by neural and hormonal signaling as well as suppression of B cell activation. Our results reveal differential heart failure development and survival at the transcriptome level and suggest candidate genes for specific signals to control adverse cardiac xenotransplantation effects.
Collapse
Affiliation(s)
- Byeonghwi Lim
- Department of Animal Science and Technology, Chung-Ang University, Anseong, Republic of Korea
| | - Min-Jae Jang
- Department of Animal Science and Technology, Chung-Ang University, Anseong, Republic of Korea
| | - Seung-Mi Oh
- Department of Animal Science and Technology, Chung-Ang University, Anseong, Republic of Korea
| | - Jin Gu No
- Animal Biotechnology Division, National Institute of Animal Science, RDA, Wanju, Republic of Korea
| | - Jungjae Lee
- Department of Animal Science and Technology, Chung-Ang University, Anseong, Republic of Korea
| | - Sang Eun Kim
- Animal Biotechnology Division, National Institute of Animal Science, RDA, Wanju, Republic of Korea
| | - Sun A. Ock
- Animal Biotechnology Division, National Institute of Animal Science, RDA, Wanju, Republic of Korea
| | - Ik Jin Yun
- Departments of Surgery, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul, Republic of Korea
| | - Junseok Kim
- Departments of Thoracic and Cardiovascular Surgery, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul, Republic of Korea
| | - Hyun Keun Chee
- Departments of Thoracic and Cardiovascular Surgery, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul, Republic of Korea
| | - Wan Seop Kim
- Departments of Pathology, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul, Republic of Korea
| | - Hee Jung Kang
- Department of Laboratory Medicine, Hallym University College of Medicine, Hallym University Sacred Heart Hospital, Anyang, Republic of Korea
| | - Kahee Cho
- Primate Organ Transplantation Centre, Genia Inc., Seongnam, Republic of Korea
| | - Keon Bong Oh
- Animal Biotechnology Division, National Institute of Animal Science, RDA, Wanju, Republic of Korea
| | - Jun-Mo Kim
- Department of Animal Science and Technology, Chung-Ang University, Anseong, Republic of Korea
| |
Collapse
|
10
|
Chornenkyy Y, Yamamoto T, Hara H, Stowell SR, Ghiran I, Robson SC, Cooper DKC. Future prospects for the clinical transfusion of pig red blood cells. Blood Rev 2023; 61:101113. [PMID: 37474379 PMCID: PMC10968389 DOI: 10.1016/j.blre.2023.101113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/23/2023] [Accepted: 07/09/2023] [Indexed: 07/22/2023]
Abstract
Transfusion of allogeneic human red blood cell (hRBCs) is limited by supply and compatibility between individual donors and recipients. In situations where the blood supply is constrained or when no compatible RBCs are available, patients suffer. As a result, alternatives to hRBCs that complement existing RBC transfusion strategies are needed. Pig RBCs (pRBCs) could provide an alternative because of their abundant supply, and functional similarities to hRBCs. The ability to genetically modify pigs to limit pRBC immunogenicity and augment expression of human 'protective' proteins has provided major boosts to this research and opens up new therapeutic avenues. Although deletion of expression of xenoantigens has been achieved in genetically-engineered pigs, novel genetic methods are needed to introduce human 'protective' transgenes into pRBCs at the high levels required to prevent hemolysis and extend RBC survival in vivo. This review addresses recent progress and examines future prospects for clinical xenogeneic pRBC transfusion.
Collapse
Affiliation(s)
- Yevgen Chornenkyy
- Department of Pathology, McGaw Medical Center of Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Takayuki Yamamoto
- Center for Transplantation Science, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA; Division of Transplantation, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA.
| | - Hidetaka Hara
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, Yunnan, China
| | - Sean R Stowell
- Joint Program in Transfusion Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ionita Ghiran
- Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, MA, USA
| | - Simon C Robson
- Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, MA, USA
| | - David K C Cooper
- Center for Transplantation Science, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| |
Collapse
|
11
|
Zhao P, Gu L, Gao Y, Pan Z, Liu L, Li X, Zhou H, Yu D, Han X, Qian L, Liu GE, Fang L, Wang Z. Young SINEs in pig genomes impact gene regulation, genetic diversity, and complex traits. Commun Biol 2023; 6:894. [PMID: 37652983 PMCID: PMC10471783 DOI: 10.1038/s42003-023-05234-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 08/09/2023] [Indexed: 09/02/2023] Open
Abstract
Transposable elements (TEs) are a major source of genetic polymorphisms and play a role in chromatin architecture, gene regulatory networks, and genomic evolution. However, their functional role in pigs and contributions to complex traits are largely unknown. We created a catalog of TEs (n = 3,087,929) in pigs and found that young SINEs were predominantly silenced by histone modifications, DNA methylation, and decreased accessibility. However, some transcripts from active young SINEs showed high tissue-specificity, as confirmed by analyzing 3570 RNA-seq samples. We also detected 211,067 dimorphic SINEs in 374 individuals, including 340 population-specific ones associated with local adaptation. Mapping these dimorphic SINEs to genome-wide associations of 97 complex traits in pigs, we found 54 candidate genes (e.g., ANK2 and VRTN) that might be mediated by TEs. Our findings highlight the important roles of young SINEs and provide a supplement for genotype-to-phenotype associations and modern breeding in pigs.
Collapse
Affiliation(s)
- Pengju Zhao
- Hainan Institute, Zhejiang University, Yongyou Industry Park, Yazhou Bay Sci-Tech City, Sanya, 572000, China
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Lihong Gu
- Institute of Animal Science & Veterinary Medicine, Hainan Academy of Agricultural Sciences, No. 14 Xingdan Road, Haikou, 571100, China
| | - Yahui Gao
- Animal Genomics and Improvement Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, USDA, Beltsville, MD, 20705, USA
| | - Zhangyuan Pan
- Department of Animal Science, University of California, Davis, CA, 95616, USA
| | - Lei Liu
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518124, China
| | - Xingzheng Li
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518124, China
| | - Huaijun Zhou
- Department of Animal Science, University of California, Davis, CA, 95616, USA
| | - Dongyou Yu
- Hainan Institute, Zhejiang University, Yongyou Industry Park, Yazhou Bay Sci-Tech City, Sanya, 572000, China
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Xinyan Han
- Hainan Institute, Zhejiang University, Yongyou Industry Park, Yazhou Bay Sci-Tech City, Sanya, 572000, China
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Lichun Qian
- Hainan Institute, Zhejiang University, Yongyou Industry Park, Yazhou Bay Sci-Tech City, Sanya, 572000, China
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - George E Liu
- Animal Genomics and Improvement Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, USDA, Beltsville, MD, 20705, USA.
| | - Lingzhao Fang
- Center for Quantitative Genetics and Genomics, Aarhus University, Aarhus, 8000, Denmark.
| | - Zhengguang Wang
- Hainan Institute, Zhejiang University, Yongyou Industry Park, Yazhou Bay Sci-Tech City, Sanya, 572000, China.
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China.
| |
Collapse
|
12
|
Cooper DK, Hara H. Xenotransplantation-A Basic Science Perspective. KIDNEY360 2023; 4:1147-1149. [PMID: 37265370 PMCID: PMC10476676 DOI: 10.34067/kid.0000000000000173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 05/03/2023] [Indexed: 06/03/2023]
Affiliation(s)
- David K.C. Cooper
- Center for Transplantation Sciences, Department of Surgery, General Hospital/Harvard Medical School, Boston, Massachusetts
| | - Hidetaka Hara
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, Yunnan, China
| |
Collapse
|
13
|
Habibabady Z, McGrath G, Kinoshita K, Maenaka A, Ikechukwu I, Elias GF, Zaletel T, Rosales I, Hara H, Pierson RN, Cooper DKC. Antibody-mediated rejection in xenotransplantation: Can it be prevented or reversed? Xenotransplantation 2023; 30:e12816. [PMID: 37548030 PMCID: PMC11101061 DOI: 10.1111/xen.12816] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 07/19/2023] [Accepted: 07/26/2023] [Indexed: 08/08/2023]
Abstract
Antibody-mediated rejection (AMR) is the commonest cause of failure of a pig graft after transplantation into an immunosuppressed nonhuman primate (NHP). The incidence of AMR compared to acute cellular rejection is much higher in xenotransplantation (46% vs. 7%) than in allotransplantation (3% vs. 63%) in NHPs. Although AMR in an allograft can often be reversed, to our knowledge there is no report of its successful reversal in a pig xenograft. As there is less experience in preventing or reversing AMR in models of xenotransplantation, the results of studies in patients with allografts provide more information. These include (i) depletion or neutralization of serum anti-donor antibodies, (ii) inhibition of complement activation, (iii) therapies targeting B or plasma cells, and (iv) anti-inflammatory therapy. Depletion or neutralization of anti-pig antibody, for example, by plasmapheresis, is effective in depleting antibodies, but they recover within days. IgG-degrading enzymes do not deplete IgM. Despite the expression of human complement-regulatory proteins on the pig graft, inhibition of systemic complement activation may be necessary, particularly if AMR is to be reversed. Potential therapies include (i) inhibition of complement activation (e.g., by IVIg, C1 INH, or an anti-C5 antibody), but some complement inhibitors are not effective in NHPs, for example, eculizumab. Possible B cell-targeted therapies include (i) B cell depletion, (ii) plasma cell depletion, (iii) modulation of B cell activation, and (iv) enhancing the generation of regulatory B and/or T cells. Among anti-inflammatory agents, anti-IL6R mAb and TNF blockers are increasingly being tested in xenotransplantation models, but with no definitive evidence that they reverse AMR. Increasing attention should be directed toward testing combinations of the above therapies. We suggest that treatment with a systemic complement inhibitor is likely to be most effective, possibly combined with anti-inflammatory agents (if these are not already being administered). Ultimately, it may require further genetic engineering of the organ-source pig to resolve the problem entirely, for example, knockout or knockdown of SLA, and/or expression of PD-L1, HLA E, and/or HLA-G.
Collapse
Affiliation(s)
- Zahra Habibabady
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Gannon McGrath
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Kohei Kinoshita
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Akihiro Maenaka
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Ileka Ikechukwu
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Gabriela F. Elias
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Tjasa Zaletel
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Ivy Rosales
- Department of Pathology, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - Hidetaka Hara
- Yunnan Xenotransplantation Engineering Research Center, Yunnan Agricultural University, Kunming, Yunnan, China
| | - Richard N. Pierson
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| | - David K. C. Cooper
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
14
|
Mubarak M. Transitioning of renal transplant pathology from allograft to xenograft and tissue engineering pathology: Are we prepared? World J Transplant 2023; 13:86-95. [PMID: 36968134 PMCID: PMC10037233 DOI: 10.5500/wjt.v13.i3.86] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 12/21/2022] [Accepted: 01/11/2023] [Indexed: 03/16/2023] Open
Abstract
Currently, the most feasible and widely practiced option for patients with end-stage organ failure is the transplantation of part of or whole organs, either from deceased or living donors. However, organ shortage has posed and is still posing a big challenge in this field. Newer options being explored are xenografts and engineered/bioengineered tissues/organs. Already small steps have been taken in this direction and sooner or later, these will become a norm in this field. However, these developments will pose different challenges for the diagnosis and management of problems as compared with traditional allografts. The approach to pathologic diagnosis of dysfunction in these settings will likely be significantly different. Thus, there is a need to increase awareness and prepare transplant diagnosticians to meet this future challenge in the field of xenotransplantation/ regenerative medicine. This review will focus on the current status of transplant pathology and how it will be changed in the future with the emerging scenario of routine xenotransplantation.
Collapse
Affiliation(s)
- Muhammed Mubarak
- Department of Histopathology, Sindh Institute of Urology and Transplantation, Karachi 74200, Sindh, Pakistan
| |
Collapse
|
15
|
Rodger D, Cooper DKC. Kidney xenotransplantation: Future clinical reality or science fiction? Nurs Health Sci 2023; 25:161-170. [PMID: 36335558 PMCID: PMC10124775 DOI: 10.1111/nhs.12994] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 10/10/2022] [Accepted: 11/02/2022] [Indexed: 11/07/2022]
Abstract
There is a global shortage of organs for transplantation and despite many governments making significant changes to their organ donation systems, there are not enough kidneys available to meet the demand. This has led scientists and clinicians to explore alternative means of meeting this organ shortfall. One of the alternatives to human organ transplantation is xenotransplantation, which is the transplantation of organs, tissues, or cells between different species. The resurgence of interest in xenotransplantation and recent scientific breakthroughs suggest that genetically engineered pigs may soon present a realistic alternative as sources of kidneys for clinical transplantation. It is therefore important for healthcare professionals to understand what is involved in xenotransplantation and its future implications for their clinical practices. First, we explore the insufficiency of different organ donation systems to meet the kidney shortage. Second, we provide a background and a summary of the progress made so far in xenotransplantation research. Third, we discuss some of the scientific, technological, ethical, and public health issues associated with xenotransplantation. Finally, we summarize the literature on the attitudes of healthcare professionals toward xenotransplantation.
Collapse
Affiliation(s)
- Daniel Rodger
- Institute of Health and Social Care, School of Allied and Community Health, London South Bank University, London, UK
- Department of Psychological Sciences, Birkbeck, University of London, London, UK
| | - David K. C. Cooper
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
16
|
Cooper DKC, Pierson RN. Milestones on the path to clinical pig organ xenotransplantation. Am J Transplant 2023; 23:326-335. [PMID: 36775767 PMCID: PMC10127379 DOI: 10.1016/j.ajt.2022.12.023] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/20/2022] [Accepted: 12/28/2022] [Indexed: 01/19/2023]
Abstract
Progress in pig organ xenotransplantation has been made largely through (1) genetic engineering of the organ-source pig to protect its tissues from the human innate immune response, and (2) development of an immunosuppressive regimen based on blockade of the CD40/CD154 costimulation pathway to prevent the adaptive immune response. In the 1980s, after transplantation into nonhuman primates (NHPs), wild-type (genetically unmodified) pig organs were rejected within minutes or hours. In the 1990s, organs from pigs expressing a human complement-regulatory protein (CD55) transplanted into NHPs receiving intensive conventional immunosuppressive therapy functioned for days or weeks. When costimulation blockade was introduced in 2000, the adaptive immune response was suppressed more readily. The identification of galactose-α1,3-galactose as the major antigen target for human and NHP anti-pig antibodies in 1991 allowed for deletion of expression of galactose-α1,3-galactose in 2003, extending pig graft survival for up to 6 months. Subsequent gene editing to overcome molecular incompatibilities between the pig and primate coagulation systems proved additionally beneficial. The identification of 2 further pig carbohydrate xenoantigens allowed the production of 'triple-knockout' pigs that are preferred for clinical organ transplantation. These combined advances enabled the first clinical pig heart transplant to be performed and opened the door to formal clinical trials.
Collapse
Affiliation(s)
- David K C Cooper
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA.
| | - Richard N Pierson
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
17
|
Salloum A, Bazzi N, Squires S, Chu T, Benedetto P, Benedetto A. Comparing the application of various engineered xenografts for skin defects: A systematic review. J Cosmet Dermatol 2023; 22:921-931. [PMID: 36409467 DOI: 10.1111/jocd.15517] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 09/23/2022] [Accepted: 10/23/2022] [Indexed: 11/23/2022]
Abstract
INTRODUCTION Xenografts are a now a cornerstone in the management of wound dressings. Promising results were achieved since 1960 in the application of skin substitute for skin defects. OBJECTIVE The objective of this study was to evaluate the efficacy of various xenografts. METHODS A literature research was conducted using the following query: 'Porcine skin dermatology substitute', 'bovine skin dermatology substitute', 'xenograft skin substitute dermatology', 'xenografts skin defect', 'porcine skin defect', 'bovine skin defect'. RESULTS The review yielded 35 articles pertaining to the topic. Main indications for porcine and bovine xenograft application were burn wounds and post-traumatic wounds, respectively. Mean discharge date or length of stay was at the 6th day after porcine application, and the time of graft healing was reported for 33.7% (n = 510) of patients. Promising results were seen with Matriderm and split-thickness skin graft. Most wounds achieved an excellent cosmetic result with full range of motion and a smooth contour appearance. A great variety of tissue substitutes exist, and the choice of graft application should depend on a patient's factors, product availability, wound type, size, and physician's factors. CONCLUSION In summary, xenografts are more economic and affordable but have higher risk of infections compared to allografts.
Collapse
Affiliation(s)
- Antoine Salloum
- Roger Williams Medical Center, Providence, Rhode Island, USA
| | - Nagham Bazzi
- Lebanese University, School of Medicine, Beirut, Lebanon
| | | | - Thomas Chu
- East Virginia Medical School, Norfolk, Virginia, USA
| | - Paul Benedetto
- Dermatologic Surgicenter, Philadelphia, Pennsylvania, USA.,Cleveland Clinic Foundation, Westin, Florida, USA
| | - Anthony Benedetto
- Dermatologic Surgicenter, Philadelphia, Pennsylvania, USA.,University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
18
|
Reichart B, Cooper DKC, Längin M, Tönjes RR, Pierson RN, Wolf E. Cardiac xenotransplantation: from concept to clinic. Cardiovasc Res 2023; 118:3499-3516. [PMID: 36461918 PMCID: PMC9897693 DOI: 10.1093/cvr/cvac180] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 10/17/2022] [Accepted: 10/21/2022] [Indexed: 12/05/2022] Open
Abstract
For many patients with terminal/advanced cardiac failure, heart transplantation is the most effective, durable treatment option, and offers the best prospects for a high quality of life. The number of potentially life-saving donated human organs is far fewer than the population who could benefit from a new heart, resulting in increasing numbers of patients awaiting replacement of their failing heart, high waitlist mortality, and frequent reliance on interim mechanical support for many of those deemed among the best candidates but who are deteriorating as they wait. Currently, mechanical assist devices supporting left ventricular or biventricular heart function are the only alternative to heart transplant that is in clinical use. Unfortunately, the complication rate with mechanical assistance remains high despite advances in device design and patient selection and management, and the quality of life of the patients even with good outcomes is only moderately improved. Cardiac xenotransplantation from genetically multi-modified (GM) organ-source pigs is an emerging new option as demonstrated by the consistent long-term success of heterotopic (non-life-supporting) abdominal and life-supporting orthotopic porcine heart transplantation in baboons, and by a recent 'compassionate use' transplant of the heart from a GM pig with 10 modifications into a terminally ill patient who survived for 2 months. In this review, we discuss pig heart xenotransplantation as a concept, including pathobiological aspects related to immune rejection, coagulation dysregulation, and detrimental overgrowth of the heart, as well as GM strategies in pigs to prevent or minimize these problems. Additional topics discussed include relevant results of heterotopic and orthotopic heart transplantation experiments in the pig-to-baboon model, microbiological and virologic safety concepts, and efficacy requirements for initiating formal clinical trials. An adequate regulatory and ethical framework as well as stringent criteria for the selection of patients will be critical for the safe clinical development of cardiac xenotransplantation, which we expect will be clinically tested during the next few years.
Collapse
Affiliation(s)
- Bruno Reichart
- Walter Brendel Centre for Experimental Medicine, Ludwig-Maximilians-Universität München, Munich 81377, Germany
| | - David K C Cooper
- Center for Transplantation Sciences, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02129, USA
- Division of Cardiac Surgery, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02114, USA
| | - Matthias Längin
- Department of Anaesthesiology, University Hospital, Ludwig-Maximilians-Universität München, Munich 81377, Germany
| | - Ralf R Tönjes
- Division of Medical Biotechnology, Paul-Ehrlich-Institute, Langen 63225, Germany
| | - Richard N Pierson
- Center for Transplantation Sciences, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02129, USA
- Division of Cardiac Surgery, Department of Surgery, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02114, USA
| | - Eckhard Wolf
- Gene Centre and Centre for Innovative Medical Models (CiMM), Ludwig-Maximilians-Universität München, Munich 81377, Germany
| |
Collapse
|
19
|
Ajima K, Tsuda N, Takaki T, Furusako S, Matsumoto S, Shinohara K, Yamashita Y, Amano S, Oyama C, Shimoda M. A porcine islet-encapsulation device that enables long-term discordant xenotransplantation in immunocompetent diabetic mice. CELL REPORTS METHODS 2023; 3:100370. [PMID: 36814843 PMCID: PMC9939365 DOI: 10.1016/j.crmeth.2022.100370] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 08/29/2022] [Accepted: 11/23/2022] [Indexed: 12/24/2022]
Abstract
Islet transplantation is an effective treatment for type 1 diabetes (T1D). However, a shortage of donors and the need for immunosuppressants are major issues. The ideal solution is to develop a source of insulin-secreting cells and an immunoprotective method. No bioartificial pancreas (BAP) devices currently meet all of the functions of long-term glycemic control, islet survival, immunoprotection, discordant xenotransplantation feasibility, and biocompatibility. We developed a device in which porcine islets were encapsulated in a highly stable and permeable hydrogel and a biocompatible immunoisolation membrane. Discordant xenotransplantation of the device into diabetic mice improved glycemic control for more than 200 days. Glycemic control was also improved in new diabetic mice "relay-transplanted" with the device after its retrieval. The easily retrieved devices exhibited almost no adhesion or fibrosis and showed sustained insulin secretion even after the two xenotransplantations. This device has the potential to be a useful BAP for T1D.
Collapse
Affiliation(s)
- Kumiko Ajima
- Pancreatic Islet Cell Transplantation Project, Research Institute National Center for Global Health and Medicine, 1-21-1 Toyama Shinjuku-ku, Tokyo 162-8655, Japan
| | - Naoto Tsuda
- Biomaterials Business Division, Mochida Pharmaceutical Co., Ltd., 722 Uenohara, Jimba, Gotemba, Shizuoka 412-8524, Japan
| | - Tadashi Takaki
- Pancreatic Islet Cell Transplantation Project, Research Institute National Center for Global Health and Medicine, 1-21-1 Toyama Shinjuku-ku, Tokyo 162-8655, Japan
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
- Takeda-CiRA Joint Program (T-CiRA), 2-26-1 Muraoka-higashi, Fujisawa-shi, Kanagawa 251-8555, Japan
| | - Shoji Furusako
- Biomaterials Business Division, Mochida Pharmaceutical Co., Ltd., 1-7 Yotsuya, Shinjuku-ku, Tokyo 160-8515, Japan
| | - Shigeki Matsumoto
- Biomaterials Business Division, Mochida Pharmaceutical Co., Ltd., 722 Uenohara, Jimba, Gotemba, Shizuoka 412-8524, Japan
| | - Koya Shinohara
- Pancreatic Islet Cell Transplantation Project, Research Institute National Center for Global Health and Medicine, 1-21-1 Toyama Shinjuku-ku, Tokyo 162-8655, Japan
| | - Yzumi Yamashita
- Pancreatic Islet Cell Transplantation Project, Research Institute National Center for Global Health and Medicine, 1-21-1 Toyama Shinjuku-ku, Tokyo 162-8655, Japan
| | - Sayaka Amano
- Pancreatic Islet Cell Transplantation Project, Research Institute National Center for Global Health and Medicine, 1-21-1 Toyama Shinjuku-ku, Tokyo 162-8655, Japan
| | - Chinatsu Oyama
- Communal Laboratory, Research Institute National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo 162-8655, Japan
| | - Masayuki Shimoda
- Pancreatic Islet Cell Transplantation Project, Research Institute National Center for Global Health and Medicine, 1-21-1 Toyama Shinjuku-ku, Tokyo 162-8655, Japan
| |
Collapse
|
20
|
Kato A, Go T, Otsuki Y, Yokota N, Soo CS, Misaki N, Yajima T, Yokomise H. Perpendicular implantation of porcine trachea extracellular matrix for enhanced xenogeneic scaffold surface epithelialization in a canine model. Front Surg 2023; 9:1089403. [PMID: 36713663 PMCID: PMC9877415 DOI: 10.3389/fsurg.2022.1089403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 12/26/2022] [Indexed: 01/13/2023] Open
Abstract
Objective The availability of clinically applied medical materials in thoracic surgery remains insufficient, especially materials for treating tracheal defects. Herein, the potential of porcine extracellular matrix (P-ECM) as a new airway reconstruction material was explored by xenotransplanting it into a canine trachea. Methods P-ECM was first transplanted into the buttocks of Narc Beagle dogs (n = 3) and its overall immuno-induced effects were evaluated. Subsequently, nine dogs underwent surgery to create a tracheal defect that was 1 × 2 cm. In group A, the P-ECM was implanted parallel to the tracheal axis (n = 3), whereas in group B the P-ECM was implanted perpendicular to the tracheal axis (n = 6). The grafts were periodically observed by bronchoscopy and evaluated postoperatively at 1 and 3 months through macroscopic and microscopic examinations. Immunosuppressants were not administered. Statistical evaluation was performed for Bronchoscopic stenosis rate, graft epithelialization rate, shrinkage rate and ECM live-implantation rate. Results No sign of P-ECM rejection was observed after its implantation in the buttocks. Bronchoscopic findings showed no improvement concerning stenosis in group A until 3 months after surgery; epithelialization of the graft site was not evident, and the ECM site appeared scarred and faded. In contrast, stenosis gradually improved in group B, with continuous epithelium within the host tissues and P-ECM. Histologically, the graft site contracted longitudinally and no epithelialization was observed in group A, whereas full epithelialization was observed on the P-ECM in group B. No sign of cartilage regeneration was confirmed in both groups. No statistically significant differences were found in bronchoscopic stenosis rate, shrinkage rate and ECM live-implantation rate, but graft epithelialization rate showed a statistically significant difference (G-A; sporadic (25%) 3, vs. G-B; full covered (100%) 3; p = 0.047). Conclusions P-ECM can support full re-epithelialization without chondrocyte regeneration, with perpendicular implantation facilitating epithelialization of the ECM. Our results showed that our decellularized tracheal matrix holds clinical potential as a biological xenogeneic material for airway defect repair.
Collapse
|
21
|
Geng Z, Zhang Q, Li T, Huang T, Wang H, Zhou Q, Deng S, Zhao Y, Li Y, Cheng C, Gonelle-Gispert C, Buhler LH, Wang Y. Advantages of the retroperitoneal retrocolic space as the transplant site for encapsulated xenogeneic islets. Xenotransplantation 2023; 30:e12787. [PMID: 36454040 DOI: 10.1111/xen.12787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 10/14/2022] [Accepted: 10/18/2022] [Indexed: 12/03/2022]
Abstract
OBJECTIVE Islet allotransplantation has demonstrated improved clinical outcomes using the hepatic portal vein as the standard infusion method. However, the current implantation site is not ideal due to the short-term thrombotic and long-term immune destruction. Meanwhile, the shortage of human organ donors further limits its application. To find a new strategy, we tested a new polymer combination for islet encapsulation and transplantation. Meanwhile, we explored a new site for xenogeneic islet transplantation in mice. METHOD We synthesized a hydrogel combining alginate plus poly-ethylene-imine (Alg/PEI) for the encapsulation of rat, neonatal porcine, and human islets. Transplantation was performed into the retroperitoneal retro-colic space of diabetic mice. Control mice received free islets under the kidney capsule or encapsulated islets into the peritoneum. The biochemical indexes were measured, and the transplanted islets were harvested for immunohistochemical staining of insulin and glucagon. RESULTS Mice receiving encapsulated rat, porcine and human islets transplanted into the retroperitoneal space maintained normoglycemia for a median of 275, 145.5, and 146 days, respectively. In contrast, encapsulated xenogeneic islets transplanted into the peritoneum, maintained function for a median of 61, 95.5, and 82 days, respectively. Meanwhile, xenogeneic islets transplanted free into the kidney capsule lost their function within 3 days after transplantation. Immunohistochemical staining of encapsulated rat, porcine and human islets, retrieved from the retroperitoneal space, allowed to distinguish morphological normal insulin expressing β- and glucagon expressing α-cells at 70, 60, and 100 days post-transplant, respectively. CONCLUSION Transplantation of Alg/PEI encapsulated xenogeneic islets into the retroperitoneal space provides a valuable new implantation strategy for the treatment of type 1 diabetes.
Collapse
Affiliation(s)
- Zhen Geng
- Health Management Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Qi Zhang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Ting Li
- Department of Rheumatology, Wenjiang District People's Hospital, Chengdu, China
| | - Ting Huang
- Department of Breast Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Hailian Wang
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Institute of Organ Transplantation, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Qiao Zhou
- Department of Rheumatology and Immunology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Shaoping Deng
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Institute of Organ Transplantation, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yanshuang Zhao
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yanjiao Li
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Chunming Cheng
- Department of Radiation Oncology, James Comprehensive Cancer Center and College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | | | - Leo H Buhler
- Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Yi Wang
- Department of Critical Care Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 610072, China
| |
Collapse
|
22
|
Lei T, Chen L, Wang K, Du S, Gonelle-Gispert C, Wang Y, Buhler LH. Genetic engineering of pigs for xenotransplantation to overcome immune rejection and physiological incompatibilities: The first clinical steps. Front Immunol 2022; 13:1031185. [PMID: 36561750 PMCID: PMC9766364 DOI: 10.3389/fimmu.2022.1031185] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022] Open
Abstract
Xenotransplantation has the potential to solve the shortfall of human organ donors. Genetically modified pigs have been considered as potential animal donors for human xenotransplantation and have been widely used in preclinical research. The genetic modifications aim to prevent the major species-specific barriers, which include humoral and cellular immune responses, and physiological incompatibilities such as complement and coagulation dysfunctions. Genetically modified pigs can be created by deleting several pig genes related to the synthesis of various pig specific antigens or by inserting human complement- and coagulation-regulatory transgenes. Finally, in order to reduce the risk of infection, genes related to porcine endogenous retroviruses can be knocked down. In this review, we focus on genetically modified pigs and comprehensively summarize the immunological mechanism of xenograft rejection and recent progress in preclinical and clinical studies. Overall, both genetically engineered pig-based xenografts and technological breakthroughs in the biomedical field provide a promising foundation for pig-to-human xenotransplantation in the future.
Collapse
Affiliation(s)
- Tiantian Lei
- Department of Pharmacy, Women and Children’s Hospital of Chongqing Medical University, Chongqing Health Center for Women and Children, Chongqing, China
| | - Lin Chen
- Department of Pharmacy, Women and Children’s Hospital of Chongqing Medical University, Chongqing Health Center for Women and Children, Chongqing, China
| | - Kejing Wang
- Department of Pharmacy, Women and Children’s Hospital of Chongqing Medical University, Chongqing Health Center for Women and Children, Chongqing, China
| | - Suya Du
- Department of Clinical Pharmacy, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Sichuan, China
| | | | - Yi Wang
- Department of Critical Care Medicine, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Leo H. Buhler
- Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
23
|
Chaban R, Habibabady Z, Hassanein W, Connolly MR, Burdorf L, Redding E, Laird C, Ranek J, Braileanu G, Sendil S, Cheng X, Sun W, O’Neill NA, Kuravi K, Hurh S, Ayares DL, Azimzadeh AM, Pierson RN. Knock-out of N-glycolylneuraminic acid attenuates antibody-mediated rejection in xenogenically perfused porcine lungs. Xenotransplantation 2022; 29:e12784. [PMID: 36250568 PMCID: PMC11093624 DOI: 10.1111/xen.12784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 07/27/2022] [Accepted: 09/13/2022] [Indexed: 01/15/2023]
Abstract
BACKGROUND Antibody-mediated rejection has long been known to be one of the major organ failure mechanisms in xenotransplantation. In addition to the porcine α1,3-galactose (α1,3Gal) epitope, N-Glycolylneuraminic acid (Neu5Gc), a sialic acid, has been identified as an important porcine antigen against which most humans have pre-formed antibodies. Here we evaluate GalTKO.hCD46 lungs with an additional cytidine monophospho-N-acetylneuraminic acid hydroxylase (CMAH) gene knock-out (Neu5GcKO) in a xenogeneic ex vivo perfusion model METHODS: Eleven GalTKO.hCD46.Neu5GcKO pig lungs were perfused for up to 6 h with fresh heparinized human blood. Six of them were treated with histamine (H) blocker famotidine and 1-thromboxane synthase inhibitor Benzylimidazole (BIA) and five were left untreated. GalTKO.hCD46 lungs without Neu5GcKO (n = 18: eight untreated and 10 BIA+H treated) served as a reference. Functional parameters, blood, and tissue samples were collected at pre-defined time points throughout the perfusion RESULTS: All but one Neu5GcKO organs maintained adequate blood oxygenation and "survived" until elective termination at 6 h whereas two reference lungs failed before elective termination at 4 h. Human anti-Neu5Gc antibody serum levels decreased during the perfusion of GalTKO.hCD46 lungs by flow cytometry (∼40% IgM, 60% IgG), whereas antibody levels in Neu5GcKO lung perfusions did not fall (IgM p = .007; IgG p < .001). Thromboxane elaboration, thrombin generation, and histamine levels were significantly reduced with Neu5GcKO lungs compared to reference in the untreated groups (p = .007, .005, and .037, respectively); treatment with BIA+H masked these changes. Activation of platelets, measured as CD62P expression on circulating platelets, was lower in Neu5GcKO experiments compared to reference lungs (p = .023), whereas complement activation (as C3a rise in plasma) was not altered. MCP-1 and lactotransferin level elevations were blunted in Neu5GcKO lung perfusions (p = .007 and .032, respectively). Pulmonary vascular resistance (PVR) rise was significantly attenuated and delayed in untreated GalTKO.hCD46.Neu5GcKO lungs in comparison to the untreated GalTKO.hCD46 lungs (p = .003) CONCLUSION: Additional Neu5GcKO in GalTKO.hCD46 lungs significantly reduces parameters associated with antibody-mediated inflammation and activation of the coagulation cascade. Knock-out of the Neu5Gc sialic acid should be beneficial to reduce innate immune antigenicity of porcine lungs in future human recipients.
Collapse
Affiliation(s)
- Ryan Chaban
- Center for Transplantation Sciences and Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Department of Cardiovascular Surgery, University Hospital of Mainz, Mainz, Germany
| | - Zahra Habibabady
- Center for Transplantation Sciences and Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Wessam Hassanein
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Margaret R. Connolly
- Center for Transplantation Sciences and Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Lars Burdorf
- Center for Transplantation Sciences and Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Revivicor, Inc., Blacksburg, Virgina, USA
| | - Emily Redding
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Christopher Laird
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Jolene Ranek
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Gheorghe Braileanu
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Selin Sendil
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Xiangfei Cheng
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Wenji Sun
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Natalie A. O’Neill
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | - Sunghoon Hurh
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Korea
| | | | - Agnes M. Azimzadeh
- Center for Transplantation Sciences and Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Richard N. Pierson
- Center for Transplantation Sciences and Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
24
|
Rodger D, Hurst DJ. Mathieu Jaboulay's (1860-1913) contribution to xenotransplantation. Xenotransplantation 2022; 29:e12765. [PMID: 35695309 PMCID: PMC9787852 DOI: 10.1111/xen.12765] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 05/23/2022] [Indexed: 12/30/2022]
Abstract
Mathieu Jaboulay (1860-1913) was a professor of clinical surgery in Lyon, France who is best known for his development of vascular anastomosis and for conducting the first reported renal xenotransplantation experiments on humans, using pig and goat kidneys to treat end-stage renal failure in 1906. His insights and pioneering techniques contributed significantly to allotransplantation and contemporary attempts at xenotransplantation. He is also credited with inventing several surgical instruments and novel surgical techniques that continue to influence vascular, general, and urological surgery to this day. However, this article will focus specifically on his notable contributions to xenotransplantation research and surgery.
Collapse
Affiliation(s)
- Daniel Rodger
- Institute of Health and Social CareLondon South Bank UniversityLondonUK
| | - Daniel J. Hurst
- Rowan University School of Osteopathic MedicineStratfordNew JerseyUSA
| |
Collapse
|
25
|
Functional Reconstruction of Denervated Muscle by Xenotransplantation of Neural Cells from Porcine to Rat. Int J Mol Sci 2022; 23:ijms23158773. [PMID: 35955906 PMCID: PMC9368947 DOI: 10.3390/ijms23158773] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/02/2022] [Accepted: 08/05/2022] [Indexed: 11/23/2022] Open
Abstract
Neural cell transplantation targeting peripheral nerves is a potential treatment regime for denervated muscle atrophy. This study aimed to develop a new therapeutic technique for intractable muscle atrophy by the xenotransplantation of neural stem cells derived from pig fetuses into peripheral nerves. In this study, we created a denervation model using neurotomy in nude rats and transplanted pig-fetus-derived neural stem cells into the cut nerve stump. Three months after transplantation, the survival of neural cells, the number and area of regenerated axons, and the degree of functional recovery by electrical stimulation of peripheral nerves were compared among the gestational ages (E 22, E 27, E 45) of the pigs. Transplanted neural cells were engrafted at all ages. Functional recovery by electric stimulation was observed at age E 22 and E 27. This study shows that the xenotransplantation of fetal porcine neural stem cells can restore denervated muscle function. When combined with medical engineering, this technology can help in developing a new therapy for paralysis.
Collapse
|
26
|
Raza SS, Hara H, Cleveland DC, Cooper DKC. The potential of genetically engineered pig heart transplantation in infants with complex congenital heart disease. Pediatr Transplant 2022; 26:e14260. [PMID: 35233893 PMCID: PMC10124767 DOI: 10.1111/petr.14260] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 02/15/2022] [Accepted: 02/16/2022] [Indexed: 12/16/2022]
Abstract
Despite advances in surgical and medical techniques, complex congenital heart disease in neonates and infants continues to be associated with significant mortality and morbidity. More than 500 infants in the USA are placed on the cardiac transplantation wait-list annually. However, there remains a critical shortage of deceased human donor organs for transplantation with a median wait-time of 4 months. Hence, infant mortality on the heart transplant wait-list in the USA is higher than for any other solid organ transplant group. Orthotopic transplantation of a pig heart as a bridge to allotransplantation might offer the best prospect of long-term survival of these patients. In recent years, there have been several advances in genetic engineering of pigs to mitigate the vigorous antibody-mediated rejection of a pig heart transplanted into a nonhuman primate. In this review, we briefly highlight (i) the history of clinical heart xenotransplantation, (ii) current advances and techniques of genetically engineering pigs, (iii) the current status of pig orthotopic cardiac graft survival in nonhuman primates, and (iv) progress toward pursuing clinical trials of cardiac xenotransplantation. Ultimately, we argue that pig heart xenotransplantation should initially be used as a bridge to cardiac allotransplantation in neonates and infants.
Collapse
Affiliation(s)
- Syed Sikandar Raza
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Hidetaka Hara
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - David C Cleveland
- Department of Cardiothoracic Surgery, Children's of Alabama, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - David K C Cooper
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
27
|
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
|
28
|
Moya-Jódar M, Coppiello G, Rodríguez-Madoz JR, Abizanda G, Barlabé P, Vilas-Zornoza A, Ullate-Agote A, Luongo C, Rodríguez-Tobón E, Navarro-Serna S, París-Oller E, Oficialdegui M, Carvajal-Vergara X, Ordovás L, Prósper F, García-Vázquez FA, Aranguren XL. One-Step In Vitro Generation of ETV2-Null Pig Embryos. Animals (Basel) 2022; 12:ani12141829. [PMID: 35883376 PMCID: PMC9311767 DOI: 10.3390/ani12141829] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 07/05/2022] [Accepted: 07/14/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary One of the latest goals in regenerative medicine is to use pluripotent stem cells to generate whole organs in vivo through the blastocyst complementation technique. This method consists of the microinjection of pluripotent stem cells into preimplantation embryos that have been genetically modified to ablate the development of a target organ. By taking advantage of the spatiotemporal clues present in the developing embryo, pluripotent stem cells are able to colonize the empty developmental niche and create the missing organ. Combining human pluripotent stem cells with genetically engineered pig embryos, it would be possible to obtain humanized organs that could be used for transplantation, and, therefore, solve the worldwide issue of insufficient availability of transplantable organs. As endothelial cells play a critical role in xenotransplantation rejection in all organs, in this study, we optimized a protocol to generate a vascular-disabled preimplantation pig embryo using the CRISPR/Cas9 system. This protocol could be used to generate avascular embryos for blastocyst complementation experiments and work towards the generation of rejection-free humanized organs in pigs. Abstract Each year, tens of thousands of people worldwide die of end-stage organ failure due to the limited availability of organs for use in transplantation. To meet this clinical demand, one of the last frontiers of regenerative medicine is the generation of humanized organs in pigs from pluripotent stem cells (PSCs) via blastocyst complementation. For this, organ-disabled pig models are needed. As endothelial cells (ECs) play a critical role in xenotransplantation rejection in every organ, we aimed to produce hematoendothelial-disabled pig embryos targeting the master transcription factor ETV2 via CRISPR-Cas9-mediated genome modification. In this study, we designed five different guide RNAs (gRNAs) against the DNA-binding domain of the porcine ETV2 gene, which were tested on porcine fibroblasts in vitro. Four out of five guides showed cleavage capacity and, subsequently, these four guides were microinjected individually as ribonucleoprotein complexes (RNPs) into one-cell-stage porcine embryos. Next, we combined the two gRNAs that showed the highest targeting efficiency and microinjected them at higher concentrations. Under these conditions, we significantly improved the rate of biallelic mutation. Hence, here, we describe an efficient one-step method for the generation of hematoendothelial-disabled pig embryos via CRISPR-Cas9 microinjection in zygotes. This model could be used in experimentation related to the in vivo generation of humanized organs.
Collapse
Affiliation(s)
- Marta Moya-Jódar
- Program of Regenerative Medicine, Centre for Applied Medical Research (CIMA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), University of Navarra, 31008 Pamplona, Spain; (M.M.-J.); (G.C.); (J.R.R.-M.); (G.A.); (P.B.); (A.U.-A.); (X.C.-V.); (F.P.)
| | - Giulia Coppiello
- Program of Regenerative Medicine, Centre for Applied Medical Research (CIMA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), University of Navarra, 31008 Pamplona, Spain; (M.M.-J.); (G.C.); (J.R.R.-M.); (G.A.); (P.B.); (A.U.-A.); (X.C.-V.); (F.P.)
| | - Juan Roberto Rodríguez-Madoz
- Program of Regenerative Medicine, Centre for Applied Medical Research (CIMA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), University of Navarra, 31008 Pamplona, Spain; (M.M.-J.); (G.C.); (J.R.R.-M.); (G.A.); (P.B.); (A.U.-A.); (X.C.-V.); (F.P.)
| | - Gloria Abizanda
- Program of Regenerative Medicine, Centre for Applied Medical Research (CIMA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), University of Navarra, 31008 Pamplona, Spain; (M.M.-J.); (G.C.); (J.R.R.-M.); (G.A.); (P.B.); (A.U.-A.); (X.C.-V.); (F.P.)
| | - Paula Barlabé
- Program of Regenerative Medicine, Centre for Applied Medical Research (CIMA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), University of Navarra, 31008 Pamplona, Spain; (M.M.-J.); (G.C.); (J.R.R.-M.); (G.A.); (P.B.); (A.U.-A.); (X.C.-V.); (F.P.)
| | - Amaia Vilas-Zornoza
- Advanced Genomics Laboratory, Program of Hemato-Oncology, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain;
| | - Asier Ullate-Agote
- Program of Regenerative Medicine, Centre for Applied Medical Research (CIMA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), University of Navarra, 31008 Pamplona, Spain; (M.M.-J.); (G.C.); (J.R.R.-M.); (G.A.); (P.B.); (A.U.-A.); (X.C.-V.); (F.P.)
- Advanced Genomics Laboratory, Program of Hemato-Oncology, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain;
| | - Chiara Luongo
- Department of Physiology, Veterinary School, International Excellence Campus for Higher Education and Research (Campus Mare Nostrum), University of Murcia, 30100 Murcia, Spain; (C.L.); (E.R.-T.); (S.N.-S.); (E.P.-O.)
- Institute for Biomedical Research of Murcia, IMIB-Arrixaca, 30100 Murcia, Spain
| | - Ernesto Rodríguez-Tobón
- Department of Physiology, Veterinary School, International Excellence Campus for Higher Education and Research (Campus Mare Nostrum), University of Murcia, 30100 Murcia, Spain; (C.L.); (E.R.-T.); (S.N.-S.); (E.P.-O.)
- Institute for Biomedical Research of Murcia, IMIB-Arrixaca, 30100 Murcia, Spain
| | - Sergio Navarro-Serna
- Department of Physiology, Veterinary School, International Excellence Campus for Higher Education and Research (Campus Mare Nostrum), University of Murcia, 30100 Murcia, Spain; (C.L.); (E.R.-T.); (S.N.-S.); (E.P.-O.)
- Institute for Biomedical Research of Murcia, IMIB-Arrixaca, 30100 Murcia, Spain
| | - Evelyne París-Oller
- Department of Physiology, Veterinary School, International Excellence Campus for Higher Education and Research (Campus Mare Nostrum), University of Murcia, 30100 Murcia, Spain; (C.L.); (E.R.-T.); (S.N.-S.); (E.P.-O.)
- Institute for Biomedical Research of Murcia, IMIB-Arrixaca, 30100 Murcia, Spain
| | | | - Xonia Carvajal-Vergara
- Program of Regenerative Medicine, Centre for Applied Medical Research (CIMA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), University of Navarra, 31008 Pamplona, Spain; (M.M.-J.); (G.C.); (J.R.R.-M.); (G.A.); (P.B.); (A.U.-A.); (X.C.-V.); (F.P.)
| | - Laura Ordovás
- Aragon Agency for Research and Development (ARAID), 50018 Zaragoza, Spain;
- Biomedical Signal Interpretation and Computational Simulation (BSICoS), Institute of Engineering Research (I3A), University of Zaragoza & Instituto de Investigación Sanitaria (IIS), 50018 Zaragoza, Spain
| | - Felipe Prósper
- Program of Regenerative Medicine, Centre for Applied Medical Research (CIMA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), University of Navarra, 31008 Pamplona, Spain; (M.M.-J.); (G.C.); (J.R.R.-M.); (G.A.); (P.B.); (A.U.-A.); (X.C.-V.); (F.P.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain
- Department of Hematology and Cell Therapy, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Francisco Alberto García-Vázquez
- Department of Physiology, Veterinary School, International Excellence Campus for Higher Education and Research (Campus Mare Nostrum), University of Murcia, 30100 Murcia, Spain; (C.L.); (E.R.-T.); (S.N.-S.); (E.P.-O.)
- Institute for Biomedical Research of Murcia, IMIB-Arrixaca, 30100 Murcia, Spain
- Correspondence: (F.A.G.-V.); (X.L.A.)
| | - Xabier L. Aranguren
- Program of Regenerative Medicine, Centre for Applied Medical Research (CIMA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), University of Navarra, 31008 Pamplona, Spain; (M.M.-J.); (G.C.); (J.R.R.-M.); (G.A.); (P.B.); (A.U.-A.); (X.C.-V.); (F.P.)
- Correspondence: (F.A.G.-V.); (X.L.A.)
| |
Collapse
|
29
|
Human Endothelial Cell Seeding in Partially Decellularized Kidneys. BIOMED RESEARCH INTERNATIONAL 2022; 2022:9018074. [PMID: 35872850 PMCID: PMC9300320 DOI: 10.1155/2022/9018074] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/25/2022] [Accepted: 06/15/2022] [Indexed: 11/20/2022]
Abstract
The excessive demand for organ transplants has promoted the development of strategies that increase the supply of immune compatible organs, such as xenotransplantation of genetically modified pig organs and the generation of bioartificial organs. We describe a method for the partial replacement of rat endothelial cells for human endothelial cells in a rat's kidney, obtaining as a final result a rat-human bioartificial kidney. First, in order to maintain parenchymal epithelial cells and selectively eliminate rat endothelial cells, three methods were evaluated in which different solutions were perfused through the renal artery: 0.1% sodium dodecyl sulfate (SDS), 0.01% SDS, and hyperosmolar solutions of sucrose. Then, partially decellularized kidneys were recellularized with human endothelial cells and finally transplanted in an anesthetized rat. The solution of 0.1% SDS achieved the highest vascular decellularization but with high degree of damage in the parenchyma side. On the contrary, 0.01% SDS and hyperosmolar solutions achieved a partial degree of endothelial decellularization. TUNEL assays reveal that hyperosmolar solutions maintained a better epithelial cell viability contrasting with 0.01% SDS. Partially decellularized kidneys were then recellularized with human endothelial cells. Histological analysis showed endothelial cells attached in almost all the vascular bed. Recellularized kidney was transplanted in an anesthetized rat. After surgery, recellularized kidney achieved complete perfusion, and urine was produced for at least 90 min posttransplant. Histological analysis showed endothelial cells attached in almost all the vascular bed. Therefore, endothelial decellularization of grafts and recellularization with human endothelial cells derived from transplant recipients can be a feasible method with the aim to reduce the damage of the grafts.
Collapse
|
30
|
Hansen-Estruch C, Porrett PM, Kumar V, Locke JE. The science of xenotransplantation for nephrologists. Curr Opin Nephrol Hypertens 2022; 31:387-393. [PMID: 35703221 DOI: 10.1097/mnh.0000000000000800] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
PURPOSE OF REVIEW The field of xenotransplantation has seen remarkable progress since its inception with recent preclinical trials in human recipients pushing kidney xenotransplantation one-step closer to clinical reality. In this review, we update practicing clinicians on recent advances in kidney xenotransplantation given the proximity of clinical trials in humans. RECENT FINDINGS Early studies in the field established the physiologic basis of xenotransplantation and suggested that the pig kidney will support human physiology. Genetic engineering of source pigs has greatly reduced the immunogenicity of kidney grafts, and studies in nonhuman primates have demonstrated the viability of kidney xenotransplants for months after transplantation. Finally, a recent study in a novel preclinical human model demonstrated that key findings in NHP experiments are generalizable to humans, namely, the absence of hyperacute rejection. SUMMARY Overall, it appears that critical physiologic, immunologic and technical barriers to implementation of clinical trials in humans have been overcome.
Collapse
Affiliation(s)
| | - Paige M Porrett
- Comprehensive Transplant Institute, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Vineeta Kumar
- Comprehensive Transplant Institute, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jayme E Locke
- Comprehensive Transplant Institute, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
31
|
Ko N, Shim J, Kim HJ, Lee Y, Park JK, Kwak K, Lee JW, Jin DI, Kim H, Choi K. A desirable transgenic strategy using GGTA1 endogenous promoter-mediated knock-in for xenotransplantation model. Sci Rep 2022; 12:9611. [PMID: 35688851 PMCID: PMC9187654 DOI: 10.1038/s41598-022-13536-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 05/25/2022] [Indexed: 11/09/2022] Open
Abstract
Pig-to-human organ transplantation is a feasible solution to resolve the shortage of organ donors for patients that wait for transplantation. To overcome immunological rejection, which is the main hurdle in pig-to-human xenotransplantation, various engineered transgenic pigs have been developed. Ablation of xeno-reactive antigens, especially the 1,3-Gal epitope (GalT), which causes hyperacute rejection, and insertion of complement regulatory protein genes, such as hCD46, hCD55, and hCD59, and genes to regulate the coagulation pathway or immune cell-mediated rejection may be required for an ideal xenotransplantation model. However, the technique for stable and efficient expression of multi-transgenes has not yet been settled to develop a suitable xenotransplantation model. To develop a stable and efficient transgenic system, we knocked-in internal ribosome entry sites (IRES)-mediated transgenes into the α 1,3-galactosyltransferase (GGTA1) locus so that expression of these transgenes would be controlled by the GGTA1 endogenous promoter. We constructed an IRES-based polycistronic hCD55/hCD39 knock-in vector to target exon4 of the GGTA1 gene. The hCD55/hCD39 knock-in vector and CRISPR/Cas9 to target exon4 of the GGTA1 gene were co-transfected into white yucatan miniature pig fibroblasts. After transfection, hCD39 expressed cells were sorted by FACS. Targeted colonies were verified using targeting PCR and FACS analysis, and used as donors for somatic cell nuclear transfer. Expression of GalT, hCD55, and hCD39 was analyzed by FACS and western blotting. Human complement-mediated cytotoxicity and human antibody binding assays were conducted on peripheral blood mononuclear cells (PBMCs) and red blood cells (RBCs), and deposition of C3 by incubation with human complement serum and platelet aggregation were analyzed in GGTA1 knock-out (GTKO)/CD55/CD39 pig cells. We obtained six targeted colonies with high efficiency of targeting (42.8% of efficiency). Selected colony and transgenic pigs showed abundant expression of targeted genes (hCD55 and hCD39). Knocked-in transgenes were expressed in various cell types under the control of the GGTA1 endogenous promoter in GTKO/CD55/CD39 pig and IRES was sufficient to express downstream expression of the transgene. Human IgG and IgM binding decreased in GTKO/CD55/CD39 pig and GTKO compared to wild-type pig PBMCs and RBCs. The human complement-mediated cytotoxicity of RBCs and PBMCs decreased in GTKO/CD55/CD39 pig compared to cells from GTKO pig. C3 was also deposited less in GTKO/CD55/CD39 pig cells than wild-type pig cells. The platelet aggregation was delayed by hCD39 expression in GTKO/CD55/CD39 pig. In the current study, knock-in into the GGTA1 locus and GGTA1 endogenous promoter-mediated expression of transgenes are an appropriable strategy for effective and stable expression of multi-transgenes. The IRES-based polycistronic transgene vector system also caused sufficient expression of both hCD55 and hCD39. Furthermore, co-transfection of CRISPR/Cas9 and the knock-in vector not only increased the knock-in efficiency but also induced null for GalT by CRISPR/Cas9-mediated double-stranded break of the target site. As shown in human complement-mediated lysis and human antibody binding to GTKO/CD55/CD39 transgenic pig cells, expression of hCD55 and hCD39 with ablation of GalT prevents an effective immunological reaction in vitro. As a consequence, our technique to produce multi-transgenic pigs could improve the development of a suitable xenotransplantation model, and the GTKO/CD55/CD39 pig developed could prolong the survival of pig-to-primate xenotransplant recipients.
Collapse
Affiliation(s)
- Nayoung Ko
- Department of Transgenic Animal Research, Optipharm, Inc., Chungcheongbuk-do, Cheongju-si, 28158, Republic of Korea
- Department of Animal Science and Biotechnology, Chungnam National University, Daejeon, Republic of Korea
| | - Joohyun Shim
- Department of Transgenic Animal Research, Optipharm, Inc., Chungcheongbuk-do, Cheongju-si, 28158, Republic of Korea
- Department of Animal Science and Biotechnology, Chungnam National University, Daejeon, Republic of Korea
| | - Hyoung-Joo Kim
- Department of Transgenic Animal Research, Optipharm, Inc., Chungcheongbuk-do, Cheongju-si, 28158, Republic of Korea
| | - Yongjin Lee
- Department of Transgenic Animal Research, Optipharm, Inc., Chungcheongbuk-do, Cheongju-si, 28158, Republic of Korea
| | - Jae-Kyung Park
- Department of Transgenic Animal Research, Optipharm, Inc., Chungcheongbuk-do, Cheongju-si, 28158, Republic of Korea
| | - Kyungmin Kwak
- Department of Transgenic Animal Research, Optipharm, Inc., Chungcheongbuk-do, Cheongju-si, 28158, Republic of Korea
| | - Jeong-Woong Lee
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology, Dajeon, Republic of Korea
| | - Dong-Il Jin
- Department of Animal Science and Biotechnology, Chungnam National University, Daejeon, Republic of Korea
| | - Hyunil Kim
- Department of Transgenic Animal Research, Optipharm, Inc., Chungcheongbuk-do, Cheongju-si, 28158, Republic of Korea
| | - Kimyung Choi
- Department of Transgenic Animal Research, Optipharm, Inc., Chungcheongbuk-do, Cheongju-si, 28158, Republic of Korea.
| |
Collapse
|
32
|
Jiang Z, Fu M, Zhu D, Wang X, Li N, Ren L, He J, Yang G. Genetically modified immunomodulatory cell-based biomaterials in tissue regeneration and engineering. Cytokine Growth Factor Rev 2022; 66:53-73. [PMID: 35690567 DOI: 10.1016/j.cytogfr.2022.05.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 05/24/2022] [Indexed: 11/25/2022]
Abstract
To date, the wide application of cell-based biomaterials in tissue engineering and regeneration is remarkably hampered by immune rejection. Reducing the immunogenicity of cell-based biomaterials has become the latest direction in biomaterial research. Recently, genetically modified cell-based biomaterials with immunomodulatory genes have become a feasible solution to the immunogenicity problem. In this review, recent advances and future challenges of genetically modified immunomodulatory cell-based biomaterials are elaborated, including fabrication approaches, mechanisms of common immunomodulatory genes, application and, more importantly, current preclinical and clinical advances. The fabrication approaches can be categorized into commonly used (e.g., virus transfection) and newly developed approaches. The immunomodulatory mechanisms of representative genes involve complicated cell signaling pathways and metabolic activities. Wide application in curing multiple end-term diseases and replacing lifelong immunosuppressive therapy in multiple cell and organ transplantation models is demonstrated. Most significantly, practices of genetically modified organ transplantation have been conducted on brain-dead human decedent and even on living patients after a series of experiments on nonhuman primates. Nevertheless, uncertain biosecurity, nonspecific effects and overlooked personalization of current genetically modified immunomodulatory cell-based biomaterials are shortcomings that remain to be overcome.
Collapse
Affiliation(s)
- Zhiwei Jiang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center of Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Mengdie Fu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center of Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Danji Zhu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center of Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Xueting Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center of Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Na Li
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center of Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Lingfei Ren
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center of Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Jin He
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center of Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China
| | - Guoli Yang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center of Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310000, China.
| |
Collapse
|
33
|
Ganchiku Y, Riella LV. Pig-to-human kidney transplantation using brain-dead donors as recipients: One giant leap, or only one small step for transplantkind? Xenotransplantation 2022; 29:e12748. [PMID: 35616243 DOI: 10.1111/xen.12748] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 04/28/2022] [Accepted: 04/28/2022] [Indexed: 12/11/2022]
Abstract
Pig kidney xenotransplantation is increasingly regarded as a realistic solution to the current shortage of human organ donors for patients with end-stage organ failure. Recently, the news of three pig-to-human transplantation cases has awakened public interest. Notably, the case by the Alabama team reported detailed and important findings for the xenotransplantation field. Using a genetically modified pig, two porcine kidneys were transplanted into a brain-dead recipient. They applied several approaches established in the preclinical NHP study, including gene-edited pig kidney graft and preoperative laboratory inspection such as crossmatching and infection screening. The pig-to-human kidney xenotransplantation had no unexpected events during surgery or evidence of hyperacute rejection. Unfortunately, the grafts did not work appropriately, and the study had to be terminated due to the decompensation of the recipient. While this study demonstrated the outstanding achievement in this research area, it also revealed remaining gaps to move xenotransplantation to the clinic. While brain-dead human recipients could reinforce the compatibility achievements of gene-edited pigs in NHP, their pro-inflammatory and pro-coagulant environment, in combination with short-duration of experiments will limit the assessment of kidney function, infection and rejection risk post-transplant, in particular antibody-mediated rejection. The use of successful immunosuppressive protocols of non-human primates xenotransplant experiments including anti-CD154 antibody will be critical to maximize the success in the first in-human trials.
Collapse
Affiliation(s)
- Yoshikazu Ganchiku
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Leonardo V Riella
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.,Division of Nephrology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
34
|
Lee H, Park EM, Ko N, Choi K, Oh KB, Kang HJ. Effect of Factor H on Complement Alternative Pathway Activation in Human Serum Remains on Porcine Cells Lacking N-Glycolylneuraminic Acid. Front Immunol 2022; 13:859261. [PMID: 35444661 PMCID: PMC9014258 DOI: 10.3389/fimmu.2022.859261] [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: 01/21/2022] [Accepted: 03/14/2022] [Indexed: 11/13/2022] Open
Abstract
Background Triple knockout (TKO) donor pigs lacking alpha-1,3-galactose (Gal), N-glycolylneuraminic acid (Neu5Gc), and Sd(a) expressions were developed to improve the clinical success of xenotransplantation. Neu5Gc, a sialic acid expressed on cell surfaces, recruits factor H to protect cells from attack by the complement system. Lack of Neu5Gc expression may cause unwanted complement activation, abrogating the potential benefit of gene-modified donor pigs. To investigate whether TKO porcine cells display increased susceptibility to complement activation in human serum, pathway-specific complement activation, apoptosis, and human platelet aggregation by porcine cells were compared between alpha-1,3-galactosyltransferase gene-knockout (GTKO) and TKO porcine cells. Methods Primary porcine peripheral blood mononuclear cells (pPBMCs) and endothelial cells (pECs) from GTKO and TKO pigs were used. Cells were incubated in human serum diluted in gelatin veronal buffer (GVB++) or Mg++-EGTA GVB, and C3 deposition and apoptotic changes in these cells were measured by flow cytometry. C3 deposition levels were also measured after incubating these cells in 10% human serum supplemented with human factor H. Platelet aggregation in human platelet-rich plasma containing GTKO or TKO pECs was analyzed. Results The C3 deposition level in GTKO pPBMCs or pECs in GVB++ was significantly higher than that of TKO pPBMCs or pECs, respectively, but C3 deposition levels in Mg++-EGTA-GVB were comparable between them. The addition of factor H into the porcine cell suspension in 10% serum in Mg++ -EGTA-GVB inhibited C3 deposition in a dose-dependent manner, and the extent of inhibition by factor H was similar between GTKO and TKO porcine cells. The percentage of late apoptotic cells in porcine cell suspension in GVB++ increased with the addition of human serum, of which the net increase was significantly less in TKO pPBMCs than in GTKO pPBMCs. Finally, the lag time of platelet aggregation in recalcified human plasma was significantly prolonged in the presence of TKO pECs compared to that in the presence of GTKO pECs. Conclusion TKO genetic modification protects porcine cells from serum-induced complement activation and apoptotic changes, and delays recalcification-induced human platelet aggregation. It does not hamper factor H recruitment on cell surfaces, allowing the suppression of alternative complement pathway activation.
Collapse
Affiliation(s)
- Haneulnari Lee
- Department of Laboratory Medicine, Hallym University College of Medicine, Anyang, South Korea
| | - Eun Mi Park
- Department of Laboratory Medicine, Hallym University College of Medicine, Anyang, South Korea
| | - Nayoung Ko
- Department of Transgenic Animal Research, Optipharm Inc., Cheongju, South Korea
| | - Kimyung Choi
- Department of Transgenic Animal Research, Optipharm Inc., Cheongju, South Korea
| | - Keon Bong Oh
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration (RDA), Wanju, South Korea
| | - Hee Jung Kang
- Department of Laboratory Medicine, Hallym University College of Medicine, Anyang, South Korea
| |
Collapse
|
35
|
Signaling cascades in the failing heart and emerging therapeutic strategies. Signal Transduct Target Ther 2022; 7:134. [PMID: 35461308 PMCID: PMC9035186 DOI: 10.1038/s41392-022-00972-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/13/2022] [Accepted: 03/20/2022] [Indexed: 12/11/2022] Open
Abstract
Chronic heart failure is the end stage of cardiac diseases. With a high prevalence and a high mortality rate worldwide, chronic heart failure is one of the heaviest health-related burdens. In addition to the standard neurohormonal blockade therapy, several medications have been developed for chronic heart failure treatment, but the population-wide improvement in chronic heart failure prognosis over time has been modest, and novel therapies are still needed. Mechanistic discovery and technical innovation are powerful driving forces for therapeutic development. On the one hand, the past decades have witnessed great progress in understanding the mechanism of chronic heart failure. It is now known that chronic heart failure is not only a matter involving cardiomyocytes. Instead, chronic heart failure involves numerous signaling pathways in noncardiomyocytes, including fibroblasts, immune cells, vascular cells, and lymphatic endothelial cells, and crosstalk among these cells. The complex regulatory network includes protein-protein, protein-RNA, and RNA-RNA interactions. These achievements in mechanistic studies provide novel insights for future therapeutic targets. On the other hand, with the development of modern biological techniques, targeting a protein pharmacologically is no longer the sole option for treating chronic heart failure. Gene therapy can directly manipulate the expression level of genes; gene editing techniques provide hope for curing hereditary cardiomyopathy; cell therapy aims to replace dysfunctional cardiomyocytes; and xenotransplantation may solve the problem of donor heart shortages. In this paper, we reviewed these two aspects in the field of failing heart signaling cascades and emerging therapeutic strategies based on modern biological techniques.
Collapse
|
36
|
Li X, Wang Y, Yang H, Dai Y. Liver and Hepatocyte Transplantation: What Can Pigs Contribute? Front Immunol 2022; 12:802692. [PMID: 35095885 PMCID: PMC8795512 DOI: 10.3389/fimmu.2021.802692] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 12/30/2021] [Indexed: 12/25/2022] Open
Abstract
About one-fifth of the population suffers from liver diseases in China, meaning that liver disorders are prominent causative factors relating to the Chinese mortality rate. For patients with end-stage liver diseases such as hepatocellular carcinoma or acute liver diseases with life-threatening liver dysfunction, allogeneic liver transplantation is the only life-saving treatment. Hepatocyte transplantation is a promising alternative for patients with acute liver failure or those considered high risk for major surgery, particularly for the bridge-to-transplant period. However, the lack of donors has become a serious global problem. The clinical application of porcine xenogeneic livers and hepatocytes remains a potential solution to alleviate the donor shortage. Pig grafts of xenotransplantation play roles in providing liver support in recipients, together with the occurrence of rejection, thrombocytopenia, and blood coagulation dysfunction. In this review, we present an overview of the development, potential therapeutic impact, and remaining barriers in the clinical application of pig liver and hepatocyte xenotransplantation to humans and non-human primates. Donor pigs with optimized genetic modification combinations and highly effective immunosuppressive regimens should be further explored to improve the outcomes of xenogeneic liver and hepatocyte transplantation.
Collapse
Affiliation(s)
- Xiaoxue Li
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, China
| | - Ying Wang
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, China.,Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Haiyuan Yang
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, China.,Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Yifan Dai
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, Nanjing, China.,Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China.,State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China.,Shenzhen Xenotransplantation Medical Engineering Research and Development Center, Institute of Translational Medicine, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, China
| |
Collapse
|
37
|
Ma Y, Xie B, Guo J, Chen Y, Zhong M, Lin Q, Hua J, Zhong J, Luo X, Yan G, Dai H, Qi Z. Leflunomide Inhibits rat-to-Mouse Cardiac Xenograft Rejection by Suppressing Adaptive Immune Cell Response and NF-κB Signaling Activation. Cell Transplant 2021; 30:9636897211054503. [PMID: 34814739 PMCID: PMC8647224 DOI: 10.1177/09636897211054503] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Xenotransplantation is a potential solution for the severe shortage of human donor organs and tissues. The generation of humanized animal models attenuates strong innate immune responses, such as complement-mediated hyperacute rejection. However, acute vascular rejection and cell mediated rejection remain primary barriers to xenotransplantation, which limits its clinical application. In this study, we systematically investigated the immunosuppressive effect of LEF using a rat-to-mouse heart xenotransplantation model. SD rat xenogeneic hearts were transplanted into C57BL/6 mice, and survived 34.5 days after LEF treatment. In contrast, BALB/c allogeneic hearts were transplanted into C57BL/6 mice, and survived 31 days after LEF treatment. Compared to normal saline treatment, LEF treatment decreased xenoreactive T cells and CD19+ B cells in recipient splenocytes. Most importantly, LEF treatment protected myocardial cells by decreasing xenoreactive T and B cell infiltration, inflammatory gene expression, and IgM deposition in grafts. In vivo assays revealed that LEF treatment eliminated xenoreactive and alloreactive T and B lymphocytes by suppressing the activation of the NF-κB signaling pathway. Taken together, these observations complement the evidence supporting the potential use of LEF in xenotransplantation.
Collapse
Affiliation(s)
- Yunhan Ma
- Fujian Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, China.,School of Medicine, Xiamen University, Xiamen, China.,Department of Clinical Sciences, Malmö, Lund University, Malmö, Sweden.,Yunhan Ma and Baiyi Xie contributed equally to this work
| | - Baiyi Xie
- Department of Urology Surgery, Ruikang Hospital affiliated to Guangxi University of Chinese Medicine, Nanning, China.,Yunhan Ma and Baiyi Xie contributed equally to this work
| | - Junjun Guo
- School of Medicine, Xiamen University, Xiamen, China
| | - Yingyu Chen
- Fujian Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, China.,School of Medicine, Xiamen University, Xiamen, China
| | - Mengya Zhong
- School of Medicine, Xiamen University, Xiamen, China
| | - Qingru Lin
- Fujian Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, China.,School of Medicine, Xiamen University, Xiamen, China
| | - Jianyu Hua
- Fujian Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, China.,School of Medicine, Xiamen University, Xiamen, China
| | - Jiaying Zhong
- Fujian Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, China.,School of Medicine, Xiamen University, Xiamen, China
| | - Xuewei Luo
- Medicinal College, Guangxi University, Nanning, China
| | - Guoliang Yan
- Fujian Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, China.,School of Medicine, Xiamen University, Xiamen, China
| | - Helong Dai
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, China.,Clinical Research Center for Organ Transplantation in Hunan Province, Changsha, China.,Clinical Immunology Center, Central South University, Changsha, China
| | - Zhongquan Qi
- Fujian Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, China.,Medicinal College, Guangxi University, Nanning, China
| |
Collapse
|
38
|
Zhao P, Zheng X, Yu Y, Hou Z, Diao C, Wang H, Kang H, Ning C, Li J, Feng W, Wang W, Liu GE, Li B, Smith J, Chamba Y, Liu JF. Mining Unknown Porcine Protein Isoforms by Tissue-based Map of Proteome Enhances Pig Genome Annotation. GENOMICS, PROTEOMICS & BIOINFORMATICS 2021; 19:772-786. [PMID: 33631433 PMCID: PMC9170766 DOI: 10.1016/j.gpb.2021.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 09/05/2019] [Accepted: 11/29/2019] [Indexed: 11/29/2022]
Abstract
A lack of the complete pig proteome has left a gap in our knowledge of the pig genome and has restricted the feasibility of using pigs as a biomedical model. In this study, we developed a tissue-based proteome map using 34 major normal pig tissues. A total of 5841 unknown protein isoforms were identified and systematically characterized, including 2225 novel protein isoforms, 669 protein isoforms from 460 genes symbolized beginning with LOC, and 2947 protein isoforms without clear NCBI annotation in the current pig reference genome. These newly identified protein isoforms were functionally annotated through profiling the pig transcriptome with high-throughput RNA sequencing of the same pig tissues, further improving the genome annotation of the corresponding protein-coding genes. Combining the well-annotated genes that have parallel expression pattern and subcellular witness, we predicted the tissue-related subcellularlocations and potential functions for these unknown proteins. Finally, we mined 3081 orthologous genes for 52.7% of unknown protein isoforms across multiple species, referring to 68 KEGG pathways as well as 23 disease signaling pathways. These findings provide valuable insights and a rich resource for enhancing studies of pig genomics and biology, as well as biomedical model application to human medicine.
Collapse
Affiliation(s)
- Pengju Zhao
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Xianrui Zheng
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Ying Yu
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Zhuocheng Hou
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Chenguang Diao
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Haifei Wang
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Huimin Kang
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Chao Ning
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Junhui Li
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Wen Feng
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Wen Wang
- Center for Ecological and Environmental Sciences, Northwestern Polytechnical University, Xi'an 710072, China
| | - George E Liu
- Animal Genomics and Improvement Laboratory, Beltsville Agricultural Research Center, U.S. Department of Agriculture, Beltsville, MD 20705, USA
| | - Bugao Li
- Department of Animal Sciences and Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, China
| | - Jacqueline Smith
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK
| | - Yangzom Chamba
- Tibet Agriculture and Animal Husbandry College, Linzhi 860000, China
| | - Jian-Feng Liu
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
39
|
Cooper DKC, Hara H. "You cannot stay in the laboratory forever"*: Taking pig kidney xenotransplantation from the laboratory to the clinic. EBioMedicine 2021; 71:103562. [PMID: 34517284 PMCID: PMC8441149 DOI: 10.1016/j.ebiom.2021.103562] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 08/02/2021] [Accepted: 08/16/2021] [Indexed: 11/18/2022] Open
Abstract
Progress in life-supporting kidney transplantation in the genetically-engineered pig-to-nonhuman primate model has been encouraging, with pig kidneys sometimes supporting life for > 1 year. What steps need to be taken by (i) the laboratory team, and (ii) the clinical team to prepare for the first clinical trial? The major topics include (i) what currently-available genetic modifications are optimal to reduce the possibility of graft rejection, (ii) what immunosuppressive therapeutic regimen is optimal, and (iii) what steps need to be taken to minimize the risk of transfer of an infectious microorganism with the graft. We suggest that patients who are unlikely to live long enough to receive a kidney from a deceased human donor would benefit from the opportunity of a period of dialysis-free support by a pig kidney, and the experience gained would enable xenotransplantation to progress much more rapidly than if we remain in the laboratory.
Collapse
Affiliation(s)
- David K C Cooper
- Xenotransplantation Program, Division of Transplantation, Department of Surgery, University of Alabama at Birmingham, 752 Lyons-Harrison Research Building, 701 19th Street South, Birmingham, AL 35294, USA.
| | - Hidetaka Hara
- Xenotransplantation Program, Division of Transplantation, Department of Surgery, University of Alabama at Birmingham, 752 Lyons-Harrison Research Building, 701 19th Street South, Birmingham, AL 35294, USA
| |
Collapse
|
40
|
Ladowski JM, Houp J, Hauptfeld-Dolejsek V, Javed M, Hara H, Cooper DKC. Aspects of histocompatibility testing in xenotransplantation. Transpl Immunol 2021; 67:101409. [PMID: 34015463 PMCID: PMC8197754 DOI: 10.1016/j.trim.2021.101409] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 05/14/2021] [Indexed: 12/16/2022]
Abstract
Xenotransplantation, using genetically-modified pigs for clinical organ transplantation, is a solution to the organ shortage. The biggest barrier to clinical implementation is the antigenicity of pig cells. Humans possess preformed antibody to pig cells that initiate antibody-mediated rejection of pig organs in primates. Advances in genetic engineering have led to the development of a pig lacking the three known glycan xenoantigens (triple-knockout [TKO] pigs). A significant number of human sera demonstrate no antibody binding to TKO pig cells. As a result of the TKO pig's low antigen expression, survival of life-supporting pig organs in immunosuppressed nonhuman primates has significantly increased, and hope has been renewed for clinical trials of xenotransplantation. It is important to understand the context in which xenotransplantation's predecessor, allotransplantation, has been successful, and the steps needed for the success of xenotransplantation. Successful allotransplantation has been based on two main immunological approaches - (i) adequate immunosuppressive therapy, and (ii) careful histocompatibility matching. In vivo studies suggest that the available immunosuppressive regimens are adequate to suppress the human anti-pig cellular response. Methods to evaluate and screen patients for the first clinical xenotransplantation trial are the next challenge. The goal of this review is to summarize the history of histocompatibility testing, and the available tools that can be utilized to determine xenograft histocompatibility.
Collapse
Affiliation(s)
- Joseph M Ladowski
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Julie Houp
- Histocompatibility Laboratory, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Mariyam Javed
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hidetaka Hara
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - David K C Cooper
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
41
|
Gao C, Sow WT, Wang Y, Wang Y, Yang D, Lee BH, Matičić D, Fang L, Li H, Zhang C. Hydrogel composite scaffolds with an attenuated immunogenicity component for bone tissue engineering applications. J Mater Chem B 2021; 9:2033-2041. [PMID: 33587079 DOI: 10.1039/d0tb02588g] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Xenogeneic bones are potential templates for bone regeneration. In this study, decellularized porcine bone powder with attenuated immunogenicity was incorporated into a photocurable hydrogel, gelatin methacryloyl (GelMA), to obtain scaffolds with good mechanical properties for bone tissue engineering. The decellularized bone powder (DCB)-GelMA hybrid scaffolds had higher compressive strength and stiffness values when the DCB content was increased. In vitro evaluations revealed the biocompatibility of these scaffolds. The scaffolds could induce human bone marrow mesenchymal stem cells (hMSCs) to undergo osteogenic differentiation even in the absence of an induction medium. The efficiency of the scaffolds for bone regeneration applications was further evaluated using an in vivo cranial bone defect model in rats. Micro-CT images showed that the hybrid scaffolds with 20% DCB content had the best effect in promoting new bone regeneration. Thus, it was concluded that the DCB-GelMA hybrid scaffolds have high potential in bone tissue engineering applications.
Collapse
Affiliation(s)
- Chenyuan Gao
- Department of Orthopaedics, Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, P. R. China. and Engineering Research Center of Clinical Functional Materials and Diagnosis & Treatment Devices of Zhejiang Province, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325011, P. R. China
| | - Wan Ting Sow
- Engineering Research Center of Clinical Functional Materials and Diagnosis & Treatment Devices of Zhejiang Province, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325011, P. R. China
| | - Yingying Wang
- School of Biomedical Engineering, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, 325035, P. R. China
| | - Yili Wang
- Engineering Research Center of Clinical Functional Materials and Diagnosis & Treatment Devices of Zhejiang Province, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325011, P. R. China
| | - Dejun Yang
- Engineering Research Center of Clinical Functional Materials and Diagnosis & Treatment Devices of Zhejiang Province, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325011, P. R. China and School of Biomedical Engineering, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, 325035, P. R. China
| | - Bae Hoon Lee
- Engineering Research Center of Clinical Functional Materials and Diagnosis & Treatment Devices of Zhejiang Province, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325011, P. R. China and School of Biomedical Engineering, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, 325035, P. R. China
| | - DraŽen Matičić
- Faculty of Veterinary Medicine, University of Zagreb, Zagreb, 10000, Croatia
| | - Lian Fang
- ENT Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, P. R. China
| | - Huaqiong Li
- Department of Orthopaedics, Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, P. R. China. and Engineering Research Center of Clinical Functional Materials and Diagnosis & Treatment Devices of Zhejiang Province, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325011, P. R. China
| | - Chunwu Zhang
- Department of Orthopaedics, Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, P. R. China.
| |
Collapse
|
42
|
Barra JM, Kozlovskaya V, Kepple JD, Seeberger KL, Kuppan P, Hunter CS, Korbutt GS, Kharlampieva E, Tse HM. Xenotransplantation of tannic acid-encapsulated neonatal porcine islets decreases proinflammatory innate immune responses. Xenotransplantation 2021; 28:e12706. [PMID: 34245064 DOI: 10.1111/xen.12706] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 06/10/2021] [Accepted: 06/27/2021] [Indexed: 01/04/2023]
Abstract
BACKGROUND Islet transplantation with neonatal porcine islets (NPIs) is a promising treatment for type 1 diabetes (T1D), but immune rejection poses a major hurdle for clinical use. Innate immune-derived reactive oxygen species (ROS) synthesis can facilitate islet xenograft destruction and enhance adaptive immune responses. METHODS To suppress ROS-mediated xenograft destruction, we utilized nanothin encapsulation materials composed of multilayers of tannic acid (TA), an antioxidant, and a neutral polymer, poly(N-vinylpyrrolidone) (PVPON). We hypothesized that (PVPON/TA)-encapsulated NPIs will maintain euglycemia and dampen proinflammatory innate immune responses following xenotransplantation. RESULTS (PVPON/TA)-encapsulated NPIs were viable and glucose-responsive similar to non-encapsulated NPIs. Transplantation of (PVPON/TA)-encapsulated NPIs into hyperglycemic C57BL/6.Rag or NOD.Rag mice restored euglycemia, exhibited glucose tolerance, and maintained islet-specific transcription factor levels similar to non-encapsulated NPIs. Gene expression analysis of (PVPON/TA)-encapsulated grafts post-transplantation displayed reduced proinflammatory Ccl5, Cxcl10, Tnf, and Stat1 while enhancing alternatively activated macrophage Retnla, Arg1, and Stat6 mRNA accumulation compared with controls. Flow cytometry analysis demonstrated significantly reduced innate immune infiltration, MHC-II, co-stimulatory molecule, and TNF expression with concomitant increases in arginase-1+ macrophages and dendritic cells. Similar alterations in immune responses were observed following xenotransplantation into immunocompetent NOD mice. CONCLUSION Our data suggest that (PVPON/TA) encapsulation of NPIs is an effective strategy to decrease inflammatory innate immune signals involved in NPI xenograft responses through STAT1/6 modulation without compromising islet function.
Collapse
Affiliation(s)
- Jessie M Barra
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA.,Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Veronika Kozlovskaya
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL, USA.,Center for Nanoscale Materials and Biointegration, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jessica D Kepple
- Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL, USA.,Department of Medicine, Division of Endocrinology Diabetes and Metabolism, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Karen L Seeberger
- Department of Surgery, Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
| | - Purushothaman Kuppan
- Department of Surgery, Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
| | - Chad S Hunter
- Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL, USA.,Department of Medicine, Division of Endocrinology Diabetes and Metabolism, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Gregory S Korbutt
- Department of Surgery, Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
| | - Eugenia Kharlampieva
- Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL, USA.,Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL, USA.,Center for Nanoscale Materials and Biointegration, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hubert M Tse
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA.,Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL, USA.,Center for Nanoscale Materials and Biointegration, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
43
|
Cohen S, Partouche S, Gurevich M, Tennak V, Mezhybovsky V, Azarov D, Soffer-Hirschberg S, Hovav B, Niv-Drori H, Weiss C, Borovich A, Cohen G, Wertheimer A, Shukrun G, Israeli M, Yahalom V, Leshem-Lev D, Perl L, Kornowski R, Wiznitzer A, Tobar A, Feinmesser M, Mor E, Atar E, Nesher E. Generation of vascular chimerism within donor organs. Sci Rep 2021; 11:13437. [PMID: 34183759 PMCID: PMC8238957 DOI: 10.1038/s41598-021-92823-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 06/16/2021] [Indexed: 01/22/2023] Open
Abstract
Whole organ perfusion decellularization has been proposed as a promising method to generate non-immunogenic organs from allogeneic and xenogeneic donors. However, the ability to recellularize organ scaffolds with multiple patient-specific cells in a spatially controlled manner remains challenging. Here, we propose that replacing donor endothelial cells alone, while keeping the rest of the organ viable and functional, is more technically feasible, and may offer a significant shortcut in the efforts to engineer transplantable organs. Vascular decellularization was achieved ex vivo, under controlled machine perfusion conditions, in various rat and porcine organs, including the kidneys, liver, lungs, heart, aorta, hind limbs, and pancreas. In addition, vascular decellularization of selected organs was performed in situ, within the donor body, achieving better control over the perfusion process. Human placenta-derived endothelial progenitor cells (EPCs) were used as immunologically-acceptable human cells to repopulate the luminal surface of de-endothelialized aorta (in vitro), kidneys, lungs and hind limbs (ex vivo). This study provides evidence that artificially generating vascular chimerism is feasible and could potentially pave the way for crossing the immunological barrier to xenotransplantation, as well as reducing the immunological burden of allogeneic grafts.
Collapse
Affiliation(s)
- Shahar Cohen
- Laboratory for Organ Bioengineering, Rabin Medical Center, Petah Tikva, Israel.
| | - Shirly Partouche
- Laboratory for Organ Bioengineering, Rabin Medical Center, Petah Tikva, Israel.,Felsenstien Medical Research Center, Rabin Medical Center, Petah Tikva, Israel
| | - Michael Gurevich
- Department of Organ Transplantation, Rabin Medical Center, Petah Tikva, Israel
| | - Vladimir Tennak
- Department of Organ Transplantation, Rabin Medical Center, Petah Tikva, Israel
| | - Vadym Mezhybovsky
- Department of Organ Transplantation, Rabin Medical Center, Petah Tikva, Israel
| | - Dmitry Azarov
- Experimental Surgery Unit, Rabin Medical Center, Petah Tikva, Israel
| | | | - Benny Hovav
- Department of Radiology, Rabin Medical Center, Petah Tikva, Israel
| | - Hagit Niv-Drori
- Department of Pathology, Rabin Medical Center, Petah Tikva, Israel
| | - Chana Weiss
- Department of Pathology, Rabin Medical Center, Petah Tikva, Israel
| | - Adi Borovich
- Helen Schneider Hospital for Women, Rabin Medical Center, Petah Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Guy Cohen
- Helen Schneider Hospital for Women, Rabin Medical Center, Petah Tikva, Israel
| | - Avital Wertheimer
- Helen Schneider Hospital for Women, Rabin Medical Center, Petah Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Golan Shukrun
- Division of Pediatric Cardiothoracic Surgery, Schneider Children's Medical Center, Petah Tikva, Israel.,Department of Cardiothoracic Surgery, Rabin Medical Center, Petah Tikva, Israel
| | - Moshe Israeli
- Tissue Typing Laboratory, Rabin Medical Center, Petah Tikva, Israel.,Zefat Academic College, Zefat, Israel
| | - Vered Yahalom
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Blood Services and Apheresis Institute, Rabin Medical Center, Petah Tikva, Israel
| | - Dorit Leshem-Lev
- Felsenstien Medical Research Center, Rabin Medical Center, Petah Tikva, Israel.,Department of Cardiology, Rabin Medical Center, Petah Tikva, Israel
| | - Leor Perl
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Cardiology, Rabin Medical Center, Petah Tikva, Israel
| | - Ran Kornowski
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Cardiology, Rabin Medical Center, Petah Tikva, Israel
| | - Arnon Wiznitzer
- Helen Schneider Hospital for Women, Rabin Medical Center, Petah Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ana Tobar
- Department of Pathology, Rabin Medical Center, Petah Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Meora Feinmesser
- Department of Pathology, Rabin Medical Center, Petah Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Eytan Mor
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Transplantation Unit, Department of Surgery B, Sheba Medical Center, Ramat Gan, Israel
| | - Eli Atar
- Department of Radiology, Rabin Medical Center, Petah Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Eviatar Nesher
- Department of Organ Transplantation, Rabin Medical Center, Petah Tikva, Israel
| |
Collapse
|
44
|
Hara H, Iwase H, Nguyen H, Miyagawa Y, Kuravi K, Foote JB, Eyestone W, Phelps C, Ayares D, Cooper DKC. Stable expression of the human thrombomodulin transgene in pig endothelial cells is associated with a reduction in the inflammatory response. Cytokine 2021; 148:155580. [PMID: 34099346 DOI: 10.1016/j.cyto.2021.155580] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 05/10/2021] [Accepted: 05/11/2021] [Indexed: 01/31/2023]
Abstract
BACKGROUND Xenotransplantation is associated with an inflammatory response. The proinflammatory cytokine, TNF-α, downregulates the expression of thrombomodulin (TBM), and induces coagulation dysfunction. Although human (h) TBM-transgenic pigs (p) have been developed to reduce coagulation dysfunction, the effect of TNF-α on the expression of hTBM and its functional activity has not been fully investigated. The aims of this study were to investigate (i) whether the expression of hTBM on pig (p) cells is down-regulated during TNF-α stimulation, and (ii) whether cells from hTBM pigs regulate the inflammatory response. METHODS TNF-α-producing T, B, and natural killer cells in blood from baboons with pig heart or kidney xenografts were investigated by flow cytometry. TNF-α staining in the grafts was detected by immunohistochemistry. Aortic endothelial cells (AECs) from GTKO/CD46 and GTKO/CD46/hTBM pigs were stimulated by hTNF-α, and the expression of the inflammatory/coagulation regulatory protein, TBM, was investigated. RESULTS After pig organ xenotransplantation, there was a trend to increases in TNF-α-producing T and natural killer cells in the blood of baboons. In vitro observations demonstrated that after hTNF-α stimulation, there was a significant reduction in the expression of endogenous pTBM on pAECs, and a significant increase in the expression of inflammatory molecules. Blocking of NF-κB signaling significantly up-regulated pTBM expression, and suppressed the inflammatory response induced by hTNF-α in pAECs. Whereas the expression of pTBM mRNA was significantly reduced by hTNF-α stimulation, hTBM expression on the GTKO/CD46/hTBM pAECs was not affected. Furthermore, after hTNF-α stimulation, there was significant suppression of expression of inflammatory molecules on GTKO/CD46/hTBM pAECs compared to GTKO/CD46 pAECs. CONCLUSIONS The stable expression of hTBM in pig cells may locally regulate the inflammatory response. This will help suppress the inflammatory response and prevent coagulation dysregulation after xenotransplantation.
Collapse
Affiliation(s)
- Hidetaka Hara
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA; Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Hayato Iwase
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA; Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Huy Nguyen
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Yuko Miyagawa
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Jeremy B Foote
- Department of Microbiology and Animal Resources Program, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | | | - David K C Cooper
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
45
|
Lucander ACK, Nguyen H, Foote JB, Cooper DKC, Hara H. Immunological selection and monitoring of patients undergoing pig kidney transplantation. Xenotransplantation 2021; 28:e12686. [PMID: 33880816 DOI: 10.1111/xen.12686] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/03/2021] [Accepted: 03/18/2021] [Indexed: 01/04/2023]
Abstract
Pig kidney xenotransplantation has the potential to alleviate the current shortage of deceased and living human organs and provide patients with end-stage renal disease with a greater opportunity for long-term survival and a better quality of life. In recent decades, advances in the genetic engineering of pigs and in immunosuppressive therapy have permitted the resolution of many historical obstacles to the success of pig kidney transplantation in nonhuman primates. Pig kidney xenotransplantation may soon be translated to the clinic. Given the potential risks of kidney xenotransplantation, particularly of immunologic rejection of the graft, potential patients must be carefully screened for inclusion in the initial clinical trials and immunologically monitored diligently post-transplantation. We provide an overview of the immunological methods we believe should be used to (i) screen potential patients for the first clinical trials to exclude those with a higher risk of rejection, and (ii) monitor patients with a pig kidney graft to determine their immunological response to the graft.
Collapse
Affiliation(s)
- Aaron C K Lucander
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Huy Nguyen
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jeremy B Foote
- Department of Microbiology and Animal Resources Program, University of Alabama at Birmingham, Birmingham, AL, USA
| | - David K C Cooper
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hidetaka Hara
- Xenotransplantation Program, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
46
|
Thompson CP, Jagdale A, Walcott G, Iwase H, Foote JB, Cron RQ, Hara H, Cleveland DC, Cooper DKC. A perspective on the potential detrimental role of inflammation in pig orthotopic heart xenotransplantation. Xenotransplantation 2021; 28:e12687. [PMID: 33786912 DOI: 10.1111/xen.12687] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 02/26/2021] [Accepted: 03/18/2021] [Indexed: 02/06/2023]
Abstract
There is a critical shortage of deceased human donor organs for transplantation. The need is perhaps most acute in neonates and infants with life-threatening congenital heart disease, in whom mechanical support devices are largely unsuccessful. If orthotopic (life-supporting) heart transplantation (OHTx) were consistently successful in the genetically engineered pig-to-nonhuman primate (NHP) model, a clinical trial of bridging with a pig heart in such patients might be justified. However, the results of pig OHTx in NHPs have been mixed and largely poor. We hypothesise that a factor is the detrimental effects of the inflammatory response that is known to develop (a) during any surgical procedure that requires cardiopulmonary bypass, and (b) immediately after an NHP recipient is exposed to a pig xenograft. We suggest that the combination of these two inflammatory responses has a direct detrimental effect on pig heart graft function, but also, and possibly of more importance, on recipient baboon pulmonary function, which further impacts survival of the pig heart graft. In addition, the inflammatory response almost certainly adversely impacts the immune response to the graft. If our hypothesis is correct, the potential steps that could be taken to reduce the inflammatory response or its effects (with varying degrees of efficacy) include (a) white blood cell filtration, (b) complement depletion or inactivation, (c) immunosuppressive therapy, (d) high-dose corticosteroid therapy, (e) cytokine/chemokine-targeted therapy, (f) ultrafiltration or CytoSorb hemoperfusion, (g) reduction in the levels of endogenous catecholamines, (h) triiodothyronine therapy and (i) genetic engineering of the organ-source pig. Prevention of the inflammatory response, or attenuation of its effects, by judicious anti-inflammatory therapy may contribute not only to early survival of the recipient of a genetically engineered pig OHTx, but also to improved long-term pig heart graft survival. This would open the possibility of initiating a clinical trial of genetically engineered pig OHTx as a bridge to allotransplantation.
Collapse
Affiliation(s)
- Charles P Thompson
- Xenotransplantation Program, Division of Transplantation, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Abhijit Jagdale
- Xenotransplantation Program, Division of Transplantation, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Gregory Walcott
- Department of Medicine/Cardiovascular Diseases, the University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hayato Iwase
- Xenotransplantation Program, Division of Transplantation, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jeremy B Foote
- Department of Microbiology and Animal Resources Program, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Randall Q Cron
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hidetaka Hara
- Xenotransplantation Program, Division of Transplantation, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - David C Cleveland
- Division of Cardiothoracic Surgery, Children's Hospital of Alabama, and Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - David K C Cooper
- Xenotransplantation Program, Division of Transplantation, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
47
|
Hurst DJ, Padilla LA, Cooper DKC, Paris W. Factors influencing attitudes toward xenotransplantation clinical trials: A report of focus group studies. Xenotransplantation 2021; 28:e12684. [PMID: 33682188 DOI: 10.1111/xen.12684] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 02/16/2021] [Accepted: 02/25/2021] [Indexed: 12/29/2022]
Abstract
BACKGROUND Clinical trials of xenotransplantation (XTx, ie, cross-species transplantation, specifically the transplantation of genetically engineered pig organs into human recipients) are due to start within a few years. METHODS Five focus groups were conducted in April-May 2019, among local religious leaders (N = 10), organ procurement staff/administrators (N = 5), patients and parents of patients who may need an organ transplant or are transplant recipients (N = 9), and local businesspersons in the community (N = 3). Groups were audio-recorded, transcribed, and analyzed with NVivo software to identify themes of participants' thoughts to XTx. RESULTS An overall Cohen's kappa statistic of 0.71 was established. In general, there was wide agreement among participants that XTx is an exciting and acceptable option to explore as an organ alternative. Concerns were expressed primarily regarding issues of animal ethics, stigma regarding how pigs are viewed in society, organ allocation logistics, quality of life after receiving a xenograft, and how XTx would be accepted by certain theological traditions. CONCLUSION Overall, various stakeholders accepted XTx as a clinical option. However, there were ethical, social, and physical concerns raised. Future studies exploring the development of education strategies that may help alleviate concerns related to XTx before it becomes a clinical alternative are needed among the general public, potential XTx candidates, and their family members.
Collapse
Affiliation(s)
- Daniel J Hurst
- Medical Professionalism, Department of Family Medicine, Rowan University School of Osteopathic Medicine, Stratford, NJ, USA
| | - Luz A Padilla
- Department of Epidemiology and Surgery, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - David K C Cooper
- UAB Xenotransplantation Program, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Wayne Paris
- Department of Social Work, Abilene Christian University, Abilene, TX, USA
| |
Collapse
|
48
|
Song M, Fitch ZW, Samy KP, Martin BM, Gao Q, Patrick Davis R, Leopardi FV, Huffman N, Schmitz R, Devi GR, Collins BH, Kirk AD. Coagulation, inflammation, and CD46 transgene expression in neonatal porcine islet xenotransplantation. Xenotransplantation 2021; 28:e12680. [PMID: 33619844 DOI: 10.1111/xen.12680] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 12/17/2020] [Accepted: 02/12/2021] [Indexed: 12/20/2022]
Abstract
BACKGROUND Thrombosis is a known consequence of intraportal islet transplantation, particularly for xenogeneic islets. To define the origins of thrombosis after islet xenotransplantation and relate it to early inflammation, we examined porcine islets transplanted into non-human primates using a dual-transplant model to directly compare islet characteristics. METHODS α1,3-Galactosyltransferase gene-knockout (GTKO) islets with and without expression of the human complement regulatory transgene CD46 (hCD46) were studied. Biologically inert polyethylene microspheres were used to examine the generic pro-thrombotic effects of particle embolization. Immunohistochemistry was performed 1 and 24 hours after transplantation. RESULTS Xeno-islet transplantation activated both extrinsic and intrinsic coagulation pathways. The intrinsic pathway was also initiated by microsphere embolization, while extrinsic pathway tissue factor (TF) and platelet aggregation were more specific to engrafted islets. hCD46 expression significantly reduced TF, platelet, fibrin, and factor XIIIa accumulation in and around islets but did not alter intrinsic factor activation. Layers of TF+ cells emerged around islets within 24 hours, particularly co-localized with vimentin, and identified as CD3+ and CD68+ cells inflammatory cells. CONCLUSIONS These findings detail the origins of thrombosis following islet xenotransplantation, relate it to early immune activation, and suggest a role for transgenic hCD46 expression in its mitigation. Layers of TF-positive inflammatory cells and fibroblasts around islets at 24 hours may have important roles in the progressive events of thrombosis, inflammatory cell recruitment, rejection, and the ultimate outcome of transplanted grafts. These suggest that the strategies targeting these elements could yield more progress toward successful xenogeneic islet engraftment and survival.
Collapse
Affiliation(s)
- Mingqing Song
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Zachary W Fitch
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Kannan P Samy
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Benjamin M Martin
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Qimeng Gao
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | | | - Francis V Leopardi
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Niki Huffman
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Robin Schmitz
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Gayathri R Devi
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Bradley H Collins
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Allan D Kirk
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
49
|
Anti-pig IgE and IgA Antibodies in Naive Primates and Nonhuman Primates With Pig Xenografts. Transplantation 2021; 105:318-327. [PMID: 32796494 DOI: 10.1097/tp.0000000000003408] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
BACKGROUND Natural preformed anti-pig IgM/IgG antibodies in primates play an important role in xenograft rejection. As it is not clear how IgE and IgA engage in the immune system in xenotransplantation, we investigated natural preformed and elicited anti-pig IgE/IgA in naive primates and after xenotransplantation in nonhuman primates. METHODS The binding of IgM/IgG/IgE/IgA antibodies to red blood cells (RBCs) from wild-type (WT), α1,3-galactosyltransferase gene-knockout (GTKO), and GTKO/cytidine monophospho-N-acetylneuraminic acid hydroxylase gene-knockout/β-1,4 N-acetylgalactosaminyltransferase 2 gene-knockout (ie, triple-knockout pigs) pigs were measured by flow cytometry in naive human (n = 50) and baboon (n = 14) sera. Antibody binding to WT and GTKO pig RBCs (pRBCs) was also measured in the sera of baboons (nonsensitized n = 7, sensitized n = 2) and rhesus monkeys (nonsensitized n = 2, sensitized n = 11) following WT or GTKO pig organ/tissue xenotransplantation. Deposition of IgM/IgG/IgE/IgA in the grafts was detected by immunohistochemistry. RESULTS The majority of humans had natural preformed IgM/IgG/IgE/IgA to WT and GTKO pRBCs. In contrast, IgM/IgG/IgE/IgA to triple-knockout pRBCs were present at lower levels and frequency (P < 0.01). Baboons also had IgM/IgG/IgE/IgA antibodies against WT pRBCs, but fewer to GTKO and triple-knockout (P < 0.01). After xenotransplantation into nonhuman primates, when IgM/IgG increased, IgE/IgA also increased, but to a lesser extent. In addition to IgM/IgG, IgE or IgA deposition was observed in rejected pig xenografts. CONCLUSIONS Primates develop serum anti-pig IgE/IgA antibodies both naturally and during xenograft rejection. The pathophysiological role, if any, of anti-pig IgE/IgA antibodies remains unknown.
Collapse
|
50
|
Lewis PL, Wells JM. Engineering-inspired approaches to study β-cell function and diabetes. Stem Cells 2021; 39:522-535. [PMID: 33497522 DOI: 10.1002/stem.3340] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 01/13/2021] [Indexed: 12/21/2022]
Abstract
Strategies to mitigate the pathologies from diabetes range from simply administering insulin to prescribing complex drug/biologic regimens combined with lifestyle changes. There is a substantial effort to better understand β-cell physiology during diabetes pathogenesis as a means to develop improved therapies. The convergence of multiple fields ranging from developmental biology to microfluidic engineering has led to the development of new experimental systems to better study complex aspects of diabetes and β-cell biology. Here we discuss the available insulin-secreting cell types used in research, ranging from primary human β-cells, to cell lines, to pluripotent stem cell-derived β-like cells. Each of these sources possess inherent strengths and weaknesses pertinent to specific applications, especially in the context of engineered platforms. We then outline how insulin-expressing cells have been used in engineered platforms and how recent advances allow for better mimicry of in vivo conditions. Chief among these conditions are β-cell interactions with other endocrine organs. This facet is beginning to be thoroughly addressed by the organ-on-a-chip community, but holds enormous potential in the development of novel diabetes therapeutics. Furthermore, high throughput strategies focused on studying β-cell biology, improving β-cell differentiation, or proliferation have led to enormous contributions in the field and will no doubt be instrumental in bringing new diabetes therapeutics to the clinic.
Collapse
Affiliation(s)
- Phillip L Lewis
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - James M Wells
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|