1
|
Abdalla MMI. Advancing diabetes management: Exploring pancreatic beta-cell restoration's potential and challenges. World J Gastroenterol 2024; 30:4339-4353. [PMID: 39494103 PMCID: PMC11525866 DOI: 10.3748/wjg.v30.i40.4339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 09/05/2024] [Accepted: 09/24/2024] [Indexed: 10/16/2024] Open
Abstract
Diabetes mellitus, characterized by chronic hyperglycemia due to insulin deficiency or resistance, poses a significant global health burden. Central to its pathogenesis is the dysfunction or loss of pancreatic beta cells, which are res-ponsible for insulin production. Recent advances in beta-cell regeneration research offer promising strategies for diabetes treatment, aiming to restore endogenous insulin production and achieve glycemic control. This review explores the physiological basis of beta-cell function, recent scientific advan-cements, and the challenges in translating these findings into clinical applications. It highlights key developments in stem cell therapy, gene editing technologies, and the identification of novel regenerative molecules. Despite the potential, the field faces hurdles such as ensuring the safety and long-term efficacy of regen-erative therapies, ethical concerns around stem cell use, and the complexity of beta-cell differentiation and integration. The review highlights the importance of interdisciplinary collaboration, increased funding, the need for patient-centered approaches and the integration of new treatments into comprehensive care strategies to overcome these challenges. Through continued research and collaboration, beta-cell regeneration holds the potential to revolutionize diabetes care, turning a chronic condition into a manageable or even curable disease.
Collapse
Affiliation(s)
- Mona Mohamed Ibrahim Abdalla
- Department of Human Biology, School of Medicine, International Medical University, Bukit Jalil 57000, Kuala Lumpur, Malaysia
| |
Collapse
|
2
|
Sun A, Hayat H, Kenyon E, Quadri T, Amos D, Perkins K, Nigam S, Tarleton D, Mallett CL, Deng CX, Qiu Z, Li W, Sempere L, Fan J, Aguirre A, Wang P. Brown Adipose Tissue as a Unique Niche for Islet Organoid Transplantation: Insights From In Vivo Imaging. Transplant Direct 2024; 10:e1658. [PMID: 38881741 PMCID: PMC11177823 DOI: 10.1097/txd.0000000000001658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 03/15/2024] [Accepted: 04/03/2024] [Indexed: 06/18/2024] Open
Abstract
Background Transplantation of human-induced pluripotent stem cell (hiPSC)-derived islet organoids is a promising cell replacement therapy for type 1 diabetes (T1D). It is important to improve the efficacy of islet organoids transplantation by identifying new transplantation sites with high vascularization and sufficient accommodation to support graft survival with a high capacity for oxygen delivery. Methods A human-induced pluripotent stem cell line (hiPSCs-L1) was generated constitutively expressing luciferase. Luciferase-expressing hiPSCs were differentiated into islet organoids. The islet organoids were transplanted into the scapular brown adipose tissue (BAT) of nonobese diabetic/severe combined immunodeficiency disease (NOD/SCID) mice as the BAT group and under the left kidney capsule (KC) of NOD/SCID mice as a control group, respectively. Bioluminescence imaging (BLI) of the organoid grafts was performed on days 1, 7, 14, 28, 35, 42, 49, 56, and 63 posttransplantation. Results BLI signals were detected in all recipients, including both the BAT and control groups. The BLI signal gradually decreased in both BAT and KC groups. However, the graft BLI signal intensity under the left KC decreased substantially faster than that of the BAT. Furthermore, our data show that islet organoids transplanted into streptozotocin-induced diabetic mice restored normoglycemia. Positron emission tomography/MRI verified that the islet organoids were transplanted at the intended location in these diabetic mice. Immunofluorescence staining revealed the presence of functional organoid grafts, as confirmed by insulin and glucagon staining. Conclusions Our results demonstrate that BAT is a potentially desirable site for islet organoid transplantation for T1D therapy.
Collapse
Affiliation(s)
- Aixia Sun
- Precision Health Program, Michigan State University, East Lansing, MI
- Department of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI
| | - Hanaan Hayat
- Precision Health Program, Michigan State University, East Lansing, MI
- Department of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI
| | - Elizabeth Kenyon
- Precision Health Program, Michigan State University, East Lansing, MI
- Department of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI
| | - Tahnia Quadri
- Precision Health Program, Michigan State University, East Lansing, MI
- Department of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI
| | - Darius Amos
- Precision Health Program, Michigan State University, East Lansing, MI
- Department of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI
- College of Osteopathic Medicine, Michigan State University, East Lansing, MI
| | - Keenan Perkins
- Florida Agricultural and Mechanical University, Tallahassee, FL
| | - Saumya Nigam
- Precision Health Program, Michigan State University, East Lansing, MI
- Department of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI
| | - Deanna Tarleton
- Precision Health Program, Michigan State University, East Lansing, MI
- Department of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI
| | - Christiane L Mallett
- Department of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, MI
| | - Cheri X Deng
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI
| | - Zhen Qiu
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, MI
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI
| | - Wen Li
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, MI
- Department of Electrical and Computer Engineering, College of Engineering, Michigan State University, East Lansing, MI
| | - Lorenzo Sempere
- Precision Health Program, Michigan State University, East Lansing, MI
- Department of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI
| | - Jinda Fan
- Department of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, MI
- Department of Chemistry, College of Natural Science, Michigan State University, East Lansing, MI
| | - Aitor Aguirre
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, MI
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI
| | - Ping Wang
- Precision Health Program, Michigan State University, East Lansing, MI
- Department of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI
| |
Collapse
|
3
|
Yang SN, Shi Y, Berggren PO. The anterior chamber of the eye technology and its anatomical, optical, and immunological bases. Physiol Rev 2024; 104:881-929. [PMID: 38206586 PMCID: PMC11381035 DOI: 10.1152/physrev.00024.2023] [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: 06/20/2023] [Revised: 11/30/2023] [Accepted: 01/05/2024] [Indexed: 01/12/2024] Open
Abstract
The anterior chamber of the eye (ACE) is distinct in its anatomy, optics, and immunology. This guarantees that the eye perceives visual information in the context of physiology even when encountering adverse incidents like inflammation. In addition, this endows the ACE with the special nursery bed iris enriched in vasculatures and nerves. The ACE constitutes a confined space enclosing an oxygen/nutrient-rich, immune-privileged, and less stressful milieu as well as an optically transparent medium. Therefore, aside from visual perception, the ACE unexpectedly serves as an excellent transplantation site for different body parts and a unique platform for noninvasive, longitudinal, and intravital microimaging of different grafts. On the basis of these merits, the ACE technology has evolved from the prototypical through the conventional to the advanced version. Studies using this technology as a versatile biomedical research platform have led to a diverse range of basic knowledge and in-depth understanding of a variety of cells, tissues, and organs as well as artificial biomaterials, pharmaceuticals, and abiotic substances. Remarkably, the technology turns in vivo dynamic imaging of the morphological characteristics, organotypic features, developmental fates, and specific functions of intracameral grafts into reality under physiological and pathological conditions. Here we review the anatomical, optical, and immunological bases as well as technical details of the ACE technology. Moreover, we discuss major achievements obtained and potential prospective avenues for this technology.
Collapse
Affiliation(s)
- Shao-Nian Yang
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden
| | - Yue Shi
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden
| | - Per-Olof Berggren
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
4
|
Wang X, Deng H, Lin J, Zhang K, Ni J, Li L, Fan G. Distinct roles of telomerase activity in age-related chronic diseases: An update literature review. Biomed Pharmacother 2023; 167:115553. [PMID: 37738798 DOI: 10.1016/j.biopha.2023.115553] [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: 07/22/2023] [Revised: 09/15/2023] [Accepted: 09/18/2023] [Indexed: 09/24/2023] Open
Abstract
Although telomerase has low activity in somatic quiescent cells, it plays an significant roles in regenerative cells such as endothelial cells, hepatocytes, epithelial cells, and hemocytes. Telomerase activity and telomere length are critical factors in age-related chronic diseases as they are closely related to cell senescence. However, whether telomerase activity plays a key role in disease progression or whether the role of telomerase is unified among different diseases are unresolved. Considering that aging is the most important risk factor for neurodegenerative and metabolic diseases, this article will analyze the evidence, mechanism, and therapeutic potential of telomerase activity in several chronic disease, including type 2 diabetes, neurodegenerative diseases, atherosclerosis, heart failure and non-alcoholic fatty liver disease, in order to provide clues for the use of telomerase activity to target the treatment of age-related chronic diseases.
Collapse
Affiliation(s)
- Xiaodan Wang
- Medical Experiment Center, Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, 300381 Tianjin, China
| | - Hao Deng
- Medical Experiment Center, Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, 300381 Tianjin, China
| | - Jingyi Lin
- Medical Experiment Center, Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, 300381 Tianjin, China
| | - Kai Zhang
- Medical Experiment Center, Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, 300381 Tianjin, China
| | - Jingyu Ni
- Medical Experiment Center, Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, 300381 Tianjin, China
| | - Lan Li
- State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae for the Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Guanwei Fan
- Medical Experiment Center, Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, 300381 Tianjin, China.
| |
Collapse
|
5
|
Frech TM, Maguire C, Petrey AC, Stoddard GJ, Donato AJ. A systemic sclerosis disease model: can inducible pluripotent stem cells fill an unmet need in defining vascular leak? Rheumatology (Oxford) 2023; 62:e226-e228. [PMID: 36575990 PMCID: PMC10321079 DOI: 10.1093/rheumatology/keac719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 12/13/2022] [Indexed: 12/29/2022] Open
Affiliation(s)
- Tracy M Frech
- Department of Internal Medicine, Division of Rheumatology and Immunology, Tennessee Valley Health Care System, Veterans Affairs Medical Center and Vanderbilt University Medical Center, Nashville, TN, USA
| | - Colin Maguire
- Clinical and Translational Science Institute, University of Utah, Salt Lake City, UT, USA
| | - Aaron C Petrey
- Molecular Medicine Program, University of Utah, Salt Lake City, UT, USA
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Gregory J Stoddard
- Study Design and Biostatistics Center, University of Utah, Salt Lake City, UT, USA
| | - Anthony J Donato
- Department of Internal Medicine, George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, UT, USA
| |
Collapse
|
6
|
In Vivo CaV3 Channel Inhibition Promotes Maturation of Glucose-Dependent Ca2+ Signaling in Human iPSC-Islets. Biomedicines 2023; 11:biomedicines11030807. [PMID: 36979793 PMCID: PMC10045717 DOI: 10.3390/biomedicines11030807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 02/26/2023] [Accepted: 03/02/2023] [Indexed: 03/10/2023] Open
Abstract
CaV3 channels are ontogenetically downregulated with the maturation of certain electrically excitable cells, including pancreatic β cells. Abnormally exaggerated CaV3 channels drive the dedifferentiation of mature β cells. This led us to question whether excessive CaV3 channels, retained mistakenly in engineered human-induced pluripotent stem cell-derived islet (hiPSC-islet) cells, act as an obstacle to hiPSC-islet maturation. We addressed this question by using the anterior chamber of the eye (ACE) of immunodeficient mice as a site for recapitulation of in vivo hiPSC-islet maturation in combination with intravitreal drug infusion, intravital microimaging, measurements of cytoplasmic-free Ca2+ concentration ([Ca2+]i) and patch clamp analysis. We observed that the ACE is well suited for recapitulation, observation and intervention of hiPSC-islet maturation. Intriguingly, intraocular hiPSC-islet grafts, retrieved intact following intravitreal infusion of the CaV3 channel blocker NNC55-0396, exhibited decreased basal [Ca2+]i levels and increased glucose-stimulated [Ca2+]i responses. Insulin-expressing cells of these islet grafts indeed expressed the NNC55-0396 target CaV3 channels. Intraocular hiPSC-islets underwent satisfactory engraftment, vascularization and light scattering without being influenced by the intravitreally infused NNC55-0396. These data demonstrate that inhibiting CaV3 channels facilitates the maturation of glucose-activated Ca2+ signaling in hiPSC-islets, supporting the notion that excessive CaV3 channels as a developmental error impede the maturation of engineer ed hiPSC-islet insulin-expressing cells.
Collapse
|
7
|
Shah P. Genomic Editing and Diabetes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1396:207-214. [DOI: 10.1007/978-981-19-5642-3_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
|
8
|
Padovano M, Scopetti M, Manetti F, Morena D, Radaelli D, D’Errico S, Di Fazio N, Frati P, Fineschi V. Pancreatic transplant surgery and stem cell therapy: Finding the balance between therapeutic advances and ethical principles. World J Stem Cells 2022; 14:577-586. [PMID: 36157914 PMCID: PMC9453273 DOI: 10.4252/wjsc.v14.i8.577] [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: 03/18/2022] [Revised: 05/24/2022] [Accepted: 07/31/2022] [Indexed: 02/07/2023] Open
Abstract
The latest achievements in the field of pancreas transplantation and stem cell therapy require an effort by the scientific community to clarify the ethical implications of pioneering treatments, often characterized by high complexity from a surgical point of view, due to transplantation of multiple organs at the same time or at different times, and from an immunological point of view for stem cell therapy. The fundamental value in the field of organ transplants is, of course, a solidarity principle, namely that of protecting the health and life of people for whom transplantation is a condition of functional recovery, or even of survival. The nature of this value is that of a concept to which the legal discipline of transplants entrusts its own ethical dignity and for which it has ensured a constitutional recognition in different systems. The general principle of respect for human life, both of the donor and of the recipient, evokes the need not to put oneself and one’s neighbor in dangerous conditions. The present ethical reflection aims to find a balance between the latest therapeutic advances and several concepts including the idea of the person, the respect due to the dead, the voluntary nature of the donation and the consent to the same, the gratuitousness of the donation, the scientific progress and the development of surgical techniques, and the policies of health promotion.
Collapse
Affiliation(s)
- Martina Padovano
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, Rome 00185, Italy
| | - Matteo Scopetti
- Department of Medical Surgical Sciences and Translational Medicine, Sapienza University of Rome, Rome 00189, Italy
| | - Federico Manetti
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, Rome 00185, Italy
| | - Donato Morena
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, Rome 00185, Italy
| | - Davide Radaelli
- Department of Medicine, Surgery and Health, University of Trieste, Trieste 34149, Italy
| | - Stefano D’Errico
- Department of Medicine, Surgery and Health, University of Trieste, Trieste 34149, Italy
| | - Nicola Di Fazio
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, Rome 00185, Italy
| | - Paola Frati
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, Rome 00185, Italy
| | - Vittorio Fineschi
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, Rome 00185, Italy
| |
Collapse
|
9
|
Zhao K, Shi Y, Yu J, Yu L, Mael A, Li Y, Kolton A, Joyce T, Odorico J, Berggren PO, Yang SN. Intracameral Microimaging of Maturation of Human iPSC Derivatives into Islet Endocrine Cells. Cell Transplant 2022; 31:9636897211066508. [PMID: 35156411 PMCID: PMC8848082 DOI: 10.1177/09636897211066508] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
We exploited the anterior chamber of the eye (ACE) of immunodeficient mice as an ectopic site for both transplantation and microimaging of engineered surrogate islets from human induced pluripotent stem cells (hiPSC-SIs). These islets contained a majority of insulin-expressing cells, positive or negative for PDX1 and NKX6.1, and a minority of glucagon- or somatostatin-positive cells. Single, non-aggregated hiPSC-SIs were satisfactorily engrafted onto the iris. They underwent gradual vascularization and progressively increased their light scattering signals, reflecting the abundance of zinc-insulin crystal packaged inside mature insulin secretory granules. Intracameral hiPSC-SIs retrieved from recipients showed enhanced insulin immunofluorescence in correlation with the parallel increase in overall vascularization and light backscattering during the post-transplantation period. This approach enables longitudinal, nondestructive and intravital microimaging of cell fates, engraftment, vascularization and mature insulin secretory granules of single hiPSC-SI grafts, and may offer a feasible and reliable means to screen compounds for promoting in vivo hiPSC-SI maturation.
Collapse
Affiliation(s)
- Kaixuan Zhao
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital L1, Stockholm, Sweden
| | - Yue Shi
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital L1, Stockholm, Sweden
| | - Jia Yu
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital L1, Stockholm, Sweden
| | - Lina Yu
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital L1, Stockholm, Sweden
| | - Amber Mael
- Regenerative Medical Solutions, Inc., Madison, WI, USA
| | - Yuxin Li
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, China
| | | | - Thomas Joyce
- Regenerative Medical Solutions, Inc., Madison, WI, USA
| | - Jon Odorico
- Regenerative Medical Solutions, Inc., Madison, WI, USA
| | - Per-Olof Berggren
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital L1, Stockholm, Sweden
| | - Shao-Nian Yang
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, Karolinska University Hospital L1, Stockholm, Sweden
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, China
| |
Collapse
|
10
|
Human Induced Pluripotent Stem Cell as a Disease Modeling and Drug Development Platform-A Cardiac Perspective. Cells 2021; 10:cells10123483. [PMID: 34943991 PMCID: PMC8699880 DOI: 10.3390/cells10123483] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/03/2021] [Accepted: 12/06/2021] [Indexed: 02/07/2023] Open
Abstract
A comprehensive understanding of the pathophysiology and cellular responses to drugs in human heart disease is limited by species differences between humans and experimental animals. In addition, isolation of human cardiomyocytes (CMs) is complicated because cells obtained by biopsy do not proliferate to provide sufficient numbers of cells for preclinical studies in vitro. Interestingly, the discovery of human-induced pluripotent stem cell (hiPSC) has opened up the possibility of generating and studying heart disease in a culture dish. The combination of reprogramming and genome editing technologies to generate a broad spectrum of human heart diseases in vitro offers a great opportunity to elucidate gene function and mechanisms. However, to exploit the potential applications of hiPSC-derived-CMs for drug testing and studying adult-onset cardiac disease, a full functional characterization of maturation and metabolic traits is required. In this review, we focus on methods to reprogram somatic cells into hiPSC and the solutions for overcome immaturity of the hiPSC-derived-CMs to mimic the structure and physiological properties of the adult human CMs to accurately model disease and test drug safety. Finally, we discuss how to improve the culture, differentiation, and purification of CMs to obtain sufficient numbers of desired types of hiPSC-derived-CMs for disease modeling and drug development platform.
Collapse
|
11
|
Ricci S, Cacialli P. Stem Cell Research Tools in Human Metabolic Disorders: An Overview. Cells 2021; 10:cells10102681. [PMID: 34685661 PMCID: PMC8534517 DOI: 10.3390/cells10102681] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/23/2021] [Accepted: 10/04/2021] [Indexed: 12/20/2022] Open
Abstract
Metabolic disorders are very common in the population worldwide and are among the diseases with the highest health utilization and costs per person. Despite the ongoing efforts to develop new treatments, currently, for many of these disorders, there are no approved therapies, resulting in a huge economic hit and tension for society. In this review, we recapitulate the recent advancements in stem cell (gene) therapy as potential tools for the long-term treatment of both inherited (lysosomal storage diseases) and acquired (diabetes mellitus, obesity) metabolic disorders, focusing on the main promising results observed in human patients and discussing the critical hurdles preventing the definitive jump of this approach from the bench to the clinic.
Collapse
Affiliation(s)
- Serena Ricci
- Department of Cell Physiology and Metabolism, School of Medicine, University of Geneva, Rue Michel Servet 1, 1206 Geneva, Switzerland;
| | - Pietro Cacialli
- Department of Pathology and Immunology, School of Medicine, University of Geneva, Rue Michel Servet 1, 1206 Geneva, Switzerland
- Correspondence:
| |
Collapse
|
12
|
Parsons RF, Baquerizo A, Kirchner VA, Malek S, Desai CS, Schenk A, Finger EB, Brennan TV, Parekh KR, MacConmara M, Brayman K, Fair J, Wertheim JA. Challenges, highlights, and opportunities in cellular transplantation: A white paper of the current landscape. Am J Transplant 2021; 21:3225-3238. [PMID: 34212485 DOI: 10.1111/ajt.16740] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 06/22/2021] [Accepted: 06/22/2021] [Indexed: 02/05/2023]
Abstract
Although cellular transplantation remains a relatively small field compared to solid organ transplantation, the prospects for advancement in basic science and clinical care remain bountiful. In this review, notable historical events and the current landscape of the field of cellular transplantation are reviewed with an emphasis on islets (allo- and xeno-), hepatocytes (including bioartificial liver), adoptive regulatory immunotherapy, and stem cells (SCs, specifically endogenous organ-specific and mesenchymal). Also, the nascent but rapidly evolving field of three-dimensional bioprinting is highlighted, including its major processing steps and latest achievements. To reach its full potential where cellular transplants are a more viable alternative than solid organ transplants, fundamental change in how the field is regulated and advanced is needed. Greater public and private investment in the development of cellular transplantation is required. Furthermore, consistent with the call of multiple national transplant societies for allo-islet transplants, the oversight of cellular transplants should mirror that of solid organ transplants and not be classified under the unsustainable, outdated model that requires licensing as a drug with the Food and Drug Administration. Cellular transplantation has the potential to bring profound benefit through progress in bioengineering and regenerative medicine, limiting immunosuppression-related toxicity, and providing markedly reduced surgical morbidity.
Collapse
Affiliation(s)
- Ronald F Parsons
- Department of Surgery, Emory Transplant Center, Emory University School of Medicine, Atlanta, Georgia
| | - Angeles Baquerizo
- Scripps Center for Cell and Organ Transplantation, La Jolla, California
| | - Varvara A Kirchner
- Division of Transplantation, Department of Surgery, University of Minnesota, Minneapolis, Minnesota
| | - Sayeed Malek
- Division of Transplant Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Chirag S Desai
- Division of Transplantation, Department of Surgery, University of North Carolina, Chapel Hill, North Carolina
| | - Austin Schenk
- Division of Transplantation, Department of Surgery, Ohio State University, Columbus, Ohio
| | - Erik B Finger
- Division of Transplantation, Department of Surgery, University of Minnesota, Minneapolis, Minnesota
| | - Todd V Brennan
- Department of Surgery, Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, California
| | - Kalpaj R Parekh
- Division of Cardiothoracic Surgery, Department of Surgery, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Malcolm MacConmara
- Division of Surgical Transplantation, Department of Surgery, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Kenneth Brayman
- Division of Transplantation, Department of Surgery, University of Virginia, Charlottesville, Virginia
| | - Jeffrey Fair
- Division of Transplant Surgery, Department of Surgery, University of Texas Medical Branch, Galveston, Texas
| | - Jason A Wertheim
- Departments of Surgery and Biomedical Engineering, University of Arizona Health Sciences, Tucson, Arizona
| | | |
Collapse
|
13
|
Wang D, Zhu Y, Huang Y, Zhu J, Zhu B, Zhao Y, Lu Y, Wang Z, Guo Y. Pancreatic Extracellular Matrix/Alginate Hydrogels Provide a Supportive Microenvironment for Insulin-Producing Cells. ACS Biomater Sci Eng 2021; 7:3793-3805. [PMID: 34251797 DOI: 10.1021/acsbiomaterials.1c00269] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Type 1 diabetes mellitus (T1DM), as an autoimmune deficiency disease, is associated with an absolute deficiency of insulin subject to islet β-cell destruction. Insulin-producing cells (IPCs) differentiated from induced pluripotent stem cells are an ideal replacement origin of β-cells, which can be applied for cell transplantation therapies in T1DM. At present, more strategies focus on inducing and differentiating to obtain IPCs; however, the unsatisfactory differentiation efficiency and the lack of ideal carriers for in vivo transplantation limited their application. It is necessary to consider the cell microenvironment by constructing a biomimetic niche to improve the differentiation and transplantation efficiency. The main components of the extracellular matrix derived from pancreatic (the niche of β-cells) decellularization were retained, which could provide the ideal extracellular microenvironment for IPCs. In this research, a hydrogel prepared with alginate (Alg) and the pancreatic extracellular matrix (pECM) was assessed for the beneficial outcomes on encapsulated IPCs. The results showed that pECM/Alg improved the differentiation efficiency and promoted insulin secretion and the expression of insulin-related genes as well. Besides, pECM/Alg-encapsulated IPCs exhibited obvious biocompatibility in vivo, which can prolong the transplantation effect and hypoglycemic function by reducing the inflammatory reaction. RNA-seq indicated that the PI3K/Akt pathway may be related to the improvement of the differentiation efficiency and function of IPCs. In general, the pECM/Alg hydrogel provides an ideal biomimetic microenvironment for IPCs and is suitable for in vivo transplantation.
Collapse
Affiliation(s)
- Dongzhi Wang
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China.,Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P. R China
| | - Yi Zhu
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China.,Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P. R China
| | - Yan Huang
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China.,Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P. R China
| | - Jiachen Zhu
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P. R China.,Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Biwen Zhu
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China.,Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P. R China
| | - Yahong Zhao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Yuhua Lu
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China
| | - Zhiwei Wang
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China
| | - Yibing Guo
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P. R China
| |
Collapse
|
14
|
Marfil-Garza BA, Shapiro AMJ, Kin T. Clinical islet transplantation: Current progress and new frontiers. JOURNAL OF HEPATO-BILIARY-PANCREATIC SCIENCES 2021; 28:243-254. [PMID: 33417749 DOI: 10.1002/jhbp.891] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/12/2020] [Accepted: 12/28/2020] [Indexed: 02/05/2023]
Abstract
Islet transplantation (IT) is now a robust treatment for selected patients with type 1 diabetes suffering from recurrent hypoglycemia and impaired awareness of hypoglycemia. A global soar of clinical islet transplant programs attests to the commitment of many institutions and researchers to advance IT as a potential cure for this devastating disease. However, many challenges limiting the widespread applicability of clinical IT remain. In this review, we will touch on the milestones in the history of IT and its path to clinical success, discuss the current challenges around IT, propose some possible solutions, and elaborate on the frontiers envisioned in the future of clinical IT.
Collapse
Affiliation(s)
| | - Andrew Mark James Shapiro
- Department of Surgery, University of Alberta, Edmonton, AB, Canada
- Clinical Islet Transplant Program, University of Alberta, Edmonton, AB, Canada
| | - Tatsuya Kin
- Clinical Islet Transplant Program, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
15
|
Ma X, Jain NM, Hitscherich P, Seetamraju S, Lee EJ. Stem Cell-Derived Insulin-Producing Cells in 3D Engineered Tissue in a Perfusion Flow Bioreactor. Tissue Eng Part A 2021; 27:1182-1191. [PMID: 33218288 DOI: 10.1089/ten.tea.2020.0231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
To circumvent the lack of donor pancreas, insulin-producing cells (IPCs) derived from pluripotent stem cells emerged as a viable cell source for the treatment of type 1 diabetes. While it has been shown that IPCs can be derived from pluripotent stem cells using various protocols, the long-term viability and functional stability of IPCs in vitro remains a challenge. Thus, the principles of three-dimensional (3D) tissue engineering and a perfusion flow bioreactor were used in this study to establish 3D microenvironment suitable for long-term in vitro culture of IPCs-derived from mouse embryonic stem cells. It was observed that in static 3D culture of IPCs, the viability decreased gradually with longer time in culture. However, when a low flow (0.02 mL/min) was continuously applied to 3D IPC containing tissues, enhanced survival and function of IPCs were demonstrated. IPCs cultured under low flow exhibited a significantly enhanced glucose responsiveness and upregulation of Ins1 compared to that of static culture. In summary, this study demonstrates the feasibility and benefits of 3D engineered tissue environment combined with perfusion flow in vitro for culturing stem cell-derived IPCs.
Collapse
Affiliation(s)
- Xiaotang Ma
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey, USA
| | - Neha M Jain
- Vanderbilt University Medical Center, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee, USA
| | - Pamela Hitscherich
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey, USA
| | - Sahiti Seetamraju
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey, USA
| | - Eun Jung Lee
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey, USA
| |
Collapse
|
16
|
Verhoeff K, Henschke SJ, Marfil-Garza BA, Dadheech N, Shapiro AMJ. Inducible Pluripotent Stem Cells as a Potential Cure for Diabetes. Cells 2021; 10:cells10020278. [PMID: 33573247 PMCID: PMC7911560 DOI: 10.3390/cells10020278] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 01/22/2021] [Accepted: 01/24/2021] [Indexed: 02/07/2023] Open
Abstract
Over the last century, diabetes has been treated with subcutaneous insulin, a discovery that enabled patients to forego death from hyperglycemia. Despite novel insulin formulations, patients with diabetes continue to suffer morbidity and mortality with unsustainable costs to the health care system. Continuous glucose monitoring, wearable insulin pumps, and closed-loop artificial pancreas systems represent an advance, but still fail to recreate physiologic euglycemia and are not universally available. Islet cell transplantation has evolved into a successful modality for treating a subset of patients with ‘brittle’ diabetes but is limited by organ donor supply and immunosuppression requirements. A novel approach involves generating autologous or immune-protected islet cells for transplant from inducible pluripotent stem cells to eliminate detrimental immune responses and organ supply limitations. In this review, we briefly discuss novel mechanisms for subcutaneous insulin delivery and define their shortfalls. We describe embryological development and physiology of islets to better understand their role in glycemic control and, finally, discuss cell-based therapies for diabetes and barriers to widespread use. In response to these barriers, we present the promise of stem cell therapy, and review the current gaps requiring solutions to enable widespread use of stem cells as a potential cure for diabetes.
Collapse
Affiliation(s)
- Kevin Verhoeff
- Department of Surgery, University of Alberta, Edmonton, AB T6G 2B7, Canada;
- Correspondence: ; Tel.: +1-780-984-1836
| | - Sarah J. Henschke
- Department of Emergency Medicine, University of Saskatchewan, Saskatoon, SK S7N 0W8, Canada;
| | | | - Nidheesh Dadheech
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB T6G 2B7, Canada;
| | - Andrew Mark James Shapiro
- FRCS (Eng) FRCSC MSM FCAHS, Clinical Islet Transplant Program, Alberta Diabetes Institute, Department of Surgery, Canadian National Transplant Research Program, Edmonton, AB T6G 2B7, Canada;
| |
Collapse
|
17
|
Baek SE, Ul-Haq A, Kim DH, Choi HW, Kim MJ, Choi HJ, Kim H. Human Organic Anion Transporting Polypeptide 1B3 Applied as an MRI-Based Reporter Gene. Korean J Radiol 2020; 21:726-735. [PMID: 32410411 PMCID: PMC7231618 DOI: 10.3348/kjr.2019.0903] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/10/2020] [Accepted: 01/19/2020] [Indexed: 12/22/2022] Open
Abstract
Objective Recent innovations in biology are boosting gene and cell therapy, but monitoring the response to these treatments is difficult. The purpose of this study was to find an MRI-reporter gene that can be used to monitor gene or cell therapy and that can be delivered without a viral vector, as viral vector delivery methods can result in long-term complications. Materials and Methods CMV promoter-human organic anion transporting polypeptide 1B3 (CMV-hOATP1B3) cDNA or CMV-blank DNA (control) was transfected into HEK293 cells using Lipofectamine. OATP1B3 expression was confirmed by western blotting and confocal microscopy. In vitro cell phantoms were made using transfected HEK293 cells cultured in various concentrations of gadoxetic acid for 24 hours, and images of the phantoms were made with a 9.4T micro-MRI. In vivo xenograft tumors were made by implanting HEK293 cells transfected with CMV-hOATP1B3 (n = 4) or CMV-blank (n = 4) in 8-week-old male nude mice, and MRI was performed before and after intravenous injection of gadoxetic acid (1.2 µL/g). Results Western blot and confocal microscopy after immunofluorescence staining revealed that only CMV-hOATP1B3-transfected HEK293 cells produced abundant OATP1B3, which localized at the cell membrane. OATP1B3 expression levels remained high through the 25th subculture cycle, but decreased substantially by the 50th subculture cycle. MRI of cell phantoms showed that only the CMV-hOATP1B3-transfected cells produced a significant contrast enhancement effect. In vivo MRI of xenograft tumors revealed that only CMV-hOATP1B3-transfected HEK293 tumors demonstrated a T1 contrast effect, which lasted for at least 5 hours. Conclusion The human endogenous OATP1B3 gene can be non-virally delivered into cells to induce transient OATP1B3 expression, leading to gadoxetic acid-mediated enhancement on MRI. These results indicate that hOATP1B3 can serve as an MRI-reporter gene while minimizing the risk of long-term complications.
Collapse
Affiliation(s)
- Song Ee Baek
- Department of Radiology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Asad Ul-Haq
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Dae Hee Kim
- Yonsei University College of Medicine, Seoul, Korea
| | | | - Myeong Jin Kim
- Department of Radiology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Hye Jin Choi
- Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Honsoul Kim
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.
| |
Collapse
|
18
|
Hu M, Cherkaoui I, Misra S, Rutter GA. Functional Genomics in Pancreatic β Cells: Recent Advances in Gene Deletion and Genome Editing Technologies for Diabetes Research. Front Endocrinol (Lausanne) 2020; 11:576632. [PMID: 33162936 PMCID: PMC7580382 DOI: 10.3389/fendo.2020.576632] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 09/17/2020] [Indexed: 12/13/2022] Open
Abstract
The inheritance of variants that lead to coding changes in, or the mis-expression of, genes critical to pancreatic beta cell function can lead to alterations in insulin secretion and increase the risk of both type 1 and type 2 diabetes. Recently developed clustered regularly interspaced short palindromic repeats (CRISPR/Cas9) gene editing tools provide a powerful means of understanding the impact of identified variants on cell function, growth, and survival and might ultimately provide a means, most likely after the transplantation of genetically "corrected" cells, of treating the disease. Here, we review some of the disease-associated genes and variants whose roles have been probed up to now. Next, we survey recent exciting developments in CRISPR/Cas9 technology and their possible exploitation for β cell functional genomics. Finally, we will provide a perspective as to how CRISPR/Cas9 technology may find clinical application in patients with diabetes.
Collapse
Affiliation(s)
- Ming Hu
- Section of Cell Biology and Functional Genomics, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Ines Cherkaoui
- Section of Cell Biology and Functional Genomics, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Shivani Misra
- Metabolic Medicine, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Guy A. Rutter
- Section of Cell Biology and Functional Genomics, Faculty of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
19
|
Budhwar S, Chakraborty M, Sethi K, Chatterjee A. Antidiabetic properties of rice and wheat bran-A review. J Food Biochem 2020; 44:e13424. [PMID: 32761956 DOI: 10.1111/jfbc.13424] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 07/16/2020] [Accepted: 07/16/2020] [Indexed: 01/09/2023]
Abstract
In the present study, the increasing demand for the by-products of rice and wheat, especially their bran part obtained by milling has been discussed along with their properties in controlling diabetes. It is composed of macronutrients and micronutrients, including fibers, and trace elements and different phytochemicals. Previously, they were being used as animal fodder or for other compost matter. Contrarily, it can be utilized for humankind to save world hunger and to cater to the need for extra food demand and eradication of malnourishment, particularly in the developing countries. The bran part can act as a defense against different chronic diseases, particularly diabetes, which accounts for 3.2 million deaths worldwide every year. Keeping this in view, the current review discusses the nutritional composition, biological, and therapeutic properties of rice and wheat bran. PRACTICAL APPLICATIONS: Properly processed agricultural wastes can yield resourceful and economical by-products. The bran part of rice and wheat is such an agricultural byproduct which is cheap and easily available. They contain vast amount of beneficial biochemical constituents. Properly processed bran part can be utilized for preparation of various value-added food products which can save the world hunger, extra food demand, and malnourishment and will be a boon for the developing countries. It can be also useful in combating several chronic diseases including diabetes through dietary intake.
Collapse
Affiliation(s)
- Savita Budhwar
- Department of Nutrition Biology, School of Interdisciplinary and Applied Life Sciences, Central University of Haryana, Mahendergarh, India
| | - Manali Chakraborty
- Department of Nutrition Biology, School of Interdisciplinary and Applied Life Sciences, Central University of Haryana, Mahendergarh, India
| | - Kashika Sethi
- Department of Nutrition Biology, School of Interdisciplinary and Applied Life Sciences, Central University of Haryana, Mahendergarh, India
| | - Arnab Chatterjee
- Department of Nutrition Biology, School of Interdisciplinary and Applied Life Sciences, Central University of Haryana, Mahendergarh, India
| |
Collapse
|
20
|
Li F, Lv Y, Li X, Yang Z, Guo T, Zhang J. Comparative Study of Two Different Islet Transplantation Sites in Mice: Hepatic Sinus Tract vs Splenic Parenchyma. Cell Transplant 2020; 29:963689720943576. [PMID: 32731817 PMCID: PMC7563812 DOI: 10.1177/0963689720943576] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Although 90% of clinical islet transplantations are performed via the portal vein approach, it is still far from the ideal transplant site. Alternative islet transplant sites are promising to reduce the islet dose required to reverse hyperglycemia, thereby improving the efficiency of islet transplantation. The aim of this study was to compare the differences in survival and metabolic function of islet grafts transplanted into the hepatic sinus tract (HST) and the splenic parenchyma (SP). Approximately 300 syngeneic mouse islets were transplanted into the HST (n = 6) and the SP (n = 6) of recipient diabetic mice, respectively. After transplantation, the glycemic control, glucose tolerance, and morphology of islet grafts were evaluated and compared in each group. The nonfasting blood glucose of the two groups of mice receiving islet transplantation gradually decreased to the normal range and sustained for more than 100 d. There is no significant difference in the time required to restore normoglycemia (P > 0.05). The results of the glucose tolerance test showed that the SP group presented a smaller area under the curve than the HST group (P < 0.05). Histopathological results showed that islet grafts in the HST and the SP were characterized with normal islet morphology and robust insulin production. Compared with the HST, islet transplantation in the SP presents better blood glucose regulation, although there is no significant difference in the time required to restore normoglycemia.
Collapse
Affiliation(s)
- Feng Li
- Hepatobiliary Surgery Department, the First Hospital of China Medical University, Shenyang, China
| | - Yi Lv
- Hepatobiliary Surgery Department, the First Hospital of China Medical University, Shenyang, China
| | - Xiaohang Li
- Hepatobiliary Surgery Department, the First Hospital of China Medical University, Shenyang, China
| | - Zhaoming Yang
- Hepatobiliary Surgery Department, the First Hospital of China Medical University, Shenyang, China
| | - Tingwei Guo
- Hepatobiliary Surgery Department, the First Hospital of China Medical University, Shenyang, China
| | - Jialin Zhang
- Hepatobiliary Surgery Department, the First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
21
|
Marfil‐Garza BA, Polishevska K, Pepper AR, Korbutt GS. Current State and Evidence of Cellular Encapsulation Strategies in Type 1 Diabetes. Compr Physiol 2020; 10:839-878. [DOI: 10.1002/cphy.c190033] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
22
|
The Influence of the Flow of Detergent and Donor Characteristics on the Extracellular Matrix Composition After Human Pancreas Decellularization. Transplant Proc 2020; 52:2043-2049. [PMID: 32527472 DOI: 10.1016/j.transproceed.2020.03.053] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 03/10/2020] [Accepted: 03/30/2020] [Indexed: 12/28/2022]
Abstract
INTRODUCTION The extracellular matrix (ECM) consists, among others, of polysaccharides, glycosaminoglycans, and proteins. It is being increasingly used in tissue bioengineering. Obtaining ECM of the highest quality through decellularization is a big challenge because of some differences in organ structure. To deprive organs of the cellular part, chemical, enzymatic, or mechanical methods are used. After decellularization, we get a scaffold made of a variety of proteins, and it is the role of these proteins that can significantly affect the maintenance of the spatial structure and be a suitable environment for cells to rebuild a specific organ. AIM Estimation of the detergent (Triton X-100) flow parameters and anthropometric donors' decellularization process accuracy on the final ECM composition. MATERIALS Five human pancreata, rejected from transplantation, were used for decellularization. All organs were harvested from brain-dead donors age 13 to 60 years. METHODS Decellularization was carried out using the flow method with Triton X-100 as an active agent. The experiment compared 5 different flow values. After decellularization, an assessment of the final DNA concentration and the protein composition was performed. Results were compared to anthropometric data of donors. In addition, a microscopic analysis was also carried out. RESULTS The best results were obtained using a flow of 120 mL/minute. A higher detergent flow was associated with a lower concentration of residual DNA in scaffold. Analysis of the protein profile with anthropometric data has shown that LAM A2 was increasing with age and LAMA5 was decreasing. Being overweight was associated with a higher proportion of COL1 and 4 and a smaller proportion of COL6.
Collapse
|
23
|
Magisson J, Sassi A, Xhema D, Kobalyan A, Gianello P, Mourer B, Tran N, Burcez CT, Bou Aoun R, Sigrist S. Safety and function of a new pre-vascularized bioartificial pancreas in an allogeneic rat model. J Tissue Eng 2020; 11:2041731420924818. [PMID: 32523669 PMCID: PMC7257875 DOI: 10.1177/2041731420924818] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 04/18/2020] [Indexed: 12/17/2022] Open
Abstract
Cell encapsulation could overcome limitations of free islets transplantation but is currently limited by inefficient cells immune protection and hypoxia. As a response to these challenges, we tested in vitro and in vivo the safety and efficacy of a new macroencapsulation device named MailPan®. Membranes of MailPan® device were tested in vitro in static conditions. Its bio-integration and level of oxygenation was assessed after implantation in non-diabetic rats. Immune protection properties were also assessed in rat with injection in the device of allogeneic islets with incompatible Major Histocompatibility Complex. Finally, function was assessed in diabetic rats with a Beta cell line injected in MailPan®. In vitro, membranes of the device showed high permeability to glucose, insulin, and rejected IgG. In rat, the device displayed good bio-integration, efficient vascularization, and satisfactory oxygenation (>5%), while positron emission tomography (PET)-scan and angiography also highlighted rapid exchanges between blood circulation and the MailPan®. The device showed its immune protection properties by preventing formation, by the rat recipient, of antibodies against encapsulated allogenic islets. Injection of a rat beta cell line into the device normalized fasting glycemia of diabetic rat with retrieval of viable cell clusters after 2 months. These data suggest that MailPan® constitutes a promising encapsulation device for widespread use of cell therapy for type 1 diabetes.
Collapse
Affiliation(s)
| | | | - Daela Xhema
- Laboratory of Experimental Surgery, Université Catholique de Louvain, Brussels, Belgium
| | | | - Pierre Gianello
- Laboratory of Experimental Surgery, Université Catholique de Louvain, Brussels, Belgium
| | - Brice Mourer
- Ecole de Chirurgie de Nancy-Lorraine, Vandoeuvre-lès-Nancy, France
| | - Nguyen Tran
- Ecole de Chirurgie de Nancy-Lorraine, Vandoeuvre-lès-Nancy, France
| | | | | | | |
Collapse
|
24
|
Black L, Zorina T. Cell-based immunomodulatory therapy approaches for type 1 diabetes mellitus. Drug Discov Today 2020; 25:380-391. [DOI: 10.1016/j.drudis.2019.11.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 11/11/2019] [Accepted: 11/30/2019] [Indexed: 12/14/2022]
|
25
|
Iosim S, MacKay M, Westover C, Mason CE. Translating current biomedical therapies for long duration, deep space missions. PRECISION CLINICAL MEDICINE 2019; 2:259-269. [PMID: 31886035 PMCID: PMC6927098 DOI: 10.1093/pcmedi/pbz022] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 11/05/2019] [Accepted: 11/06/2019] [Indexed: 12/12/2022] Open
Abstract
It is been shown that spaceflight-induced molecular, cellular, and physiologic changes cause alterations across many modalities of the human body, including cardiovascular, musculoskeletal, hematological, immunological, ocular, and neurological systems. The Twin Study, a multi-year, multi-omic study of human response to spaceflight, provided detailed and comprehensive molecular and cellular maps of the human response to radiation, microgravity, isolation, and stress. These rich data identified epigenetic, gene expression, inflammatory, and metabolic responses to spaceflight, facilitating a better biomedical roadmap of features that should be monitored and safe-guarded in upcoming missions. Further, by exploring new developments in pre-clinical models and clinical trials, we can begin to design potential cellular interventions for exploration-class missions to Mars and potentially farther. This paper will discuss the overall risks astronauts face during spaceflight, what is currently known about human response to these risks, what pharmaceutical interventions exist for use in space, and which tools of precision medicine and cellular engineering could be applied to aerospace and astronaut medicine.
Collapse
Affiliation(s)
- Sonia Iosim
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10021, USA
| | - Matthew MacKay
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10021, USA.,The WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY 10021, USA
| | - Craig Westover
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10021, USA
| | - Christopher E Mason
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10021, USA.,The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021, USA.,The Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA.,The WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY 10021, USA
| |
Collapse
|
26
|
Barcia Durán JG, Lis R, Rafii S. Haematopoietic stem cell reprogramming and the hope for a universal blood product. FEBS Lett 2019; 593:3253-3265. [PMID: 31725897 DOI: 10.1002/1873-3468.13681] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 10/31/2019] [Accepted: 11/12/2019] [Indexed: 12/13/2022]
Abstract
Haematopoietic stem cells (HSCs) are the only adult stem cells with a demonstrated clinical use, even though a tractable method to maintain and expand human HSCs in vitro has not yet been found. Owing to the introduction of transplantation strategies for the treatment of haematological malignancies and, more recently, the promise of gene therapy, the need to improve the generation, manipulation and scalability of autologous or allogeneic HSCs has risen steeply over the past decade. In that context, reprogramming strategies based on the expression of exogenous transcription factors have emerged as a means to produce functional HSCs in vitro. These approaches largely stem from the assumption that key master transcription factors direct the expression of downstream target genes thereby triggering haematopoiesis. Both somatic and pluripotent cells have been used to this end, yielding variable results in terms of haematopoietic phenotype and functionality. Here, we present an overview of the haematopoietic reprogramming methods reported to date, provide the appropriate historical context and offer some critical insight about where the field stands at present.
Collapse
Affiliation(s)
- José Gabriel Barcia Durán
- Division of Regenerative Medicine, Department of Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, NY, USA
| | - Raphaël Lis
- Division of Regenerative Medicine, Department of Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, NY, USA.,Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine and Infertility, Weill Cornell Medicine, New York, NY, USA
| | - Shahin Rafii
- Division of Regenerative Medicine, Department of Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|