1
|
Peng X, Rao G, Li X, Tong N, Tian Y, Fu X. Preclinical models for Type 1 Diabetes Mellitus - A practical approach for research. Int J Med Sci 2023; 20:1644-1661. [PMID: 37859703 PMCID: PMC10583179 DOI: 10.7150/ijms.86566] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Accepted: 09/18/2023] [Indexed: 10/21/2023] Open
Abstract
Numerous preclinical models have been developed to advance biomedical research in type 1 diabetes mellitus (T1DM). They are essential for improving our knowledge of T1DM development and progression, allowing researchers to identify potential therapeutic targets and evaluate the effectiveness of new medications. A deeper comprehension of these models themselves is critical not only to determine the optimal strategies for their utilization but also to fully unlock their potential applications in both basic and translational research. Here, we will comprehensively summarize and discuss the applications, advantages, and limitations of the commonly used animal models for human T1DM and also overview the up-to-date human tissue bioengineering models for the investigation of T1DM. By combining these models with a better understanding of the pathophysiology of T1DM, we can enhance our insights into disease initiation and development, ultimately leading to improved therapeutic responses and outcomes.
Collapse
Affiliation(s)
| | | | | | | | | | - Xianghui Fu
- Department of Endocrinology and Metabolism, Center for Diabetes Metabolism Research, Cancer Center West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
| |
Collapse
|
2
|
Wu T, Cai Z, Niu F, Qian B, Sun P, Yang N, Pang J, Mei H, Chang X, Chen F, Zhu Y, Li Y, Wu FG, Zhang Y, Lei T, Han X. Lentinan confers protection against type 1 diabetes by inducing regulatory T cell in spontaneous non-obese diabetic mice. Nutr Diabetes 2023; 13:4. [PMID: 37031163 PMCID: PMC10082833 DOI: 10.1038/s41387-023-00233-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 02/10/2023] [Accepted: 03/16/2023] [Indexed: 04/10/2023] Open
Abstract
BACKGROUND Lentinan (LNT) is a complex fungal component that possesses effective antitumor and immunostimulating properties. However, there is a paucity of studies regarding the effects and mechanisms of LNT on type 1 diabetes. OBJECTIVE In the current study, we investigated whether an intraperitoneal injection of LNT can diminish the risk of developing type 1 diabetes (T1D) in non-obese diabetic (NOD) mice and further examined possible mechanisms of LNT's effects. METHODS Pre-diabetic female NOD mice 8 weeks of age, NOD mice with 140-160 mg/dL, 200-230 mg/dL or 350-450 mg/dL blood glucose levels were randomly divided into two groups and intraperitoneally injected with 5 mg/kg LNT or PBS every other day. Then, blood sugar levels, pancreas slices, spleen, PnLN and pancreas cells from treatment mice were examined. RESULTS Our results demonstrated that low-dosage injections (5 mg/kg) of LNT significantly suppressed immunopathology in mice with autoimmune diabetes but increased the Foxp3+ regulatory T cells (Treg cells) proportion in mice. LNT treatment induced the production of Tregs in the spleen and PnLN cells of NOD mice in vitro. Furthermore, the adoptive transfer of Treg cells extracted from LNT-treated NOD mice confirmed that LNT induced Treg function in vivo and revealed an enhanced suppressive capacity as compared to the Tregs isolated from the control group. CONCLUSION LNT was capable of stimulating the production of Treg cells from naive CD4 + T cells, which implies that LNT exhibits therapeutic values as a tolerogenic adjuvant and may be used to reverse hyperglycaemia in the early and late stages of T1D.
Collapse
Affiliation(s)
- Tijun Wu
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, 211166, China
| | - Zhi Cai
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Fandi Niu
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, 211166, China
| | - Bin Qian
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, 211166, China
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 2111198, China
| | - Peng Sun
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, 211166, China
| | - Nan Yang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, 211166, China
| | - Jing Pang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200001, China
| | - Hongliang Mei
- Department of Pharmacy, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Xiaoai Chang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, 211166, China
| | - Fang Chen
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, 211166, China
| | - Yunxia Zhu
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, 211166, China
| | - Yating Li
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, 211166, China
| | - Fu-Gen Wu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Yaqin Zhang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, 211166, China.
| | - Ting Lei
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Xiao Han
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
3
|
Isolation and Purification of Human Pancreatic Islets. Methods Mol Biol 2022; 2592:219-232. [PMID: 36507997 DOI: 10.1007/978-1-0716-2807-2_16] [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: 12/15/2022]
Abstract
Successful islet isolation is the key to islet transplantation in diabetic patients. However, islet isolation is a technically complex and time-consuming manual process. Optimizing the islet isolation process can improve islet yield and quality, reduce operators, and thus reduce costs.The isolation and purification of human islets include pancreas acquisition and preservation, pancreas digestion, islet purification, islet culture, and islet quality identification. Briefly, after the duodenum was removed, the pancreas was trimmed, the main pancreatic duct was intubated at the distal end of the pancreatic head, collagenase was injected into the pancreatic duct, and the perfused pancreatic tissue was cut and then digested in a Ricordi chamber. A digestion temperature of 37 °C was continuously used to assess the number of samples and the integrity of the lysed and released islets. At the end of the digestion process, collect the digested tissue in a 500 mL centrifuge tube prefilled with 25 mL of cold (4 °C) human serum albumin and centrifuge twice at 150 g for 3 min. After mixing with UW solution as islet storage solution, put it on ice (shake occasionally to prevent clumping) after 30 min. Digested pancreatic tissue was centrifuged at 2200 rpm for 5 min in a COBE 2991 cell processor to isolate islets from exocrine tissue using a continuous density gradient. The purified islet fractions were washed twice in HBSS supplemented with 10% human serum albumin and finally collected in CMRL1066 medium supplemented with the corresponding liquid. The purity of purified islets was calculated by DTZ staining, the survival rate of islets was calculated by FDA/PI staining, and islet function was determined by in vitro glucose-stimulated insulin secretion test.
Collapse
|
4
|
Liebman C, Loya S, Lawrence M, Bashoo N, Cho M. Stimulatory responses in α- and β-cells by near-infrared (810 nm) photobiomodulation. JOURNAL OF BIOPHOTONICS 2022; 15:e202100257. [PMID: 34837336 DOI: 10.1002/jbio.202100257] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 11/22/2021] [Accepted: 11/23/2021] [Indexed: 06/13/2023]
Abstract
Significant efforts have been committed to better understand and regulate insulin secretion as it has direct implications on diabetes. The first phase of biphasic insulin secretion in response to glucose lasts about 10 minutes, followed by a more sustained release persisting several hours. Attenuated insulin release in the first phase is typically associated with abnormal β-cells. While near-infrared photobiomodulation (PBM) demonstrates potential for multiple therapeutic applications, photostimulatory effects on α- and β-cells remain to be further elucidated. Herein, we demonstrate that 810 nm PBM exposure at fluence of 9 J/cm2 can elevate the intracellular reactive oxygen species within 15 minutes following photostimulation. In addition, calcium spiking showed an approximately 3-fold increase in both ATC1 (α-cells) and BTC6 (β-cells) and correlates with hormone secretion in response to PBM stimulation. Our findings could lay a foundation for the development of non-biologic therapeutics that can augment islet transplantation.
Collapse
Affiliation(s)
- Caleb Liebman
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, USA
| | - Sheccid Loya
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, USA
| | | | | | - Michael Cho
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, USA
| |
Collapse
|
5
|
Chung WY, Pollard CA, Kumar R, Drogemuller CJ, Naziruddin B, Stover C, Issa E, Isherwood J, Cooke J, Levy MF, Coates PTH, Garcea G, Dennison AR. A comparison of the inflammatory response following autologous compared with allogenic islet cell transplantation. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:98. [PMID: 33569400 PMCID: PMC7867892 DOI: 10.21037/atm-20-3519] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background The initial response to islet transplantation and the subsequent acute inflammation is responsible for significant attrition of islets following both autologous and allogenic procedures. This multicentre study compares this inflammatory response using cytokine profiles and complement activation. Methods Inflammatory cytokine and complement pathway activity were examined in two cohorts of patients undergoing total pancreatectomy followed either by autologous (n=11) or allogenic (n=6) islet transplantation. Two patients who underwent total pancreatectomy alone (n=2) served as controls. Results The peak of cytokine production occurred immediately following induction of anaesthesia and during surgery. There was found to be a greater elevation of the following cytokines: TNF-alpha (P<0.01), MCP-1 (P=0.0013), MIP-1α (P=0.001), MIP-1β (P=0.00020), IP-10 (P=0.001), IL-8 (P=0.004), IL-1α (P=0.001), IL-1ra (0.0018), IL-10 (P=0.001), GM-CSF (P=0.001), G-CSF (P=0.0198), and Eotaxin (P=0.01) in the allogenic group compared to autografts and controls. Complement activation and consumption was observed in all three pathways, and there were no significant differences in between the groups although following allogenic transplantation ∆IL-10 and ∆VEGF levels were significantly elevated those patients who became insulin-independent compared with those who were insulin-dependent. Conclusions The cytokine profiles following islet transplantation suggests a significantly greater acute inflammatory response following allogenic islet transplantation compared with auto-transplantation although a significant, non-specific inflammatory response occurs following both forms of islet transplantation.
Collapse
Affiliation(s)
- Wen Yuan Chung
- Department of Hepatobiliary and Pancreatic Surgery, Leicester General Hospital, Leicester, UK
| | - Cristina A Pollard
- Department of Hepatobiliary and Pancreatic Surgery, Leicester General Hospital, Leicester, UK
| | - Rohan Kumar
- Department of Hepatobiliary and Pancreatic Surgery, Leicester General Hospital, Leicester, UK
| | | | | | - Cordula Stover
- Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, UK
| | - Eyad Issa
- Department of Hepatobiliary and Pancreatic Surgery, Leicester General Hospital, Leicester, UK
| | - John Isherwood
- Department of Hepatobiliary and Pancreatic Surgery, Leicester General Hospital, Leicester, UK
| | - Jill Cooke
- Department of Hepatobiliary and Pancreatic Surgery, Leicester General Hospital, Leicester, UK
| | - Marlon F Levy
- Baylor Research Institute, Dallas & Fort Worth, TX, USA
| | - P Toby H Coates
- Australian Islet Consortium, Royal Adelaide Hospital, South Australia, Australia
| | - Giuseppe Garcea
- Department of Hepatobiliary and Pancreatic Surgery, Leicester General Hospital, Leicester, UK
| | - Ashley R Dennison
- Department of Hepatobiliary and Pancreatic Surgery, Leicester General Hospital, Leicester, UK
| |
Collapse
|
6
|
Narayanan S, Bhutiani N, Adamson DT, Jones CM. Pancreatectomy, Islet Cell Transplantation, and Nutrition Considerations. Nutr Clin Pract 2020; 36:385-397. [PMID: 33002260 DOI: 10.1002/ncp.10578] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Pancreatic islet transplantation is a reliable approach for treating insulin-deficient diabetes. This established β-cell replacement approach has shown considerable improvements in the last 2 decades. It has helped achieve metabolic homeostasis and safe outcomes for a subset of patients with type 1 diabetes and severe pancreatitis. Nutrition support, until recently, was considered as a secondary factor, merely identified as a means of providing all the necessary nutrients for such patients. However, new literature suggests that several factors, such as the route, timing, quantity, and composition of all the nutrients administered, have key disease-altering properties and are vital during the perioperative management of such patients. This review will highlight the benefits of performing the clinical islet transplantation on a subgroup of patients with type 1 diabetes and pancreatitis and summarize new data that identify the pivotal role of nutrition support as a critical intervention in their management.
Collapse
Affiliation(s)
- Siddharth Narayanan
- Division of Transplantation, Hiram C. Polk Jr. MD Department of Surgery, University of Louisville, Louisville, Kentucky, USA
| | - Neal Bhutiani
- Division of Transplantation, Hiram C. Polk Jr. MD Department of Surgery, University of Louisville, Louisville, Kentucky, USA
| | - Dylan T Adamson
- Division of Transplantation, Hiram C. Polk Jr. MD Department of Surgery, University of Louisville, Louisville, Kentucky, USA
| | - Christopher M Jones
- Division of Transplantation, Hiram C. Polk Jr. MD Department of Surgery, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
7
|
Ludwig B, Wolf E. Transplantation und künstliches Pankreas. DIABETOLOGE 2020. [DOI: 10.1007/s11428-020-00670-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
8
|
Weir GC, Gaglia J, Bonner-Weir S. Inadequate β-cell mass is essential for the pathogenesis of type 2 diabetes. Lancet Diabetes Endocrinol 2020; 8:249-256. [PMID: 32006519 PMCID: PMC7098467 DOI: 10.1016/s2213-8587(20)30022-x] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 11/19/2019] [Accepted: 12/03/2019] [Indexed: 12/16/2022]
Abstract
For patients with type 1 diabetes, it is accepted among the scientific community that there is a marked reduction in β-cell mass; however, with type 2 diabetes, there is disagreement as to whether this reduction in mass occurs in every case. Some have argued that β-cell mass in some patients with type 2 diabetes is normal and that the cause of the hyperglycaemia in these patients is a functional abnormality of insulin secretion. In this Personal View, we argue that a deficient β-cell mass is essential for the development of type 2 diabetes. The main point is that there are enormous (≥10 fold) variations in insulin sensitivity and insulin secretion in the general population, with a very close correlation between these two factors for any individual. Although β-cell mass cannot be accurately measured in living patients, it is highly likely that it too is highly correlated with insulin sensitivity and secretion. Thus, our argument is that a person with type 2 diabetes can have a β-cell mass that is the same as a person without type 2 diabetes, but because they are insulin resistant, the mass is inadequate and responsible for their diabetes. Because the abnormal insulin secretion of diabetes is caused by dysglycaemia and can be largely reversed with glycaemic control, it is a less serious problem than the reduction in β-cell mass, which is far more difficult to restore.
Collapse
Affiliation(s)
- Gordon C Weir
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA.
| | - Jason Gaglia
- Section on Immunobiology, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Susan Bonner-Weir
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
9
|
Elsharkawi I, Parambath D, Saber-Ayad M, Khan AA, El-Serafi AT. Exploring the effect of epigenetic modifiers on developing insulin-secreting cells. Hum Cell 2019; 33:1-9. [PMID: 31755075 DOI: 10.1007/s13577-019-00292-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 10/11/2019] [Indexed: 12/24/2022]
Abstract
Diabetes is a worldwide health problem with increasing incidence. The current management modalities did not succeed to decrease comorbidities. This study aimed at enhancing the regenerative solution for diabetes by improving the differentiation of mesenchymal stromal cells (MSC) into glucose-sensitive, insulin-secreting cells through an epigenetic modification approach. A 3-day treatment protocol with the epigenetic modifiers, either decitabine (5-aza-2'-deoxycytidine; Aza); a DNA methylation inhibitor or Vorinostat (suberoylanilide hydroxamic acid; SAHA); a histone deacetylase inhibitor was added to two different human stem cell lines. The cells followed a multi-step differentiation protocol that provided the critical triggers in a temporal approach. Aza-pretreated group showed higher intracellular expression of insulin and the transcription factor 'PDX-1'. The cells responded to the high glucose challenge by secreting insulin in the media, as shown by ELISA. Gene expression showed induction of the genes for insulin, the glucose transporter 2, glucokinase, as well as the transcription factors MafA and NKX6.1. Although SAHA showed upregulation of insulin secretion, in comparison to control, the cells could not respond to the high glucose challenge. Interestingly, Aza-treated cells showed a significant decrease in the global DNA methylation level at the end of the culture. In conclusion, this additional step with Aza could enhance the response of MSC to the classical differentiation protocol for insulin-secreting cells and may help in establishing a regenerative solution for patients with diabetes.
Collapse
Affiliation(s)
| | | | - Maha Saber-Ayad
- College of Medicine, University of Sharjah, Sharjah, UAE.,Sharjah Institute for Medical Research, University of Sharjah, Sharjah, UAE.,College of Medicine, Cairo University, Cairo, Egypt
| | - Amir Ali Khan
- Department of Applied Biology, College of Science, University of Sharjah, Sharjah, UAE
| | - Ahmed T El-Serafi
- College of Medicine, University of Sharjah, Sharjah, UAE. .,Faculty of Medicine, Suez Canal University, Ismailia, Egypt. .,Department of Hand Surgery and Plastic Surgery and Burns, 401A, Building 462, Floor 11, Linköping University Hospital, P. O. Box: 581 85, Linköping, Sweden.
| |
Collapse
|
10
|
Kanzawa T, Hirai T, Fukuda H, Katsumata H, Ishii R, Ikemiyagi M, Ishii Y, Saiga K, Okumi M, Tanabe K. Combination therapy of an iNKT cell ligand and CD40-CD154 blockade establishes islet allograft acceptance in nonmyeloablative bone marrow transplant recipients. Acta Diabetol 2019; 56:541-550. [PMID: 30758788 DOI: 10.1007/s00592-019-01289-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 01/09/2019] [Indexed: 01/16/2023]
Abstract
AIMS Islet transplantation is an effective therapeutic option for type 1 diabetes. Although maintenance immunosuppression therapy is required to prevent allogeneic rejection and recurrence of autoimmunity, long-term allograft survival has not yet been achieved partly because of its adverse effects. The induction of donor-specific immunotolerance is a promising approach for long-term allograft survival without maintenance immunosuppression therapy. We previously reported that combination therapy using a liposomal ligand for invariant natural killer T cells, RGI-2001, and anti-CD154 antibody established mixed hematopoietic chimerism for the induction of donor-specific immunotolerance. This study investigated whether the protocol could promote islet allograft acceptance in experimental diabetes. METHODS Streptozotocin-induced diabetic BALB/c mice were transplanted with bone marrow cells from C57BL/6 donors and received combination therapy of RGI-2001 and anti-CD154 antibody after 3-Gy total body irradiation. 3 Weeks after bone marrow transplantation, islets isolated from C57BL/6 donors were transplanted under the kidney capsule. RESULTS Mixed chimerism was established in diabetic mice receiving the tolerance induction protocol. After islet transplantation, blood glucose levels improved and normoglycemia persisted for over 100 days. Hyperglycemia recurred after islet grafts were removed. Histopathological examinations showed insulin-positive staining and absence of cellular infiltration in the islet grafts. T cells of recipients showed donor-specific hyporesponsiveness, and anti-donor antibodies were not detected. CONCLUSIONS The tolerance induction protocol with combination therapy of RGI-2001 and anti-CD154 antibody promoted islet allograft acceptance in a mouse diabetic model. This protocol may be clinically applied to islet transplantation for type 1 diabetes mellitus.
Collapse
Affiliation(s)
- Taichi Kanzawa
- Department of Urology, Tokyo Women's Medical University, Tokyo, Japan
| | - Toshihito Hirai
- Department of Urology, Tokyo Women's Medical University, Tokyo, Japan.
| | - Hironori Fukuda
- Department of Urology, Tokyo Women's Medical University, Tokyo, Japan
| | - Haruki Katsumata
- Department of Urology, Tokyo Women's Medical University, Tokyo, Japan
| | - Rumi Ishii
- Department of Urology, Tokyo Women's Medical University, Tokyo, Japan
| | - Masako Ikemiyagi
- Department of Urology, Tokyo Women's Medical University, Tokyo, Japan
| | - Yasuyuki Ishii
- Vaccine Innovation Laboratory, RIKEN Cluster for Science, Technology and Innovation Hub (RCSTI), Yokohama, Japan
- REGiMMUNE Corporation, Tokyo, Japan
| | - Kan Saiga
- Department of Urology, Tokyo Women's Medical University, Tokyo, Japan
- Department of Urology, Jyoban Hospital of Tokiwa Foundation, Fukushima, Japan
| | - Masayoshi Okumi
- Department of Urology, Tokyo Women's Medical University, Tokyo, Japan
| | - Kazunari Tanabe
- Department of Urology, Tokyo Women's Medical University, Tokyo, Japan
| |
Collapse
|
11
|
Perez-Basterrechea M, Esteban MM, Vega JA, Obaya AJ. Tissue-engineering approaches in pancreatic islet transplantation. Biotechnol Bioeng 2018; 115:3009-3029. [PMID: 30144310 DOI: 10.1002/bit.26821] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 08/08/2018] [Accepted: 08/14/2018] [Indexed: 12/15/2022]
Abstract
Pancreatic islet transplantation is a promising alternative to whole-pancreas transplantation as a treatment of type 1 diabetes mellitus. This technique has been extensively developed during the past few years, with the main purpose of minimizing the complications arising from the standard protocols used in organ transplantation. By using a variety of strategies used in tissue engineering and regenerative medicine, pancreatic islets have been successfully introduced in host patients with different outcomes in terms of islet survival and functionality, as well as the desired normoglycemic control. Here, we describe and discuss those strategies to transplant islets together with different scaffolds, in combination with various cell types and diffusible factors, and always with the aim of reducing host immune response and achieving islet survival, regardless of the site of transplantation.
Collapse
Affiliation(s)
- Marcos Perez-Basterrechea
- Unidad de Terapia Celular y Medicina Regenerativa, Servicio de Hematología y Hemoterapia, Hospital Universitario Central de Asturias (HUCA), Oviedo, Spain.,Plataforma de Terapias Avanzadas, Instituto de Investigación Biosanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Manuel M Esteban
- Departamento de Biología Funcional, Universidad de Oviedo, Oviedo, Spain
| | - Jose A Vega
- Departamento de Morfología y Biología Celular, Universidad de Oviedo, Oviedo, Spain.,Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago, Chile
| | - Alvaro J Obaya
- Departamento de Biología Funcional, Universidad de Oviedo, Oviedo, Spain
| |
Collapse
|
12
|
Green AD, Vasu S, Flatt PR. Cellular models for beta-cell function and diabetes gene therapy. Acta Physiol (Oxf) 2018; 222. [PMID: 29226587 DOI: 10.1111/apha.13012] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 11/29/2017] [Accepted: 12/01/2017] [Indexed: 02/06/2023]
Abstract
Diabetes is characterized by the destruction and/or relative dysfunction of insulin-secreting beta-cells in the pancreatic islets of Langerhans. Consequently, considerable effort has been made to understand the physiological processes governing insulin production and secretion in these cells and to elucidate the mechanisms involved in their deterioration in the pathogenesis of diabetes. To date, considerable research has exploited clonal beta-cell lines derived from rodent insulinomas. Such cell lines have proven to be a great asset in diabetes research, in vitro drug testing, and studies of beta-cell physiology and provide a sustainable, and in many cases, more practical alternative to the use of animals or primary tissue. However, selection of the most appropriate rodent beta cell line is often challenging and no single cell line entirely recapitulates the properties of human beta-cells. The generation of stable human beta-cell lines would provide a much more suitable model for studies of human beta-cell physiology and pathology and could potentially be used as a readily available source of implantable insulin-releasing tissue for cell-based therapies of diabetes. In this review, we discuss the history, development, functional characteristics and use of available clonal rodent beta-cell lines, as well as reflecting on recent advances in the generation of human-derived beta-cell lines, their use in research studies and their potential for cell therapy of diabetes.
Collapse
Affiliation(s)
- A. D. Green
- SAAD Centre for Pharmacy & Diabetes; School of Biomedical Sciences; University of Ulster; Coleraine UK
| | - S. Vasu
- SAAD Centre for Pharmacy & Diabetes; School of Biomedical Sciences; University of Ulster; Coleraine UK
- Cell Growth and Metabolism Section; Diabetes, Endocrinology, and Obesity Branch; NIDDK; National Institutes of Health; Bethesda MD USA
| | - P. R. Flatt
- SAAD Centre for Pharmacy & Diabetes; School of Biomedical Sciences; University of Ulster; Coleraine UK
| |
Collapse
|
13
|
Hao W, Wookwyk A, Beam C, Bahnson HT, Palmer JP, Greenbaum CJ. Assessment of β Cell Mass and Function by AIRmax and Intravenous Glucose in High-Risk Subjects for Type 1 Diabetes. J Clin Endocrinol Metab 2017; 102:4428-4434. [PMID: 29040630 PMCID: PMC5718693 DOI: 10.1210/jc.2017-01713] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 10/04/2017] [Indexed: 01/13/2023]
Abstract
CONTEXT There is little information regarding β cell mass in individuals at early stages of type 1 diabetes (T1D). OBJECTIVE To investigate both acute insulin response to arginine at hyperglycemia (AIRmax), as a correlate of β cell mass, and β cell function by the intravenous glucose tolerance test (IVGTT) in subjects at early stages of T1D. DESIGN/SETTING/PARTICIPANTS Forty subjects were enrolled: (1) low-risk group: relatives of patients with T1D with 0 to 1 antibody (n = 21) and (2) high-risk group: relatives with ≥2 antibodies (n = 19). MAIN OUTCOME MEASURE Acute insulin and C-peptide responses to IVGTT and to AIRmax. Participants underwent two IVGTT and AIRmax procedures on different days. RESULTS AIRmax was reproducible, well tolerated, and correlated to first-phase insulin response (FPIR) from IVGTT (r = 0.779). The high-risk group had greater impaired β cell function compared with the low-risk group, determined both by lower mean FPIR and a greater number of subjects below an established threshold for abnormal function [10 of 19 (52.6%) versus 4 of 21 (19%)]. There was a heterogeneous AIRmax response in these subjects with low FPIR, ranging from 38 to 250 μU/mL. CONCLUSIONS There is significant variation in insulin secretory reserve as assessed by AIRmax in family members with low β cell function assessed by FPIR. As AIRmax is a functional measure of β cell mass, these data suggest heterogeneity in disease pathogenesis in which mass is preserved in relation to function in some individuals. The tolerability and reproducibility of AIRmax suggest it could be a useful stratification measure in clinical trials of disease-modifying therapy.
Collapse
Affiliation(s)
- Wei Hao
- Diabetes Clinical Research Program, Benaroya Research Institute at Virginia Mason, Seattle, Washington 98101
| | - Alyssa Wookwyk
- Division of Epidemiology and Biostatistics, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, Michigan 49007
| | - Craig Beam
- Division of Epidemiology and Biostatistics, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, Michigan 49007
| | - Henry T. Bahnson
- Diabetes Clinical Research Program, Benaroya Research Institute at Virginia Mason, Seattle, Washington 98101
| | - Jerry P. Palmer
- Department of Veterans Affairs, Puget Sound Health Care System, and University of Washington, Seattle, Washington 98195
| | - Carla J. Greenbaum
- Diabetes Clinical Research Program, Benaroya Research Institute at Virginia Mason, Seattle, Washington 98101
| |
Collapse
|
14
|
Espes D, Lau J, Carlsson PO. MECHANISMS IN ENDOCRINOLOGY: Towards the clinical translation of stem cell therapy for type 1 diabetes. Eur J Endocrinol 2017; 177:R159-R168. [PMID: 28487297 DOI: 10.1530/eje-17-0080] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 04/24/2017] [Accepted: 05/08/2017] [Indexed: 01/21/2023]
Abstract
Insulin-producing cells derived from human embryonic stem cells (hESCs) or induced pluripotent stem cells (iPSCs) have for long been a promising, but elusive treatment far from clinical translation into type 1 diabetes therapy. However, the field is now on the verge of moving such insulin-producing cells into clinical trials. Although stem cell therapies provide great opportunities, there are also potential risks such as teratoma formation associated with the treatment. Many considerations are needed on how to proceed with clinical translation, including whether to use hESCs or iPSCs, and whether encapsulation of tissue will be needed. This review aims to give an overview of the current knowledge of stem cell therapy outcomes in animal models of type 1 diabetes and a proposed road map towards the clinical setting with special focus on the potential risks and hurdles which needs to be considered. From a clinical point of view, transplantation of insulin-producing cells derived from stem cells must be performed without immune suppression in order to be an attractive treatment option. Although costly and highly labour intensive, patient-derived iPSCs would be the only solution, if not clinically successful encapsulation or tolerance induction protocols are introduced.
Collapse
Affiliation(s)
- Daniel Espes
- Departments of Medical Cell Biology
- Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Joey Lau
- Departments of Medical Cell Biology
| | - Per-Ola Carlsson
- Departments of Medical Cell Biology
- Medical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
15
|
Pereira LX, Viana CTR, Orellano LAA, Almeida SA, Vasconcelos AC, Goes ADM, Birbrair A, Andrade SP, Campos PP. Synthetic matrix of polyether-polyurethane as a biological platform for pancreatic regeneration. Life Sci 2017; 176:67-74. [PMID: 28336399 DOI: 10.1016/j.lfs.2017.03.015] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 03/17/2017] [Accepted: 03/19/2017] [Indexed: 02/07/2023]
Abstract
AIMS Several alternative cellular approaches using biomaterials to host insulin-producing cells derived from stem cells have been developed to overcome the limitations of type 1 diabetes treatment (exogenous insulin injection). However, none seem to fulfill all requirements needed to induce pancreatic cells successful colonization of the scaffolds. Here, we report a polymeric platform adherent to the native mice pancreas filled with human adipose stem cells (hASCs) that was able to induce growth of pancreatic parenchyma. MAIN METHODS Synthetic polyether-polyurethane discs were placed adjacent to pancreas of normoglycemic and streptozotocin-induced diabetic mice. At day 4 post implantation, 1×106 hASCs were injected intra-implant in groups of normoglycemic and diabetic mice. Immunohistochemistry analysis of the implants was performed to identify insulin positive cells in the newly formed tissue. In addition, metabolic, inflammatory and angiogenic parameters were carried out in those mice. KEY FINDINGS This study provides evidence of the ability of a biohybrid device to induce the growth of differentiated pancreas parenchyma in both normoglycemic and streptozotocin-induced diabetic mice as detected by histological analysis. Glucose metabolism and body weight of hyperglycemic mice bearing hASCs implants improved. SIGNIFICANCE The synthetic porous scaffold bearing hASC cells placed adjacent to the native animal pancreas exhibits the potential to be exploited in future cell-based type 1 diabetes therapies.
Collapse
Affiliation(s)
- Luciana Xavier Pereira
- Department of General Pathology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Celso Tarso Rodrigues Viana
- Department of General Pathology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Laura Alejandra Ariza Orellano
- Department of General Pathology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Simone Aparecida Almeida
- Department of General Pathology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Anilton Cesar Vasconcelos
- Department of General Pathology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Alexander Birbrair
- Department of General Pathology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Silvia Passos Andrade
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Paula Peixoto Campos
- Department of General Pathology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
16
|
Alsahli M, Gerich JE. Hypoglycemia in Diabetes Mellitus. PRINCIPLES OF DIABETES MELLITUS 2017:367-383. [DOI: 10.1007/978-3-319-18741-9_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
17
|
Yamashita-Sugahara Y, Matsumoto M, Ohtaka M, Nishimura K, Nakanishi M, Mitani K, Okazaki Y. An inhibitor of fibroblast growth factor receptor-1 (FGFR1) promotes late-stage terminal differentiation from NGN3+ pancreatic endocrine progenitors. Sci Rep 2016; 6:35908. [PMID: 27786288 PMCID: PMC5081516 DOI: 10.1038/srep35908] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 10/07/2016] [Indexed: 12/31/2022] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) provide a potential resource for regenerative medicine. To identify the signalling pathway(s) contributing to the development of functional β cells, we established a tracing model consisting of dual knock-in hiPSCs (INS-Venus/NGN3-mCherry) (hIveNry) expressing the fluorescent proteins Venus and mCherry under the control of intrinsic insulin (INS) and neurogenin 3 (NGN3) promoters, respectively. hIveNry iPSCs differentiated into NGN3- and mCherry-positive endocrine progenitors and then into Venus-positive β cells expressing INS, PDX1, NKX6.1, and glucokinase (GCK). Using these cells, we conducted high-throughput screening of chemicals and identified a specific kinase inhibitor of fibroblast growth factor receptor 1 (FGFR1) that acted in a stage-dependent manner to promote the terminal differentiation of pancreatic endocrine cells, including β cells, from the intermediate stage of pancreatic endocrine progenitors while blocking the early development of pancreatic progenitors. This FGFR1 inhibitor augmented the expression of functional β cell markers (SLC30A8 and ABCC8) and improved glucose-stimulated INS secretion. Our findings indicate that the hIveNry model could provide further insights into the mechanisms of hiPS-derived β cell differentiation controlled by FGFR1-mediated regulatory pathways in a temporal-dependent fashion.
Collapse
Affiliation(s)
- Yzumi Yamashita-Sugahara
- Division of Functional Genomics and Systems Medicine, Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan
| | - Masahito Matsumoto
- Division of Functional Genomics and Systems Medicine, Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan
| | - Manami Ohtaka
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Ibaraki, Japan
| | - Ken Nishimura
- Laboratory of Gene Regulation, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Mahito Nakanishi
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology (AIST), Ibaraki, Japan
| | - Kohnosuke Mitani
- Division of Gene Therapy, Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan
| | - Yasushi Okazaki
- Division of Functional Genomics and Systems Medicine, Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan
| |
Collapse
|
18
|
Robertson RP. Spontaneous Hypoglycemia After Islet Transplantation: The Case For Using Non-Hepatic Sites. J Clin Endocrinol Metab 2016; 101:3571-3574. [PMID: 27610653 PMCID: PMC5052339 DOI: 10.1210/jc.2016-2850] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
This Perspective provides a brief history of intrahepatic alloislet and autoislet transplantation in humans and an update of the recent success rates. It also examines the important role that hypoglycemia plays in clinical outcomes. On the one hand, recurrent serious hypoglycemic episodes related to insulin therapy are a major criterion for alloislet transplantation. On the other hand, spontaneous clinical hypoglycemia, perhaps related to the accompanying Roux-en-Y procedure for total pancreatectomy, is a complication of autoislet transplantation. Complex alterations in glucagon secretion compromise counter-regulation of hypoglycemia in both situations. The glucagon response to hypoglycemia is intrinsically defective in type 1 diabetes before transplant because of the absence of physiological regulation of α-cell secretion by neighboring β-cells. Glucagon secretion from intrahepatic islets during systemic hypoglycemia is also defective, although β-cells in the graft are normally regulated by glucose and arginine. My personal perspective is that the latter is caused by intrahepatic glycogenolysis stimulated by systemic hypoglycemia with consequent increases in intrahepatic glucose flux, which incorrectly signals intrahepatic α-cells to be quiescent. This defect is liver-specific, which strongly suggests modifying the current approach to islet transplantation by placing a portion of allo- and autoislets in nonhepatic sites in addition to hepatic sites to ensure physiological glucagon secretion as a strategy to ameliorate post-transplant hypoglycemia.
Collapse
Affiliation(s)
- R Paul Robertson
- Pacific NW Diabetes Research Institute, Divisions of Endocrinology and Metabolism, Universities of Washington and Minnesota, Seattle, Washington 98122
| |
Collapse
|
19
|
Demeester S, Balke EM, Van der Auwera BJ, Gillard P, Hilbrands R, Lee D, Van de Velde U, Ling Z, Roep BO, Pipeleers DG, Gorus FK, Keymeulen B. HLA-A*24 Carrier Status and Autoantibody Surges Posttransplantation Associate With Poor Functional Outcome in Recipients of an Islet Allograft. Diabetes Care 2016; 39:1060-4. [PMID: 27208324 DOI: 10.2337/dc15-2768] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 03/28/2016] [Indexed: 02/03/2023]
Abstract
OBJECTIVE We investigated whether changes in islet autoantibody profile and presence of HLA risk markers, reported to predict rapid β-cell loss in pre-type 1 diabetes, associate with poor functional outcome in islet allograft recipients. RESEARCH DESIGN AND METHODS Forty-one patients received ≥2.3 million β-cells/kg body wt in one to two intraportal implantations. Outcome after 6-18 months was assessed by C-peptide (random and stimulated), insulin dose, and HbA1c. RESULTS Patients carrying HLA-A*24-positive or experiencing a significant autoantibody surge within 6 months after the first transplantation (n = 19) had lower C-peptide levels (P ≤ 0.003) and higher insulin needs (P < 0.001) despite higher HbA1c levels (P ≤ 0.018). They became less often insulin independent (16% vs. 68%, P = 0.002) and remained less often C-peptide positive (47% vs. 100%, P < 0.001) than recipients lacking both risk factors. HLA-A*24 positivity or an autoantibody surge predicted insulin dependence (P = 0.007). CONCLUSIONS HLA-A*24 and early autoantibody surge after islet implantation associate with poor functional graft outcome.
Collapse
Affiliation(s)
- Simke Demeester
- Diabetes Research Center, Brussels Free University, Brussels, Belgium
| | - Else M Balke
- Diabetes Research Center, Brussels Free University, Brussels, Belgium
| | | | - Pieter Gillard
- Diabetes Research Center, Brussels Free University, Brussels, Belgium Department of Endocrinology, University Hospitals Leuven, Leuven, Belgium
| | - Robert Hilbrands
- Diabetes Research Center, Brussels Free University, Brussels, Belgium
| | - DaHae Lee
- Diabetes Research Center, Brussels Free University, Brussels, Belgium Department of Endocrinology, University Hospitals Leuven, Leuven, Belgium
| | | | - Zhidong Ling
- Diabetes Research Center, Brussels Free University, Brussels, Belgium
| | - Bart O Roep
- Department of Immunohaematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Frans K Gorus
- Diabetes Research Center, Brussels Free University, Brussels, Belgium
| | - Bart Keymeulen
- Diabetes Research Center, Brussels Free University, Brussels, Belgium
| |
Collapse
|
20
|
Forlenza GP, Nathan BM, Moran AM, Dunn TB, Beilman GJ, Pruett TL, Bellin MD. Successful Application of Closed-Loop Artificial Pancreas Therapy After Islet Autotransplantation. Am J Transplant 2016; 16:527-34. [PMID: 26588810 PMCID: PMC4844547 DOI: 10.1111/ajt.13539] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 09/13/2015] [Accepted: 09/18/2015] [Indexed: 01/25/2023]
Abstract
Total pancreatectomy with islet autotransplantation (TPIAT) may relieve the pain of chronic pancreatitis while avoiding postsurgical diabetes. Minimizing hyperglycemia after TPIAT limits beta cell apoptosis during islet engraftment. Closed-loop (CL) therapy combining an insulin pump with a continuous glucose monitor (CGM) has not been investigated previously in islet transplant recipients. Our objective was to determine the feasibility and efficacy of CL therapy to maintain glucose profiles close to normoglycemia following TPIAT. Fourteen adult subjects (36% male; aged 35.9 ± 11.4 years) were randomized to subcutaneous insulin via CL pump (n = 7) or multiple daily injections with blinded CGM (n = 7) for 72 h at transition from intravenous to subcutaneous insulin. Mean serum glucose values were significantly lower in the CL pump group than in the control group (111 ± 4 vs. 130 ± 13 mg/dL; p = 0.003) without increased risk of hypoglycemia (percentage of time <70 mg/dL: CL pump 1.9%, control 4.8%; p = 0.46). Results from this pilot study suggest that CL therapy is superior to conventional therapy in maintaining euglycemia without increased hypoglycemia. This technology shows significant promise to safely maintain euglycemic targets during the period of islet engraftment following islet transplantation.
Collapse
Affiliation(s)
- Gregory P. Forlenza
- Department of Pediatrics, University of Minnesota Medical Center, Minneapolis, MN, 55454, United States,Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Denver, CO, 80045, United States
| | - Brandon M. Nathan
- Department of Pediatrics, University of Minnesota Medical Center, Minneapolis, MN, 55454, United States
| | - Antoinette M. Moran
- Department of Pediatrics, University of Minnesota Medical Center, Minneapolis, MN, 55454, United States
| | - Ty B. Dunn
- Department of Surgery, University of Minnesota Medical Center, Minneapolis, MN, 55454, United States
| | - Gregory J. Beilman
- Department of Surgery, University of Minnesota Medical Center, Minneapolis, MN, 55454, United States
| | - Timothy L. Pruett
- Department of Surgery, University of Minnesota Medical Center, Minneapolis, MN, 55454, United States
| | - Melena D. Bellin
- Department of Pediatrics, University of Minnesota Medical Center, Minneapolis, MN, 55454, United States
| |
Collapse
|
21
|
Hawthorne WJ. Necessities for a Clinical Islet Program. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 938:67-88. [PMID: 27586423 DOI: 10.1007/978-3-319-39824-2_6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
For more than two decades we have been refining advances in islet cell transplantation as a clinical therapy for patients suffering from type 1 diabetes. A great deal of effort has gone to making this a viable therapy for a broader range of patients with type 1 diabetes. Clinical results have progressively improved, demonstrating clinical outcomes on par with other organ transplants, specifically in terms of insulin independence, graft and patient survival. We are now at the point where islet cell transplantation, in the form of allotransplantation, has become accepted as a clinical therapy in adult patients affected by type 1 diabetes, in particular those suffering from severe hypoglycaemic unawareness. This chapter provides an overview on how this has been undertaken over the years to provide outcomes on par with other organ transplantation results. In particular this chapter focuses on the processes and facilities that are required to establish a clinical islet isolation and transplantation program. It also outlines the very important underpinning processes of selection of the organ donor for islet isolation, the processes of organ donor operation and preservation of the pancreas by various means and the ideal ways to best improve outcomes for human islet cell isolation. Providing these more optimal conditions we can underpin the isolation processes to provide islets for transplantation and as such a safe, effective and feasible therapeutic option for an increasing number of patients suffering from type 1 diabetes with severe hypoglycaemic unawareness.
Collapse
Affiliation(s)
- Wayne J Hawthorne
- National Pancreas and Islet Transplant Laboratories, The Westmead Institute for Medical Research, Westmead, NSW, 2145, Australia. .,Department of Surgery, Westmead Clinical School, Westmead Hospital, University of Sydney, Westmead, NSW, 2145, Australia.
| |
Collapse
|
22
|
Päth G, Seufert J. Stammzelltherapie des Diabetes mellitus Typ 1. DIABETOLOGE 2015. [DOI: 10.1007/s11428-015-0024-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
23
|
Ludwig B, Ludwig S. Transplantable bioartificial pancreas devices: current status and future prospects. Langenbecks Arch Surg 2015; 400:531-40. [DOI: 10.1007/s00423-015-1314-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 06/04/2015] [Indexed: 02/08/2023]
|
24
|
Choudhary P, Rickels MR, Senior PA, Vantyghem MC, Maffi P, Kay TW, Keymeulen B, Inagaki N, Saudek F, Lehmann R, Hering BJ. Evidence-informed clinical practice recommendations for treatment of type 1 diabetes complicated by problematic hypoglycemia. Diabetes Care 2015; 38:1016-29. [PMID: 25998294 PMCID: PMC4439532 DOI: 10.2337/dc15-0090] [Citation(s) in RCA: 150] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Problematic hypoglycemia, defined as two or more episodes per year of severe hypoglycemia or as one episode associated with impaired awareness of hypoglycemia, extreme glycemic lability, or major fear and maladaptive behavior, is a challenge, especially for patients with long-standing type 1 diabetes. Individualized therapy for such patients should include a composite target: optimal glucose control without problematic hypoglycemia. Therefore, we propose a tiered, four-stage algorithm based on evidence of efficacy given the limitations of educational, technological, and transplant interventions. All patients with problematic hypoglycemia should undergo structured or hypoglycemia-specific education programs (stage 1). Glycemic and hypoglycemia treatment targets should be individualized and reassessed every 3-6 months. If targets are not met, one diabetes technology-continuous subcutaneous insulin infusion or continuous glucose monitoring-should be added (stage 2). For patients with continued problematic hypoglycemia despite education (stage 1) and one diabetes technology (stage 2), sensor-augmented insulin pumps preferably with an automated low-glucose suspend feature and/or very frequent contact with a specialized hypoglycemia service can reduce hypoglycemia (stage 3). For patients whose problematic hypoglycemia persists, islet or pancreas transplant should be considered (stage 4). This algorithm provides an evidence-informed approach to resolving problematic hypoglycemia; it should be used as a guide, with individual patient circumstances directing suitability and acceptability to ensure the prudent use of technology and scarce transplant resources. Standardized reporting of hypoglycemia outcomes and inclusion of patients with problematic hypoglycemia in studies of new interventions may help to guide future therapeutic strategies.
Collapse
Affiliation(s)
| | - Michael R Rickels
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Peter A Senior
- Department of Medicine, Division of Endocrinology, University of Alberta, Edmonton, Canada
| | - Marie-Christine Vantyghem
- Endocrinology and Metabolism Department, INSERM U1190, European Genomics Institute for Diabetes, Lille University Hospital, Lille Cedex, France
| | - Paola Maffi
- Diabetes Research Institute, Scientific Institute Ospedale San Raffaele, Milan, Italy
| | - Thomas W Kay
- Immunology and Diabetes Unit, St. Vincent's Institute, University of Melbourne, Melbourne, Australia
| | - Bart Keymeulen
- Diabetes Clinic and Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Nobuya Inagaki
- Department of Diabetes and Clinical Nutrition, Kyoto University, Kyoto, Japan
| | - Frantisek Saudek
- Diabetes Center, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Roger Lehmann
- Department of Endocrinology and Diabetology, University of Zurich, Zurich, Switzerland
| | - Bernhard J Hering
- Schulze Diabetes Institute and Department of Surgery, University of Minnesota, Minneapolis, MN
| |
Collapse
|
25
|
Affiliation(s)
- William T Cefalu
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA
| | | | - Jay S Skyler
- Division of Endocrinology, University of Miami, Miami, FL
| |
Collapse
|