1
|
Niu F, Liu W, Ren Y, Tian Y, Shi W, Li M, Li Y, Xiong Y, Qian L. β-cell neogenesis: A rising star to rescue diabetes mellitus. J Adv Res 2024; 62:71-89. [PMID: 37839502 PMCID: PMC11331176 DOI: 10.1016/j.jare.2023.10.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 10/08/2023] [Accepted: 10/08/2023] [Indexed: 10/17/2023] Open
Abstract
BACKGROUND Diabetes Mellitus (DM), a chronic metabolic disease characterized by elevated blood glucose, is caused by various degrees of insulin resistance and dysfunctional insulin secretion, resulting in hyperglycemia. The loss and failure of functional β-cells are key mechanisms resulting in type 1 diabetes mellitus (T1DM) and type 2 diabetes mellitus (T2DM). AIM OF REVIEW Elucidating the underlying mechanisms of β-cell failure, and exploring approaches for β-cell neogenesis to reverse β-cell dysfunction may provide novel strategies for DM therapy. KEY SCIENTIFIC CONCEPTS OF REVIEW Emerging studies reveal that genetic susceptibility, endoplasmic reticulum (ER) stress, oxidative stress, islet inflammation, and protein modification linked to multiple signaling pathways contribute to DM pathogenesis. Over the past few years, replenishing functional β-cell by β-cell neogenesis to restore the number and function of pancreatic β-cells has remarkably exhibited a promising therapeutic approach for DM therapy. In this review, we provide a comprehensive overview of the underlying mechanisms of β-cell failure in DM, highlight the effective approaches for β-cell neogenesis, as well as discuss the current clinical and preclinical agents research advances of β-cell neogenesis. Insights into the challenges of translating β-cell neogenesis into clinical application for DM treatment are also offered.
Collapse
Affiliation(s)
- Fanglin Niu
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, the Affiliated Hospital of Northwest University, Xi'an No.3 Hospital, Xi'an, Shaanxi, PR China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Wenxuan Liu
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, the Affiliated Hospital of Northwest University, Xi'an No.3 Hospital, Xi'an, Shaanxi, PR China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Yuanyuan Ren
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, the Affiliated Hospital of Northwest University, Xi'an No.3 Hospital, Xi'an, Shaanxi, PR China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Ye Tian
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, the Affiliated Hospital of Northwest University, Xi'an No.3 Hospital, Xi'an, Shaanxi, PR China; Department of Neurology, Affiliated Hospital of Northwest University, Xi'an No.3 Hospital, Xi'an, Shaanxi, China
| | - Wenzhen Shi
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, the Affiliated Hospital of Northwest University, Xi'an No.3 Hospital, Xi'an, Shaanxi, PR China; Medical Research Center, the affiliated Hospital of Northwest University, Xi'an No.3 Hospital, Xi'an, Shaanxi, China
| | - Man Li
- Department of Endocrinology, the Affiliated Hospital of Northwest University, Xi'an No.3 Hospital, Xi'an, Shaanxi, China
| | - Yujia Li
- Department of Endocrinology, the Affiliated Hospital of Northwest University, Xi'an No.3 Hospital, Xi'an, Shaanxi, China
| | - Yuyan Xiong
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, the Affiliated Hospital of Northwest University, Xi'an No.3 Hospital, Xi'an, Shaanxi, PR China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Lu Qian
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, the Affiliated Hospital of Northwest University, Xi'an No.3 Hospital, Xi'an, Shaanxi, PR China; Department of Endocrinology, the Affiliated Hospital of Northwest University, Xi'an No.3 Hospital, Xi'an, Shaanxi, China
| |
Collapse
|
2
|
Kim MH, Thanuthanakhun N, Kino-Oka M. A simple tool for the synchronous differentiation of human induced pluripotent stem cells into pancreatic progenitors. Biotechnol J 2024; 19:e2300364. [PMID: 37955342 DOI: 10.1002/biot.202300364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 10/01/2023] [Accepted: 11/09/2023] [Indexed: 11/14/2023]
Abstract
Efficient differentiation of human induced pluripotent stem cells (hiPSCs) into functional pancreatic cells holds great promise for diabetes research and treatment. However, a robust culture strategy for producing pancreatic progenitors with high homogeneity is lacking. Here, we established a simple differentiation strategy for generating synchronous iPSC-derived pancreatic progenitors via a two-step method of sequential cell synchronization using botulinum hemagglutinin (HA), an E-cadherin function-blocking agent. Of the various methods tested, the first-step synchronization method with HA exposure induces a synchronous switch from E- to N-cadherin and N- to E-cadherin expression by spatially controlling heterogeneous cell distribution, subsequently improving their competency for directed differentiation into definitive endodermal cells from iPSCs. The iPSC-derived definitive endodermal cells can efficiently generate PDX1+ and NKX6.1+ pancreatic progenitor cells in high yields. The PDX1+ and PDX1+ /NKX6.1+ cell densities showed 1.6- and 2.2-fold increases, respectively, compared with those from unsynchronized cultures. The intra-run and inter-run coefficient of variation were below 10%, indicating stable and robust differentiation across different cultures and runs. Our approach is a simple and efficient strategy to produce large quantities of differentiated cells with the highest homogeneity during multistage pancreatic progenitor differentiation, providing a potential tool for guided differentiation of iPSCs to functional insulin-producing cells.
Collapse
Affiliation(s)
- Mee-Hae Kim
- Department of Biotechnology, Graduate School of Engineering, Osaka University, Suita, Osaka, Japan
| | - Naruchit Thanuthanakhun
- Department of Biotechnology, Graduate School of Engineering, Osaka University, Suita, Osaka, Japan
| | - Masahiro Kino-Oka
- Department of Biotechnology, Graduate School of Engineering, Osaka University, Suita, Osaka, Japan
- Research Base for Cell Manufacturability, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
3
|
Kuo YC, Lin SY, De S, Rajesh R. Regeneration of Pancreatic Cells Using Optimized Nanoparticles and l-Glutamic Acid-Gelatin Scaffolds with Controlled Topography and Grafted Activin A/BMP4. ACS Biomater Sci Eng 2023; 9:6208-6224. [PMID: 37882705 DOI: 10.1021/acsbiomaterials.3c00791] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2023]
Abstract
Regeneration of insulin-producing cells (IPCs) from induced pluripotent stem cells (iPSCs) under controlled conditions has a lot of promise to emulate the pancreatic mechanism in vivo as a foundation of cell-based diabetic therapy. l-Glutamic acid-gelatin scaffolds with orderly pore sizes of 160 and 200 μm were grafted with activin A and bone morphogenic proteins 4 (BMP4) to differentiate iPSCs into definitive endoderm (DE) cells, which were then guided with fibroblast growth factor 7 (FGF7)-grafted retinoic acid (RA)-loaded solid lipid nanoparticles (FR-SLNs) to harvest IPCs. Response surface methodology was adopted to optimize the l-glutamic acid-to-gelatin ratio of scaffolds and to optimize surfactant concentration and lipid proportion in FR-SLNs. Experimental results of immunofluorescence, flow cytometry, and western blots revealed that activin A (100 ng/mL)-BMP4 (50 ng/mL)-l-glutamic acid (5%)-gelatin (95%) scaffolds provoked the largest number of SOX17-positive DE cells from iPSCs. Treatment with FGF7 (50 ng/mL)-RA (600 ng/mL)-SLNs elicited the highest number of PDX1-positive β-cells from differentiated DE cells. To imitate the natural pancreas, the scaffolds with controlled topography were appropriate for IPC production with sufficient insulin secretion. Hence, the current scheme using FR-SLNs and activin A-BMP4-l-glutamic acid-gelatin scaffolds in the two-stage differentiation of iPSCs can be promising for replacing impaired β-cells in diabetic management.
Collapse
Affiliation(s)
- Yung-Chih Kuo
- Department of Chemical Engineering, National Chung Cheng University, Chia-Yi, Taiwan 62102, ROC
- Advanced Institute of Manufacturing with High-tech Innovations, National Chung Cheng University, Chia-Yi, Taiwan 62102, ROC
| | - Sheng-Yuan Lin
- Department of Chemical Engineering, National Chung Cheng University, Chia-Yi, Taiwan 62102, ROC
| | - Sourav De
- Department of Chemical Engineering, National Chung Cheng University, Chia-Yi, Taiwan 62102, ROC
| | - Rajendiran Rajesh
- Department of Chemical Engineering, National Chung Cheng University, Chia-Yi, Taiwan 62102, ROC
| |
Collapse
|
4
|
Braam MJS, Zhao J, Liang S, Ida S, Kloostra NK, Iworima DG, Tang M, Baker RK, Quiskamp N, Piret JM, Kieffer TJ. Protocol development to further differentiate and transition stem cell-derived pancreatic progenitors from a monolayer into endocrine cells in suspension culture. Sci Rep 2023; 13:8877. [PMID: 37264038 DOI: 10.1038/s41598-023-35716-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 05/23/2023] [Indexed: 06/03/2023] Open
Abstract
The generation of functional β-cells from human pluripotent stem cells (hPSCs) for cell replacement therapy and disease modeling of diabetes is being investigated by many groups. We have developed a protocol to harvest and aggregate hPSC-derived pancreatic progenitors generated using a commercially available kit into near uniform spheroids and to further differentiate the cells toward an endocrine cell fate in suspension culture. Using a static suspension culture platform, we could generate a high percentage of insulin-expressing, glucose-responsive cells. We identified FGF7 as a soluble factor promoting aggregate survival with no inhibitory effect on endocrine gene expression. Notch inhibition of pancreatic progenitor cells during aggregation improved endocrine cell induction in vitro and improved graft function following implantation and further differentiation in mice. Thus we provide an approach to promote endocrine formation from kit-derived pancreatic progenitors, either through extended culture or post implant.
Collapse
Affiliation(s)
- Mitchell J S Braam
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Jia Zhao
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Shenghui Liang
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Shogo Ida
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Nick K Kloostra
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Diepiriye G Iworima
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Mei Tang
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Robert K Baker
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | | | - James M Piret
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Department of Chemical and Biological Engineering, University of British Columbia, Vancouver, BC, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Timothy J Kieffer
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, Canada.
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada.
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
5
|
Stem Cell-Derived Islets for Type 2 Diabetes. Int J Mol Sci 2022; 23:ijms23095099. [PMID: 35563490 PMCID: PMC9105352 DOI: 10.3390/ijms23095099] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/23/2022] [Accepted: 04/28/2022] [Indexed: 02/04/2023] Open
Abstract
Since the discovery of insulin a century ago, insulin injection has been a primary treatment for both type 1 (T1D) and type 2 diabetes (T2D). T2D is a complicated disea se that is triggered by the dysfunction of insulin-producing β cells and insulin resistance in peripheral tissues. Insulin injection partially compensates for the role of endogenous insulin which promotes glucose uptake, lipid synthesis and organ growth. However, lacking the continuous, rapid, and accurate glucose regulation by endogenous functional β cells, the current insulin injection therapy is unable to treat the root causes of the disease. Thus, new technologies such as human pluripotent stem cell (hPSC)-derived islets are needed for both identifying the key molecular and genetic causes of T2D and for achieving a long-term treatment. This perspective review will provide insight into the efficacy of hPSC-derived human islets for treating and understanding T2D. We discuss the evidence that β cells should be the primary target for T2D treatment, the use of stem cells for the modeling of T2D and the potential use of hPSC-derived islet transplantation for treating T2D.
Collapse
|
6
|
Gao D, Dai P, Fan Z, Wang J, Zhang Y. The Roles of Different Multigene Combinations of Pdx1, Ngn3, Sox9, Pax4, and Nkx2.2 in the Reprogramming of Canine ADSCs Into IPCs. Cell Transplant 2022; 31:9636897221081483. [PMID: 35236160 PMCID: PMC8902191 DOI: 10.1177/09636897221081483] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Adipose-derived mesenchymal stem cells (ADSCs) are ideal sources for the treatment of diabetes, and the differentiation of ADSCs into insulin-producing cells (IPCs) through transfection of exogenous regulatory genes in vitro has been studied in depth. The differentiation of ADSCs is strictly regulated by a variety of transcription factors such as Pdx1, Ngn3, Pax4, Nkx2.2, and Sox9. However, whether these genes can coordinately regulate the differentiation of ADSCs into IPCs is still unknown. In this study, five multigene coexpressing adenovirus vectors (pAdTrack-Pdx1-Ngn3-AdEasy, pAdTrack-Pdx1-Ngn3-Sox9-AdEasy, pAdTrack-Pdx1-Ngn3-Pax4-Sox9-AdEasy, pAdTrack-Pdx1-Ngn3-Nkx2.2-Sox9-AdEasy, and pAdTrack-Pdx1-Ngn3-Nkx2.2-Pax4-AdEasy) were constructed, and then the stocks of the packaged adenoviruses were used to infect the canine ADSCs (cADSCs). Based on results of morphological observation, dithizone staining, sugar-stimulated insulin secretion test, cellular insulin immunofluorescence assays, and the detection of pancreatic β-cell development-related genes in the induced cells, the best induction combination (pAdTrack-Pdx1-Ngn3-Nkx2.2-Pax4-AdEasy) was identified after comparative screening. This study provides a theoretical reference and an experimental basis for further research on stem cell replacement therapy for diabetes.
Collapse
Affiliation(s)
- Dengke Gao
- College of Veterinary Medicine, Northwest A&F University, Xianyang, China
| | - Pengxiu Dai
- College of Veterinary Medicine, Northwest A&F University, Xianyang, China
| | - Zhixin Fan
- College of Veterinary Medicine, Northwest A&F University, Xianyang, China
| | - Jinglu Wang
- College of Veterinary Medicine, Northwest A&F University, Xianyang, China
| | - Yihua Zhang
- College of Veterinary Medicine, Northwest A&F University, Xianyang, China
| |
Collapse
|
7
|
iPSCs-laden GDF8-grafted aldehyde hyaluronic acid-polyacrylamide inverted colloidal crystal constructs with controlled release of CHIR99021 and retinoic acid to generate insulin-producing cells. J Taiwan Inst Chem Eng 2020. [DOI: 10.1016/j.jtice.2020.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
8
|
In Vitro Generation of Glucose-Responsive Insulin-Secreting Cells from PDX1-Overexpressing Human-Induced Pluripotent Stem Cell Derived from Diabetic Patient. ASAIO J 2019; 64:819-826. [PMID: 29210770 DOI: 10.1097/mat.0000000000000728] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Pancreatic and duodenal homeobox 1 (PDX1), a member of the homeodomain-containing transcription factor family, is a key transcription factor for pancreas development and mature β-cell function. In this study, induced overexpression of PDX1 resulted in producing susceptible cells for pancreatic differentiation and was well beneficial to enhance β-cell production, maturation, function, and survival. Induced PDX1 overexpression in harmony with a set of signaling molecules involves in guiding the signaling pathways toward pancreas development, leaded to high-efficient in vitro generation of ectopic insulin-producing cells (IPCs) with the effectively reduced number of polyhormonal cells and increased number of insulin (INS) single-positive cells. This strategy yielded 85.61% glucose-responsive insulin-positive cells in vitro, which was seven times higher than the basal level, and electron microscopy images revealed the presence of mature β-cell secretory granules. The generation of glucose-responsive insulin-secreting β-like cells from human-induced pluripotent stem cells (hiPSCs) in vitro would provide a promising approach to produce an unprecedented cell source for cell transplantation therapy in diabetes without the ethical obstacle of embryonic stem cells and would bypass immune rejection. These cells are an invaluable source for disease modeling, drug discovery, and pharmacogenomics studies as well.
Collapse
|
9
|
Balboa D, Saarimäki-Vire J, Otonkoski T. Concise Review: Human Pluripotent Stem Cells for the Modeling of Pancreatic β-Cell Pathology. Stem Cells 2018; 37:33-41. [PMID: 30270471 PMCID: PMC7379656 DOI: 10.1002/stem.2913] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 08/21/2018] [Accepted: 08/28/2018] [Indexed: 12/11/2022]
Abstract
Pancreatic β‐cells are the only source of insulin. Disturbances in β‐cell development or function may thus result in insulin deficiency or excess, presenting as hyper‐ or hypoglycemia. It is increasingly evident that common forms of diabetes (types 1 and 2) are pathogenically heterogeneous. Development of efficient therapies is dependent on reliable disease models. Although animal models are remarkably useful research tools, they present limitations because of species differences. As an alternative, human pluripotent stem cell technologies offer multiple possibilities for the study of human diseases in vitro. In the last decade, advances in the derivation of induced pluripotent stem cells from diabetic patients, combined with β‐cell differentiation protocols, have resulted in the generation of useful disease models for diabetes. First disease models have been focusing on monogenic diabetes. The development of genome editing technologies, more advanced differentiation protocols and humanized mouse models based on transplanted cells have opened new horizons for the modeling of more complex forms of β‐cell dysfunction. We present here the incremental progress made in the modeling of diabetes using pluripotent stem cells. We discuss the current challenges and opportunities of these approaches to dissect β‐cell pathology and devise new pharmacological and cell replacement therapies. stem cells2019;37:33–41
Collapse
Affiliation(s)
- Diego Balboa
- Research Programs Unit, Molecular Neurology, Biomedicum Stem Cell Centre, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Jonna Saarimäki-Vire
- Research Programs Unit, Molecular Neurology, Biomedicum Stem Cell Centre, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Timo Otonkoski
- Research Programs Unit, Molecular Neurology, Biomedicum Stem Cell Centre, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Children's Hospital, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| |
Collapse
|
10
|
Lloyd RV, McNicoll AM. Annual review issue: An overview of 50 years of progress in endocrine pathology. Histopathology 2018; 72:4-5. [PMID: 29239043 DOI: 10.1111/his.13289] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Ricardo V Lloyd
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Anne-Marie McNicoll
- Molecular and Cellular Pathology, The University of Queensland, Brisbane, Australia
| |
Collapse
|
11
|
Abstract
PURPOSE OF REVIEW The immunosuppressive agent cyclosporine was first reported to lower daily insulin dose and improve glycemic control in patients with new-onset type 1 diabetes (T1D) in 1984. While renal toxicity limited cyclosporine's extended use, this observation ignited collaborative efforts to identify immunotherapeutic agents capable of safely preserving β cells in patients with or at risk for T1D. RECENT FINDINGS Advances in T1D prediction and early diagnosis, together with expanded knowledge of the disease mechanisms, have facilitated trials targeting specific immune cell subsets, autoantigens, and pathways. In addition, clinical responder and non-responder subsets have been defined through the use of metabolic and immunological readouts. Herein, we review emerging T1D biomarkers within the context of recent and ongoing T1D immunotherapy trials. We also discuss responder/non-responder analyses in an effort to identify therapeutic mechanisms, define actionable pathways, and guide subject selection, drug dosing, and tailored combination drug therapy for future T1D trials.
Collapse
Affiliation(s)
- Laura M Jacobsen
- Department of Pediatrics, College of Medicine, University of Florida Diabetes Institute, Gainesville, FL, USA
| | - Brittney N Newby
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida Diabetes Institute, 1275 Center Drive, Biomedical Sciences Building J-589, Box 100275, Gainesville, FL, 32610, USA
| | - Daniel J Perry
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida Diabetes Institute, 1275 Center Drive, Biomedical Sciences Building J-589, Box 100275, Gainesville, FL, 32610, USA
| | - Amanda L Posgai
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida Diabetes Institute, 1275 Center Drive, Biomedical Sciences Building J-589, Box 100275, Gainesville, FL, 32610, USA
| | - Michael J Haller
- Department of Pediatrics, College of Medicine, University of Florida Diabetes Institute, Gainesville, FL, USA
| | - Todd M Brusko
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida Diabetes Institute, 1275 Center Drive, Biomedical Sciences Building J-589, Box 100275, Gainesville, FL, 32610, USA.
| |
Collapse
|
12
|
Little D, Luft C, Mosaku O, Lorvellec M, Yao Z, Paillusson S, Kriston-Vizi J, Gandhi S, Abramov AY, Ketteler R, Devine MJ, Gissen P. A single cell high content assay detects mitochondrial dysfunction in iPSC-derived neurons with mutations in SNCA. Sci Rep 2018; 8:9033. [PMID: 29899557 PMCID: PMC5998042 DOI: 10.1038/s41598-018-27058-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 05/25/2018] [Indexed: 12/21/2022] Open
Abstract
Mitochondrial dysfunction is implicated in many neurodegenerative diseases including Parkinson's disease (PD). Induced pluripotent stem cells (iPSCs) provide a unique cell model for studying neurological diseases. We have established a high-content assay that can simultaneously measure mitochondrial function, morphology and cell viability in iPSC-derived dopaminergic neurons. iPSCs from PD patients with mutations in SNCA and unaffected controls were differentiated into dopaminergic neurons, seeded in 384-well plates and stained with the mitochondrial membrane potential dependent dye TMRM, alongside Hoechst-33342 and Calcein-AM. Images were acquired using an automated confocal screening microscope and single cells were analysed using automated image analysis software. PD neurons displayed reduced mitochondrial membrane potential and altered mitochondrial morphology compared to control neurons. This assay demonstrates that high content screening techniques can be applied to the analysis of mitochondria in iPSC-derived neurons. This technique could form part of a drug discovery platform to test potential new therapeutics for PD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Daniel Little
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London, United Kingdom.
| | - Christin Luft
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London, United Kingdom
| | - Olukunbi Mosaku
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London, United Kingdom
| | - Maëlle Lorvellec
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London, United Kingdom
| | - Zhi Yao
- Sobell Department of Motor Neuroscience and Movement Disorders, UCL Institute of Neurology, Queen Square, London, United Kingdom
- The Francis Crick Institute, 1 Midland Road, King's Cross, London, United Kingdom
| | - Sébastien Paillusson
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London, United Kingdom
| | - Janos Kriston-Vizi
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London, United Kingdom
| | - Sonia Gandhi
- Sobell Department of Motor Neuroscience and Movement Disorders, UCL Institute of Neurology, Queen Square, London, United Kingdom
- The Francis Crick Institute, 1 Midland Road, King's Cross, London, United Kingdom
| | - Andrey Y Abramov
- Department of Molecular Neuroscience, University College London, Institute of Neurology, Queen Square, London, United Kingdom
| | - Robin Ketteler
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London, United Kingdom
| | - Michael J Devine
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London, United Kingdom.
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, United Kingdom.
| | - Paul Gissen
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London, United Kingdom
| |
Collapse
|
13
|
Huang Y, Wan J, Guo Y, Zhu S, Wang Y, Wang L, Guo Q, Lu Y, Wang Z. Transcriptome Analysis of Induced Pluripotent Stem Cell (iPSC)-derived Pancreatic β-like Cell Differentiation. Cell Transplant 2018; 26:1380-1391. [PMID: 28901190 PMCID: PMC5680972 DOI: 10.1177/0963689717720281] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Diabetes affects millions of people worldwide, and β-cell replacement is one of the promising new strategies for treatment. Induced pluripotent stem cells (iPSCs) can differentiate into any cell type, including pancreatic β cells, providing a potential treatment for diabetes. However, the molecular mechanisms underlying the differentiation of iPSC-derived β cells have not yet been fully elucidated. Here, we generated pancreatic β-like cells from mouse iPSCs using a 3-step protocol and performed deep RNA sequencing to get a transcriptional landscape of iPSC-derived pancreatic β-like cells during the selective differentiation period. We then focused on the differentially expressed genes (DEGs) during the time course of the differentiation period, and these genes underwent Gene Ontology annotation and Kyoto Encyclopedia of Genes and Genomes pathway analysis. In addition, gene-act networks were constructed for these DEGs, and the expression of pivotal genes detected by quantitative real-time polymerase chain reaction was well correlated with RNA sequence (RNA-seq). Overall, our study provides valuable information regarding the transcriptome changes in β cells derived from iPSCs during differentiation, elucidates the biological process and pathways underlying β-cell differentiation, and promotes the identification and functional analysis of potential genes that could be used for improving functional β-cell generation from iPSCs.
Collapse
Affiliation(s)
- Yan Huang
- 1 Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Jian Wan
- 1 Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Yibing Guo
- 2 Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Shajun Zhu
- 1 Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Yao Wang
- 1 Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Lei Wang
- 1 Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Qingsong Guo
- 1 Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Yuhua Lu
- 1 Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Zhiwei Wang
- 1 Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| |
Collapse
|
14
|
Millette K, Georgia S. Gene Editing and Human Pluripotent Stem Cells: Tools for Advancing Diabetes Disease Modeling and Beta-Cell Development. Curr Diab Rep 2017; 17:116. [PMID: 28980194 DOI: 10.1007/s11892-017-0947-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
PURPOSE OF REVIEW This review will focus on the multiple approaches to gene editing and address the potential use of genetically modified human pluripotent stem cell-derived beta cells (SC-β) as a tool to study human beta-cell development and model their function in diabetes. We will explore how new variations of CRISPR/Cas9 gene editing may accelerate our understanding of beta-cell developmental biology, elucidate novel mechanisms that establish and regulate beta-cell function, and assist in pioneering new therapeutic modalities for treating diabetes. RECENT FINDINGS Improvements in CRISPR/Cas9 target specificity and homology-directed recombination continue to advance its use in engineering stem cells to model and potentially treat disease. We will review how CRISPR/Cas9 gene editing is informing our understanding of beta-cell development and expanding the therapeutic possibilities for treating diabetes and other diseases. Here we focus on the emerging use of gene editing technology, specifically CRISPR/Cas9, as a means of manipulating human gene expression to gain novel insights into the roles of key factors in beta-cell development and function. Taken together, the combined use of SC-β cells and CRISPR/Cas9 gene editing will shed new light on human beta-cell development and function and accelerate our progress towards developing new therapies for patients with diabetes.
Collapse
Affiliation(s)
- Katelyn Millette
- Center for Endocrinology, Diabetes and Metabolism, Department of Pediatrics, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Senta Georgia
- Center for Endocrinology, Diabetes and Metabolism, Department of Pediatrics, Children's Hospital Los Angeles, Los Angeles, CA, USA.
- Departments of Pediatrics and Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
- Developmental Biology and Regenerative Medicine Program, Saban Research Institute of Children's Hospital Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
15
|
Abstract
The pancreas is a complex organ with exocrine and endocrine components. Many pathologies impair exocrine function, including chronic pancreatitis, cystic fibrosis and pancreatic ductal adenocarcinoma. Conversely, when the endocrine pancreas fails to secrete sufficient insulin, patients develop diabetes mellitus. Pathology in either the endocrine or exocrine pancreas results in devastating economic and personal consequences. The current standard therapy for treating patients with type 1 diabetes mellitus is daily exogenous insulin injections, but cell sources of insulin provide superior glycaemic regulation and research is now focused on the goal of regenerating or replacing β cells. Stem-cell-based models might be useful to study exocrine pancreatic disorders, and mesenchymal stem cells or secreted factors might delay disease progression. Although the standards that bioengineered cells must meet before being considered as a viable therapy are not yet established, any potential therapy must be acceptably safe and functionally superior to current therapies. Here, we describe progress and challenges in cell-based methods to restore pancreatic function, with a focus on optimizing the site for cell delivery and decreasing requirements for immunosuppression through encapsulation. We also discuss the tools and strategies being used to generate exocrine pancreas and insulin-producing β-cell surrogates in situ and highlight obstacles to clinical application.
Collapse
|
16
|
Late-stage differentiation of embryonic pancreatic β-cells requires Jarid2. Sci Rep 2017; 7:11643. [PMID: 28912479 PMCID: PMC5599523 DOI: 10.1038/s41598-017-11691-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 08/24/2017] [Indexed: 02/04/2023] Open
Abstract
Jarid2 is a component of the Polycomb Repressor complex 2 (PRC2), which is responsible for genome-wide H3K27me3 deposition, in embryonic stem cells. However, Jarid2 has also been shown to exert pleiotropic PRC2-independent actions during embryogenesis. Here, we have investigated the role of Jarid2 during pancreas development. Conditional ablation of Jarid2 in pancreatic progenitors results in reduced endocrine cell area at birth due to impaired endocrine cell differentiation and reduced prenatal proliferation. Inactivation of Jarid2 in endocrine progenitors demonstrates that Jarid2 functions after endocrine specification. Furthermore, genome-wide expression analysis reveals that Jarid2 is required for the complete activation of the insulin-producing β-cell differentiation program. Jarid2-deficient pancreases exhibit impaired deposition of RNAPII-Ser5P, the initiating form of RNAPII, but no changes in H3K27me3, at the promoters of affected endocrine genes. Thus, our study identifies Jarid2 as a fine-tuner of gene expression during late stages of pancreatic endocrine cell development. These findings are relevant for generation of transplantable stem cell-derived β-cells.
Collapse
|
17
|
Cnop M, Toivonen S, Igoillo-Esteve M, Salpea P. Endoplasmic reticulum stress and eIF2α phosphorylation: The Achilles heel of pancreatic β cells. Mol Metab 2017; 6:1024-1039. [PMID: 28951826 PMCID: PMC5605732 DOI: 10.1016/j.molmet.2017.06.001] [Citation(s) in RCA: 179] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 05/19/2017] [Accepted: 06/01/2017] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Pancreatic β cell dysfunction and death are central in the pathogenesis of most if not all forms of diabetes. Understanding the molecular mechanisms underlying β cell failure is important to develop β cell protective approaches. SCOPE OF REVIEW Here we review the role of endoplasmic reticulum stress and dysregulated endoplasmic reticulum stress signaling in β cell failure in monogenic and polygenic forms of diabetes. There is substantial evidence for the presence of endoplasmic reticulum stress in β cells in type 1 and type 2 diabetes. Direct evidence for the importance of this stress response is provided by an increasing number of monogenic forms of diabetes. In particular, mutations in the PERK branch of the unfolded protein response provide insight into its importance for human β cell function and survival. The knowledge gained from different rodent models is reviewed. More disease- and patient-relevant models, using human induced pluripotent stem cells differentiated into β cells, will further advance our understanding of pathogenic mechanisms. Finally, we review the therapeutic modulation of endoplasmic reticulum stress and signaling in β cells. MAJOR CONCLUSIONS Pancreatic β cells are sensitive to excessive endoplasmic reticulum stress and dysregulated eIF2α phosphorylation, as indicated by transcriptome data, monogenic forms of diabetes and pharmacological studies. This should be taken into consideration when devising new therapeutic approaches for diabetes.
Collapse
Key Words
- ATF, activating transcription factor
- CHOP, C/EBP homologous protein
- CRISPR, clustered regularly interspaced short palindromic repeats
- CReP, constitutive repressor of eIF2α phosphorylation
- Diabetes
- ER, endoplasmic reticulum
- ERAD, ER-associated degradation
- Endoplasmic reticulum stress
- GCN2, general control non-derepressible-2
- GIP, glucose-dependent insulinotropic polypeptide
- GLP-1, glucagon-like peptide 1
- GWAS, genome-wide association study
- HNF1A, hepatocyte nuclear factor 1-α
- HRI, heme-regulated inhibitor kinase
- IAPP, islet amyloid polypeptide
- IER3IP1, immediate early response-3 interacting protein-1
- IRE1, inositol-requiring protein-1
- ISR, integrated stress response
- Insulin
- Islet
- MEHMO, mental retardation, epilepsy, hypogonadism and -genitalism, microcephaly and obesity
- MODY, maturity-onset diabetes of the young
- NRF2, nuclear factor, erythroid 2 like 2
- PBA, 4-phenyl butyric acid
- PERK, PKR-like ER kinase
- PKR, protein kinase RNA
- PP1, protein phosphatase 1
- PPA, phenylpropenoic acid glucoside
- Pancreatic β cell
- Pdx1, pancreatic duodenal homeobox 1
- RIDD, regulated IRE1-dependent decay
- RyR2, type 2 ryanodine receptor/Ca2+ release channel
- SERCA, sarcoendoplasmic reticulum Ca2+ ATPase
- TUDCA, taurine-conjugated ursodeoxycholic acid derivative
- UPR, unfolded protein response
- WFS, Wolfram syndrome
- XBP1, X-box binding protein 1
- eIF2, eukaryotic translation initiation factor 2
- eIF2α
- hESC, human embryonic stem cell
- hPSC, human pluripotent stem cell
- hiPSC, human induced pluripotent stem cell
- uORF, upstream open reading frame
Collapse
Affiliation(s)
- Miriam Cnop
- ULB Center for Diabetes Research, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
- Division of Endocrinology, Erasmus Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Sanna Toivonen
- ULB Center for Diabetes Research, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Mariana Igoillo-Esteve
- ULB Center for Diabetes Research, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Paraskevi Salpea
- ULB Center for Diabetes Research, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
18
|
Lee MO, Jeon H, Son MY, Lee SC, Cho YS. Clump-passaging-based efficient 3D culture of human pluripotent stem cells under chemically defined conditions. Biochem Biophys Res Commun 2017; 493:723-730. [PMID: 28859981 DOI: 10.1016/j.bbrc.2017.08.124] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 08/28/2017] [Indexed: 12/27/2022]
Abstract
Large-scale production of human pluripotent stem cells (hPSCs) in an efficient and safe manner is crucial to the successful application of hPSCs in biomedical research and regenerative medicine. Three-dimensional culture methods for hPSCs have been extensively studied using single-cell passaging approaches; however, these techniques have been challenged by the induction of massive cell death and accumulation of genomic abnormalities. In this work, we developed and optimized a novel, simple clump-passaging method for in vitro hPSCs 3-dimensional (3D) culture that can be exploited for large-scale production. Fully grown hPSC spheroids were dissociated into smaller-sized spheroid clumps by simple treatment with enzyme-free dissociation buffer, and clumped hPSCs were inoculated and maintained for 3D suspension culture. Our clump-passaging method effectively increased the hPSCs survival rate after subculture and supported scalable hPSCs 3D expansion. We also tested and selected chemically defined media formulations that are suitable for 3D culture and commercially available. Overall, our clump-passaging and expansion method demonstrated high survival and expansion rates for hPSC spheroids compared with conventional methods and may also have the advantage of maintaining genomic stability.
Collapse
Affiliation(s)
- Mi-Ok Lee
- Stem Cell Research Laboratory, Immunotherapy Convergence Research Center, KRIBB, Daejeon, Republic of Korea.
| | - Hyejin Jeon
- Stem Cell Research Laboratory, Immunotherapy Convergence Research Center, KRIBB, Daejeon, Republic of Korea.
| | - Mi-Young Son
- Stem Cell Research Center, KRIBB, Daejeon, Republic of Korea; Department of Bioscience, KRIBB School, University of Science & Technology, Daejeon, Republic of Korea.
| | - Sang Chul Lee
- Research Center for Metabolic Regulation, KRIBB, Daejeon, Republic of Korea.
| | - Yee Sook Cho
- Stem Cell Research Laboratory, Immunotherapy Convergence Research Center, KRIBB, Daejeon, Republic of Korea; Department of Bioscience, KRIBB School, University of Science & Technology, Daejeon, Republic of Korea.
| |
Collapse
|
19
|
Jiang FX, Li K, Archer M, Mehta M, Jamieson E, Charles A, Dickinson JE, Matsumoto M, Morahan G. Differentiation of Islet Progenitors Regulated by Nicotinamide into Transcriptome-Verified β Cells That Ameliorate Diabetes. Stem Cells 2017; 35:1341-1354. [DOI: 10.1002/stem.2567] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 11/21/2016] [Accepted: 12/14/2016] [Indexed: 12/11/2022]
Affiliation(s)
- Fang-Xu Jiang
- Islet Cell Development Program, Harry Perkins Institute of Medical Research, and Centre for Medical Research
- Centre for Diabetes Research, Harry Perkins Institute of Medical Research, and Centre for Medical Research
| | - Kevin Li
- Islet Cell Development Program, Harry Perkins Institute of Medical Research, and Centre for Medical Research
- Centre for Diabetes Research, Harry Perkins Institute of Medical Research, and Centre for Medical Research
| | | | - Munish Mehta
- Centre for Diabetes Research, Harry Perkins Institute of Medical Research, and Centre for Medical Research
| | - Emma Jamieson
- Centre for Diabetes Research, Harry Perkins Institute of Medical Research, and Centre for Medical Research
| | - Adrian Charles
- School of Women's and Infants' Health; The University of Western Australia; Nedlands Australia
| | - Jan E. Dickinson
- School of Women's and Infants' Health; The University of Western Australia; Nedlands Australia
| | | | - Grant Morahan
- Centre for Diabetes Research, Harry Perkins Institute of Medical Research, and Centre for Medical Research
| |
Collapse
|
20
|
Stevens S. Synthetic Biology in Cell and Organ Transplantation. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a029561. [PMID: 28003184 DOI: 10.1101/cshperspect.a029561] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The transplantation of cells and organs has an extensive history, with blood transfusion and skin grafts described as some of the earliest medical interventions. The speed and efficiency of the human immune system evolved to rapidly recognize and remove pathogens; the human immune system also serves as a barrier against the transplant of cells and organs from even highly related donors. Although this shows the remarkable effectiveness of the immune system, the engineering of cells and organs that will survive in a host patient over the long term remains a steep challenge. Progress in the understanding of host immune responses to donor cells and organs, combined with the rapid advancement in synthetic biology applications, allows the rational engineering of more effective solutions for transplantation.
Collapse
Affiliation(s)
- Sean Stevens
- Mammalian Synthetic Biology, Synthetic Genomics, Inc., La Jolla, California 92037
| |
Collapse
|
21
|
Martinez-Sanchez A, Rutter GA, Latreille M. MiRNAs in β-Cell Development, Identity, and Disease. Front Genet 2017; 7:226. [PMID: 28123396 PMCID: PMC5225124 DOI: 10.3389/fgene.2016.00226] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 12/21/2016] [Indexed: 12/22/2022] Open
Abstract
Pancreatic β-cells regulate glucose metabolism by secreting insulin, which in turn stimulates the utilization or storage of the sugar by peripheral tissues. Insulin insufficiency and a prolonged period of insulin resistance are usually the core components of type 2 diabetes (T2D). Although, decreased insulin levels in T2D have long been attributed to a decrease in β-cell function and/or mass, this model has recently been refined with the recognition that a loss of β-cell “identity” and dedifferentiation also contribute to the decline in insulin production. MicroRNAs (miRNAs) are key regulatory molecules that display tissue-specific expression patterns and maintain the differentiated state of somatic cells. During the past few years, great strides have been made in understanding how miRNA circuits impact β-cell identity. Here, we review current knowledge on the role of miRNAs in regulating the acquisition of the β-cell fate during development and in maintaining mature β-cell identity and function during stress situations such as obesity, pregnancy, aging, or diabetes. We also discuss how miRNA function could be harnessed to improve our ability to generate β-cells for replacement therapy for T2D.
Collapse
Affiliation(s)
- Aida Martinez-Sanchez
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College London London, UK
| | - Guy A Rutter
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College London London, UK
| | - Mathieu Latreille
- Cellular Identity and Metabolism Group, MRC London Institute of Medical SciencesLondon, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College LondonLondon, UK
| |
Collapse
|
22
|
Sommese L, Zullo A, Mancini FP, Fabbricini R, Soricelli A, Napoli C. Clinical relevance of epigenetics in the onset and management of type 2 diabetes mellitus. Epigenetics 2017; 12:401-415. [PMID: 28059593 DOI: 10.1080/15592294.2016.1278097] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Epigenetics is involved in the altered expression of gene networks that underlie insulin resistance and insufficiency. Major genes controlling β-cell differentiation and function, such as PAX4, PDX1, and GLP1 receptor, are epigenetically controlled. Epigenetics can cause insulin resistance through immunomediated pro-inflammatory actions related to several factors, such as NF-kB, osteopontin, and Toll-like receptors. Hereafter, we provide a critical and comprehensive summary on this topic with a particular emphasis on translational and clinical aspects. We discuss the effect of epigenetics on β-cell regeneration for cell replacement therapy, the emerging bioinformatics approaches for analyzing the epigenetic contribution to type 2 diabetes mellitus (T2DM), the epigenetic core of the transgenerational inheritance hypothesis in T2DM, and the epigenetic clinical trials on T2DM. Therefore, prevention or reversion of the epigenetic changes occurring during T2DM development may reduce the individual and societal burden of the disease.
Collapse
Affiliation(s)
- Linda Sommese
- a U.O.C. Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology, Regional Reference Laboratory of Transplant Immunology , Department of Internal and Specialty Medicine , Azienda Ospedaliera Universitaria (AOU), Università degli Studi della Campania "Luigi Vanvitelli ," Italy.,b Department of Experimental Medicine , Second University of Naples , Italy
| | - Alberto Zullo
- c Department of Sciences and Technologies , University of Sannio , Benevento , Italy.,d CEINGE-Advanced Biotechnologies , Naples , Italy
| | | | - Rossella Fabbricini
- a U.O.C. Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology, Regional Reference Laboratory of Transplant Immunology , Department of Internal and Specialty Medicine , Azienda Ospedaliera Universitaria (AOU), Università degli Studi della Campania "Luigi Vanvitelli ," Italy
| | - Andrea Soricelli
- e IRCCS Research Institute SDN , Naples , Italy.,f Department of Studies of Institutions and Territorial Systems , University of Naples Parthenope , Naples , Italy
| | - Claudio Napoli
- a U.O.C. Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology, Regional Reference Laboratory of Transplant Immunology , Department of Internal and Specialty Medicine , Azienda Ospedaliera Universitaria (AOU), Università degli Studi della Campania "Luigi Vanvitelli ," Italy.,e IRCCS Research Institute SDN , Naples , Italy.,g Department of Medical, Surgical, Neurological, Metabolic and Geriatric Sciences , Second University of Naples , Italy
| |
Collapse
|
23
|
Jeffery N, Harries LW. β-cell differentiation status in type 2 diabetes. Diabetes Obes Metab 2016; 18:1167-1175. [PMID: 27550203 DOI: 10.1111/dom.12778] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 08/16/2016] [Accepted: 08/17/2016] [Indexed: 12/12/2022]
Abstract
Type 2 diabetes (T2D) affects 415 million people worldwide and is characterized by chronic hyperglycaemia and insulin resistance, progressing to insufficient insulin production, as a result of β-cell failure. Over time, chronic hyperglycaemia can ultimately lead to loss of β-cell function, leaving patients insulin-dependent. Until recently the loss of β-cell mass seen in T2D was considered to be the result of increased rates of apoptosis; however, it has been proposed that apoptosis alone cannot account for the extent of β-cell mass loss seen in the disease, and that a loss of function may also occur as a result of changes in β-cell differentiation status. In the present review, we consider current knowledge of determinants of β-cell fate in the context of understanding its relevance to disease process in T2D, and also the impact of a diabetogenic environment (hyperglycaemia, hypoxia, inflammation and dyslipidaemia) on the expression of genes involved in maintenance of β-cell identity. We describe current knowledge of the impact of the diabetic microenvironment on gene regulatory processes such alternative splicing, the expression of disallowed genes and epigenetic modifications. Elucidating the molecular mechanisms that underpin changes to β-cell differentiation status and the concomitant β-cell failure offers potential treatment targets for the future management of patients with T2D.
Collapse
Affiliation(s)
- Nicola Jeffery
- Department of Molecular Genetics, Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, University of Exeter, Devon, UK
| | - Lorna W Harries
- Department of Molecular Genetics, Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, University of Exeter, Devon, UK
| |
Collapse
|
24
|
Johnson JD. The quest to make fully functional human pancreatic beta cells from embryonic stem cells: climbing a mountain in the clouds. Diabetologia 2016; 59:2047-57. [PMID: 27473069 DOI: 10.1007/s00125-016-4059-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 06/23/2016] [Indexed: 01/10/2023]
Abstract
The production of fully functional insulin-secreting cells to treat diabetes is a major goal of regenerative medicine. In this article, I review progress towards this goal over the last 15 years from the perspective of a beta cell biologist. I describe the current state-of-the-art, and speculate on the general approaches that will be required to identify and achieve our ultimate goal of producing functional beta cells. The need for deeper phenotyping of heterogeneous cultures of stem cell derived islet-like cells in parallel with a better understanding of the heterogeneity of the target cell type(s) is emphasised. This deep phenotyping should include high-throughput single-cell analysis, as well as comprehensive 'omics technologies to provide unbiased characterisation of cell products and human beta cells. There are justified calls for more detailed and well-powered studies of primary human pancreatic beta cell physiology, and I propose online databases of standardised human beta cell responses to physiological stimuli, including both functional and metabolomic/proteomic/transcriptomic profiles. With a concerted, community-wide effort, including both basic and applied scientists, beta cell replacement will become a clinical reality for patients with diabetes.
Collapse
Affiliation(s)
- James D Johnson
- Diabetes Research Group, Life Sciences Institute, Department of Cellular and Physiological Sciences, University of British Columbia, 5358-2350 Health Sciences Mall, Vancouver, BC, Canada, V6T 1Z3.
| |
Collapse
|
25
|
Scharfmann R, Didiesheim M, Richards P, Chandra V, Oshima M, Albagli O. Mass production of functional human pancreatic β-cells: why and how? Diabetes Obes Metab 2016; 18 Suppl 1:128-36. [PMID: 27615142 DOI: 10.1111/dom.12728] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 05/17/2016] [Indexed: 12/17/2022]
Abstract
Diabetes (either type 1 or type 2) is due to insufficient functional β-cell mass. Research has, therefore, aimed to discover new ways to maintain or increase either β-cell mass or function. For this purpose, rodents have mainly been used as model systems and a large number of discoveries have been made. Meanwhile, although we have learned that rodent models represent powerful systems to model β-cell development, function and destruction, we realize that there are limitations when attempting to transfer the data to what is occurring in humans. Indeed, while human β-cells share many similarities with rodent β-cells, they also differ on a number of important parameters. In this context, developing ways to study human β-cell development, function and death represents an important challenge. This review will describe recent data on the development and use of convenient sources of human β-cells that should be useful tools to discover new ways to modulate functional β-cell mass in humans.
Collapse
Affiliation(s)
- R Scharfmann
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France.
| | - M Didiesheim
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France
| | - P Richards
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France
| | - V Chandra
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France
| | - M Oshima
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France
| | - O Albagli
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France
| |
Collapse
|
26
|
Beer NL, Gloyn AL. Genome-edited human stem cell-derived beta cells: a powerful tool for drilling down on type 2 diabetes GWAS biology. F1000Res 2016; 5:F1000 Faculty Rev-1711. [PMID: 27508066 PMCID: PMC4955023 DOI: 10.12688/f1000research.8682.1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/11/2016] [Indexed: 12/30/2022] Open
Abstract
Type 2 diabetes (T2D) is a disease of pandemic proportions, one defined by a complex aetiological mix of genetic, epigenetic, environmental, and lifestyle risk factors. Whilst the last decade of T2D genetic research has identified more than 100 loci showing strong statistical association with disease susceptibility, our inability to capitalise upon these signals reflects, in part, a lack of appropriate human cell models for study. This review discusses the impact of two complementary, state-of-the-art technologies on T2D genetic research: the generation of stem cell-derived, endocrine pancreas-lineage cells and the editing of their genomes. Such models facilitate investigation of diabetes-associated genomic perturbations in a physiologically representative cell context and allow the role of both developmental and adult islet dysfunction in T2D pathogenesis to be investigated. Accordingly, we interrogate the role that patient-derived induced pluripotent stem cell models are playing in understanding cellular dysfunction in monogenic diabetes, and how site-specific nucleases such as the clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9 system are helping to confirm genes crucial to human endocrine pancreas development. We also highlight the novel biology gleaned in the absence of patient lines, including an ability to model the whole phenotypic spectrum of diabetes phenotypes occurring both in utero and in adult cells, interrogating the non-coding 'islet regulome' for disease-causing perturbations, and understanding the role of other islet cell types in aberrant glycaemia. This article aims to reinforce the importance of investigating T2D signals in cell models reflecting appropriate species, genomic context, developmental time point, and tissue type.
Collapse
Affiliation(s)
- Nicola L. Beer
- Oxford Centre for Diabetes Endocrinology and Metabolism, Churchill Hospital, Oxford, UK,
| | - Anna L. Gloyn
- Oxford Centre for Diabetes Endocrinology and Metabolism, Churchill Hospital, Oxford, UK,Wellcome Trust Centre for Human Genetics, Oxford, UK,Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford, UK
| |
Collapse
|
27
|
Endothelial Progenitor Cells in Diabetic Microvascular Complications: Friends or Foes? Stem Cells Int 2016; 2016:1803989. [PMID: 27313624 PMCID: PMC4903148 DOI: 10.1155/2016/1803989] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 01/05/2016] [Accepted: 04/18/2016] [Indexed: 12/24/2022] Open
Abstract
Despite being featured as metabolic disorder, diabetic patients are largely affected by hyperglycemia-induced vascular abnormality. Accumulated evidence has confirmed the beneficial effect of endothelial progenitor cells (EPCs) in coronary heart disease. However, antivascular endothelial growth factor (anti-VEGF) treatment is the main therapy for diabetic retinopathy and nephropathy, indicating the uncertain role of EPCs in the pathogenesis of diabetic microvascular disease. In this review, we first illustrate how hyperglycemia induces metabolic and epigenetic changes in EPCs, which exerts deleterious impact on their number and function. We then discuss how abnormal angiogenesis develops in eyes and kidneys under diabetes condition, focusing on “VEGF uncoupling with nitric oxide” and “competitive angiopoietin 1/angiopoietin 2” mechanisms that are shared in both organs. Next, we dissect the nature of EPCs in diabetic microvascular complications. After we overview the current EPCs-related strategies, we point out new EPCs-associated options for future exploration. Ultimately, we hope that this review would uncover the mysterious nature of EPCs in diabetic microvascular disease for therapeutics.
Collapse
|
28
|
Saxena P, Heng BC, Bai P, Folcher M, Zulewski H, Fussenegger M. A programmable synthetic lineage-control network that differentiates human IPSCs into glucose-sensitive insulin-secreting beta-like cells. Nat Commun 2016; 7:11247. [PMID: 27063289 PMCID: PMC4831023 DOI: 10.1038/ncomms11247] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Accepted: 03/04/2016] [Indexed: 02/06/2023] Open
Abstract
Synthetic biology has advanced the design of standardized transcription control
devices that programme cellular behaviour. By coupling synthetic signalling cascade-
and transcription factor-based gene switches with reverse and differential
sensitivity to the licensed food additive vanillic acid, we designed a synthetic
lineage-control network combining vanillic acid-triggered mutually exclusive
expression switches for the transcription factors Ngn3 (neurogenin 3; OFF-ON-OFF)
and Pdx1 (pancreatic and duodenal homeobox 1; ON-OFF-ON) with the concomitant
induction of MafA (V-maf musculoaponeurotic fibrosarcoma oncogene homologue A;
OFF-ON). This designer network consisting of different network topologies
orchestrating the timely control of transgenic and genomic Ngn3, Pdx1 and MafA
variants is able to programme human induced pluripotent stem cells (hIPSCs)-derived
pancreatic progenitor cells into glucose-sensitive insulin-secreting beta-like
cells, whose glucose-stimulated insulin-release dynamics are comparable to human
pancreatic islets. Synthetic lineage-control networks may provide the missing link
to genetically programme somatic cells into autologous cell phenotypes for
regenerative medicine. Synthetic biology offers the potential for the design and
implementation of rationally designed, complex genetic programmes. Here the authors
design a genetic network to trigger the differentiation of patient derived IPSCs into
beta-like cells.
Collapse
Affiliation(s)
- Pratik Saxena
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, CH-4058 Basel, Switzerland
| | - Boon Chin Heng
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, CH-4058 Basel, Switzerland
| | - Peng Bai
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, CH-4058 Basel, Switzerland
| | - Marc Folcher
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, CH-4058 Basel, Switzerland
| | - Henryk Zulewski
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, CH-4058 Basel, Switzerland.,Division of Endocrinology, Diabetes and Metabolism, University Hospital Basel, Petersgraben 4, CH-4031 Basel, Switzerland
| | - Martin Fussenegger
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, CH-4058 Basel, Switzerland.,Faculty of Science, University of Basel, Mattenstrasse 26, CH-4058 Basel, Switzerland
| |
Collapse
|
29
|
Mojibian M, Glavas MM, Kieffer TJ. Engineering the gut for insulin replacement to treat diabetes. J Diabetes Investig 2016; 7 Suppl 1:87-93. [PMID: 27186362 PMCID: PMC4854511 DOI: 10.1111/jdi.12479] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Accepted: 01/06/2016] [Indexed: 12/11/2022] Open
Abstract
The gut epithelium's large surface area, its direct exposure to ingested nutrients, its vast stem cell population and its immunotolerogenic environment make it an excellent candidate for therapeutic cells to treat diabetes. Thus, several attempts have been made to coax immature gut cells to differentiate into insulin-producing cells by altering the expression patterns of specific transcription factors. Furthermore, because of similarities in enteroendocrine and pancreatic endocrine cell differentiation pathways, other approaches have used genetically engineered enteroendocrine cells to produce insulin in addition to their endogenous secreted hormones. Several studies support the utility of both of these approaches for the treatment of diabetes. Converting a patient's own gut cells into meal-regulated insulin factories in a safe and immunotolerogenic environment is an attractive approach to treat and potentially cure diabetes. Here, we review work on these approaches and indicate where we feel further advancements are required.
Collapse
Affiliation(s)
- Majid Mojibian
- Laboratory of Molecular and Cellular Medicine Department of Cellular and Physiological Sciences Life Sciences Institute University of British Columbia Vancouver British Columbia Canada
| | - Maria M Glavas
- Laboratory of Molecular and Cellular Medicine Department of Cellular and Physiological Sciences Life Sciences Institute University of British Columbia Vancouver British Columbia Canada
| | - Timothy J Kieffer
- Laboratory of Molecular and Cellular Medicine Department of Cellular and Physiological Sciences Life Sciences Institute University of British Columbia Vancouver British Columbia Canada
| |
Collapse
|
30
|
Altabas V, Altabas K, Kirigin L. Endothelial progenitor cells (EPCs) in ageing and age-related diseases: How currently available treatment modalities affect EPC biology, atherosclerosis, and cardiovascular outcomes. Mech Ageing Dev 2016; 159:49-62. [PMID: 26919825 DOI: 10.1016/j.mad.2016.02.009] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 01/25/2016] [Accepted: 02/22/2016] [Indexed: 12/15/2022]
Abstract
Endothelial progenitor cells (EPCs) are mononuclear cells that circulate in the blood and are derived from different tissues, expressing cell surface markers that are similar to mature endothelial cells. The discovery of EPCs has lead to new insights in vascular repair and atherosclerosis and also a new theory for ageing. EPCs from the bone marrow and some other organs aid in vascular repair by migrating to distant vessels where they differentiate into mature endothelial cells and replace old and injured endothelial cells. The ability of EPCs to repair vascular damage depends on their number and functionality. Currently marketed drugs used in a variety of diseases can modulate these characteristics. In this review, the effect of currently available treatment options for cardiovascular and metabolic disorders on EPC biology will be discussed. The various EPC-based therapies that will be discussed include lipid-lowering agents, antihypertensive agents, antidiabetic drugs, phosphodiesteraze inhibitors, hormones, as well as EPC capturing stents.
Collapse
Affiliation(s)
- Velimir Altabas
- Department of Internal Medicine, University Clinical Hospital "Sestre milosrdnice", Zagreb, Croatia.
| | - Karmela Altabas
- Department of Internal Medicine, University Clinical Hospital "Sestre milosrdnice", Zagreb, Croatia.
| | - Lora Kirigin
- Department of Internal Medicine, University Clinical Hospital "Sestre milosrdnice", Zagreb, Croatia.
| |
Collapse
|