1
|
Koç IY, Beler M, Ünal İ, Paker S, Emekli-Alturfan E, Alturfan AA, Cansız D. Investigating the effect of radiofrequency electromagnetic field exposure on molecular pathways related to insulin resistance and adipogenesis in zebrafish embryos - A pilot study without quantitative exposure metrics. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 954:176038. [PMID: 39245375 DOI: 10.1016/j.scitotenv.2024.176038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 08/06/2024] [Accepted: 09/03/2024] [Indexed: 09/10/2024]
Abstract
In recent years, obesity has become a global problem in children and adolescents, in parallel with the rapid increase in the use of information and communication technology. Recognizing the embryonic causes of obesity may help prevent adverse adult health outcomes. In our study, we hypothesized that radiofrequency-electromagnetic field (RF-EMF) exposure during embryogenesis would affect the molecular mechanisms related to adipogenesis and insulin resistance in zebrafish. To achieve this, we set up a system that emits RF-EMF in the 900 MHz band and subjected zebrafish embryos to its RF-EMF. We created two groups in which we exposed 30 min (EMF-30) and 60 min (EMF-60) per day, and a control group that was not exposed to RF-EMF. We ended the exposure at 96 hpf and analyzed the expression of lepa, ins, and pparg that are involved in the regulation of glucose and lipid metabolism. In addition, we analyzed oxidative stress parameters, embryonic development, and locomotor activity. We found decreased mRNA transcript abundance of lepa, ins, pparg, and activities of superoxide dismutase and acetylcholine esterase, along with increased lipid peroxidation (LPO), nitric oxide (NO), and glutathione S-transferase (GST). Locomotor activity increased in the EMF-30 group and decreased in the EMF-60 group. Our results showed that exposure to RF-EMF during the embryonic period disrupted the molecular pathways related to insulin resistance and adipogenesis in zebrafish. However, due to limited available resources, we were not able to appropriately quantify the actual RF exposure strength of the samples. Hence the results reported here should only be seen as preliminary, and further studies employing high quality exposure apparatus and dosimetry should be carried out in future.
Collapse
Affiliation(s)
- Irmak Yaldız Koç
- Faculty of Medicine, Department of Medical Biochemistry, Istanbul Medipol University, Istanbul, Turkey
| | - Merih Beler
- Institute of Health Sciences, Marmara University, Istanbul, Turkey
| | - İsmail Ünal
- Institute of Health Sciences, Marmara University, Istanbul, Turkey
| | - Selçuk Paker
- Faculty of Electrical and Electronic Engineering, Istanbul Technical University, Istanbul, Turkey
| | - Ebru Emekli-Alturfan
- Faculty of Dentistry, Department of Basic Medical Sciences, Marmara University, Istanbul, Turkey
| | - A Ata Alturfan
- Faculty of Medicine, Department of Biochemistry, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Derya Cansız
- Faculty of Medicine, Department of Medical Biochemistry, Istanbul Medipol University, Istanbul, Turkey.
| |
Collapse
|
2
|
Kaplan G, Beler M, Ünal I, Karagöz A, Eğilmezer G, Üstündağ ÜV, Cansız D, Alturfan AA, Emekli-Alturfan E. Diethylhexyl phthalate exposure amplifies oxidant and inflammatory response in fetal hyperglycemia model predisposing insulin resistance in zebrafish embryos. Toxicol Ind Health 2024; 40:232-243. [PMID: 38467557 DOI: 10.1177/07482337241238475] [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] [Indexed: 03/13/2024]
Abstract
Exposure of zebrafish embryos to glucose is a suitable model for the fetal hyperglycemia seen in gestational diabetes. Diethylhexyl phthalate (DEHP), which is considered an endocrine-disrupting chemical, is one of the most common phthalate derivatives used in stretching plastic and is encountered in every area where plastic is used in daily life. In the present study, the effects of DEHP on pathways related to insulin resistance and obesity were examined in zebrafish embryos exposed to glucose as a fetal hyperglycemia model. Zebrafish embryos were exposed to DEHP, glucose, and glucose + DEHP for 72 h post-fertilization (hpf), and developmental parameters and locomotor activities were monitored. At 72 hpf ins, lepa, pparγ, atf4a, and il-6 expressions were determined by RT-PCR. Glucose, lipid peroxidation (LPO), nitric oxide (NO) levels, glutathione S-transferase (GST), superoxide dismutase (SOD), and acetylcholine esterase (AChE) activities were measured spectrophotometrically. Compared with the control group, glucose, LPO, GST activity, il6, and atf4a expressions increased in all exposure groups, while body length, locomotor, and SOD activities decreased. While AChE activity decreased in the DEHP and glucose groups, it increased in the glucose + DEHP group. Although glucose exposure increased pparγ and lepa expressions, DEHP significantly decreased the expressions of pparγ and lepa both in the DEHP and glucose + DEHP groups. Our findings showed that DEHP amplified oxidant and inflammatory responses in this fetal hyperglycemia model, predisposing insulin resistance in zebrafish embryos.
Collapse
Affiliation(s)
- Gül Kaplan
- Institute of Health Sciences, Department Biochemistry, Marmara University, Istanbul, Turkey
| | - Merih Beler
- Institute of Health Sciences, Department Biochemistry, Marmara University, Istanbul, Turkey
| | - Ismail Ünal
- Institute of Health Sciences, Department Biochemistry, Marmara University, Istanbul, Turkey
| | - Atakan Karagöz
- Institute of Health Sciences, Department Biochemistry, Marmara University, Istanbul, Turkey
| | - Gizem Eğilmezer
- Institute of Health Sciences, Department Biochemistry, Marmara University, Istanbul, Turkey
| | - Ünsal Veli Üstündağ
- Department of Biochemistry, Faculty of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Derya Cansız
- Department of Biochemistry, Faculty of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - A Ata Alturfan
- Department of Biochemistry, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Ebru Emekli-Alturfan
- Department of Biochemistry, Faculty of Dentistry, Marmara University, Istanbul, Turkey
| |
Collapse
|
3
|
Dayer D, Bayati V, Ebrahimi M. Manipulation of Sonic Hedgehog Signaling Pathway in Maintenance, Differentiation, and Endocrine Activity of Insulin-Producing Cells: A Systematic Review. IRANIAN JOURNAL OF MEDICAL SCIENCES 2024; 49:65-76. [PMID: 38356490 PMCID: PMC10862108 DOI: 10.30476/ijms.2023.95425.2678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 10/30/2022] [Accepted: 11/01/2022] [Indexed: 02/16/2024]
Abstract
Background Some studies have evaluated the manipulation of the sonic hedgehog (Shh) signaling pathway to generate more efficient insulin-producing cells (IPCs). In a systematic review, we evaluated in vitro and in vivo studies on the effect of inhibition or activation of the Shh pathway on the production, differentiation, maintenance, and endocrine activity of IPCs. Methods A systematic review was conducted using all available experimental studies published between January 2000 and November 2022. The review aimed at determining the effect of Shh manipulation on the differentiation of stem cells (SCs) into IPCs. Keywords and phrases using medical subject headings were extracted, and a complete search was performed in Web of Science, Embase, ProQuest, PubMed, Scopus, and Cochrane Library databases. The inclusion criteria were manipulation of Shh in SCs, SCs differentiation into IPCs, and endocrine activity of mature IPCs. Articles with incomplete data and duplications were excluded. Results A total of 208 articles were initially identified, out of which 11 articles were included in the study. The effect of Shh inhibition in the definitive endoderm stage to produce functional IPCs were confirmed. Some studies showed the importance of Shh re-activation at late-stage differentiation for the generation of efficient IPCs. It is proposed that baseline concentrations of Shh in mature pancreatic β-cells affect insulin secretion and endocrine activities of the cells. However, Shh overexpression in pancreatic β-cells ultimately leads to improper endocrine function and inadequate glucose-sensing insulin secretion. Conclusion Accurate manipulation of the Shh signaling pathway can be an effective approach in the production and maintenance of functional IPCs.
Collapse
Affiliation(s)
- Dian Dayer
- Cellular and Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Vahid Bayati
- Cellular and Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Anatomy, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mina Ebrahimi
- Thalassemia and Hemoglobinopathy Research Center, Research Institute of Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
4
|
Nihad M, Shenoy P S, Bose B. Cell therapy research for Diabetes: Pancreatic β cell differentiation from pluripotent stem cells. Diabetes Res Clin Pract 2021; 181:109084. [PMID: 34673084 DOI: 10.1016/j.diabres.2021.109084] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 09/24/2021] [Accepted: 09/28/2021] [Indexed: 12/16/2022]
Abstract
Human pluripotent stem cells (PSCs), both embryonic and induced pluripotent stem cells (iPSCs), have been differentiated into pancreatic β isletsin vitrofor more than a decade. The idea is to get enough β cells for cell transplantation for diabetics. Finding a standard cell therapy for diabetes is essential because of the logarithmic increase in the global population of people with diabetes and the insufficient availability of the human cadaveric pancreas. Moreover, with better insights into developmental biology, thein vitroβ cell differentiation protocols have depended on thein vivoβ cell organogenesis. Various protocols for pancreatic β cell differentiation have been developed. Such protocols are based on the modulation of cell signalling pathways with growth factors, small molecules, RNAi approaches, directed differentiation using transcription factors, genome editing. Growth factor free differentiation protocols, epigenetic modulations, 3D differentiation approaches, and encapsulation strategies have also been reported for better glycemic control and endocrine modulations. Here, we have reviewed various aforementionedin vitroβ cell differentiation protocols from human PSCs, their respective comparisons, challenges, past, present, and future. The literature has been reviewed primarily from PubMed from the year 2000 till date using the mentioned keywords.
Collapse
Affiliation(s)
- Muhammad Nihad
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Pincode-575 018, Karnataka, India
| | - Sudheer Shenoy P
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Pincode-575 018, Karnataka, India
| | - Bipasha Bose
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Pincode-575 018, Karnataka, India.
| |
Collapse
|
5
|
Gyimah E, Dong X, Xu H, Zhang Z, Mensah JK. Embryonic Exposure to Low Concentrations of Bisphenol A and S Altered Genes Related to Pancreatic β-Cell Development and DNA Methyltransferase in Zebrafish. ARCHIVES OF ENVIRONMENTAL CONTAMINATION AND TOXICOLOGY 2021; 80:450-460. [PMID: 33471154 DOI: 10.1007/s00244-021-00812-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 01/02/2021] [Indexed: 06/12/2023]
Abstract
Bisphenol A (BPA) and bisphenol S (BPS) are implicated in the development of metabolic disorders, such diabetes mellitus. However, the epigenetic mechanism underlying the pancreatic β-cell dysregulation for both BPA/BPS needs clarification. This exploratory study was designed to investigate whether embryonic exposure to low BPA/BPS concentrations impair early pancreatic β-cell differentiation as well as DNA methylation in its gene expression profile using an in vivo model, zebrafish. Zebrafish embryos were exposed to 0, 0.01, 0.03, 0.1, 0.3, and 1.0 µM BPA/BPS at 4-h post fertilization (hpf) until 120 hpf. BPA/BPS-induced effects on pancreatic-related genes, insulin gene, and DNA methylation-associated genes were assessed at developmental stages (24-120 hpf), while glucose level was measure at the 120 hpf. The insulin expression levels decreased at 72-120 hpf for 1.0 µM BPA, while 0.32 and 0.24-fold of insulin expression were elicited by 0.3 and 1 µM BPS respectively at 72 hpf. Significant elevation of glucose levels; 16.3% (for 1.0 µM BPA), 7.20% (for 0.3 µM BPS), and 74.09% (for 1.0 µM BPS) higher than the control groups were observed. In addition, pancreatic-related genes pdx-1, foxa2, ptfla, and isl1 were significantly interfered compared with the untreated group. Moreover, the maintenance methylation gene, dnmt1, was monotonically and significantly decreased at early stage of development following BPA exposure but remained constant for BPS treatment relative to the control group. DNMT3a and DNMT3b orthologs were distinctively altered following BPA/BPS embryonic exposure. Our data indicated that embryonic exposure to low concentration of BPA/BPS can impair the normal expressions of pancreatic-associated genes and DNA methylation pattern of selected genes in zebrafish early development.
Collapse
Affiliation(s)
- Eric Gyimah
- Institute of Environmental Health and Ecological Security, School of Environment and Safety Engineering, Jiangsu University, Zhenjiang, 212013, China
| | - Xing Dong
- Institute of Environmental Health and Ecological Security, School of Environment and Safety Engineering, Jiangsu University, Zhenjiang, 212013, China
| | - Hai Xu
- Institute of Environmental Health and Ecological Security, School of Environment and Safety Engineering, Jiangsu University, Zhenjiang, 212013, China.
| | - Zhen Zhang
- Institute of Environmental Health and Ecological Security, School of Environment and Safety Engineering, Jiangsu University, Zhenjiang, 212013, China
| | - John Kenneth Mensah
- Department of Chemistry, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| |
Collapse
|
6
|
Tran R, Moraes C, Hoesli CA. Developmentally-Inspired Biomimetic Culture Models to Produce Functional Islet-Like Cells From Pluripotent Precursors. Front Bioeng Biotechnol 2020; 8:583970. [PMID: 33117786 PMCID: PMC7576674 DOI: 10.3389/fbioe.2020.583970] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 09/08/2020] [Indexed: 12/28/2022] Open
Abstract
Insulin-producing beta cells sourced from pluripotent stem cells hold great potential as a virtually unlimited cell source to treat diabetes. Directed pancreatic differentiation protocols aim to mimic various stimuli present during embryonic development through sequential changes of in vitro culture conditions. This is commonly accomplished by the timed addition of soluble signaling factors, in conjunction with cell-handling steps such as the formation of 3D cell aggregates. Interestingly, when stem cells at the pancreatic progenitor stage are transplanted, they form functional insulin-producing cells, suggesting that in vivo microenvironmental cues promote beta cell specification. Among these cues, biophysical stimuli have only recently emerged in the context of optimizing pancreatic differentiation protocols. This review focuses on studies of cell–microenvironment interactions and their impact on differentiating pancreatic cells when considering cell signaling, cell–cell and cell–ECM interactions. We highlight the development of in vitro cell culture models that allow systematic studies of pancreatic cell mechanobiology in response to extracellular matrix proteins, biomechanical effects, soluble factor modulation of biomechanics, substrate stiffness, fluid flow and topography. Finally, we explore how these new mechanical insights could lead to novel pancreatic differentiation protocols that improve efficiency, maturity, and throughput.
Collapse
Affiliation(s)
- Raymond Tran
- Department of Chemical Engineering, McGill University, Montreal, QC, Canada
| | - Christopher Moraes
- Department of Chemical Engineering, McGill University, Montreal, QC, Canada.,Department of Biomedical Engineering, McGill University, Montreal, QC, Canada
| | - Corinne A Hoesli
- Department of Chemical Engineering, McGill University, Montreal, QC, Canada.,Department of Biomedical Engineering, McGill University, Montreal, QC, Canada
| |
Collapse
|
7
|
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
|
8
|
Abstract
Regenerative medicine with stem cells holds great hope for the treatment of degenerative disease. The medical potential of embryonic stem cells remains relatively untapped at this point, and significant scientific hurdles remain to be overcome before these cells might be considered safe and effective for uses in patients. Meanwhile, adult stem cells have begun to show significant capabilities of their own in repair of damaged tissues, in both animal models and early patient trials.
Collapse
|
9
|
Salmikangas P, Menezes-Ferreira M, Reischl I, Tsiftsoglou A, Kyselovic J, Borg JJ, Ruiz S, Flory E, Trouvin JH, Celis P, Ancans J, Timon M, Pante G, Sladowski D, Lipnik-Stangelj M, Schneider CK. Manufacturing, characterization and control of cell-based medicinal products: challenging paradigms toward commercial use. Regen Med 2015; 10:65-78. [DOI: 10.2217/rme.14.65] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
During the past decade, a large number of cell-based medicinal products have been tested in clinical trials for the treatment of various diseases and tissue defects. However, licensed products and those approaching marketing authorization are still few. One major area of challenge is the manufacturing and quality development of these complex products, for which significant manipulation of cells might be required. While the paradigms of quality, safety and efficacy must apply also to these innovative products, their demonstration may be demanding. Demonstration of comparability between production processes and batches may be difficult for cell-based medicinal products. Thus, the development should be built around a well-controlled manufacturing process and a qualified product to guarantee reproducible data from nonclinical and clinical studies.
Collapse
Affiliation(s)
| | | | | | | | - Jan Kyselovic
- • Comenius University, Department of Pharmacology & Toxicology, Slovakia
| | | | - Sol Ruiz
- • Agencia Española de Medicamentos y Productos Sanitarios, Division of Biologicals & Biotechnology, Madrid, Spain
| | - Egbert Flory
- • Medical Biotechnology Division, Paul-Ehrlich-Institut, Langen, Germany
| | | | | | - Janis Ancans
- • University of Latvia, Faculty of Biology, Riga, Latvia
| | - Marcos Timon
- • Agencia Española de Medicamentos y Productos Sanitarios, Division of Biologicals & Biotechnology, Madrid, Spain
| | | | - Dariusz Sladowski
- • Medical University of Warsaw, Department of Transplantology & Central Tissue Bank, Warsaw, Poland
| | | | | |
Collapse
|
10
|
Tan G, Elefanty AG, Stanley EG. β-cell regeneration and differentiation: how close are we to the 'holy grail'? J Mol Endocrinol 2014; 53:R119-29. [PMID: 25385843 DOI: 10.1530/jme-14-0188] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Diabetes can be managed by careful monitoring of blood glucose and timely delivery of exogenous insulin. However, even with fastidious compliance, people with diabetes can suffer from numerous complications including atherosclerosis, retinopathy, neuropathy, and kidney disease. This is because delivery of exogenous insulin coupled with glucose monitoring cannot provide the fine level of glucose control normally provided by endogenous β-cells in the context of intact islets. Moreover, a subset of people with diabetes lack awareness of hypoglycemic events; a status that can have grave consequences. Therefore, much effort has been focused on replacing lost or dysfunctional β-cells with cells derived from other sources. The advent of stem cell biology and cellular reprogramming strategies have provided impetus to this work and raised hopes that a β-cell replacement therapy is on the horizon. In this review, we look at two components that will be required for successful β-cell replacement therapy: a reliable and safe source of β-cells and a mechanism by which such cells can be delivered and protected from host immune destruction. Particular attention is paid to insulin-producing cells derived from pluripotent stem cells because this platform addresses the issue of scale, one of the more significant hurdles associated with potential cell-based therapies. We also review methods for encapsulating transplanted cells, a technique that allows grafts to evade immune attack and survive for a long term in the absence of ongoing immunosuppression. In surveying the literature, we conclude that there are still several substantial hurdles that need to be cleared before a stem cell-based β-cell replacement therapy for diabetes becomes a reality.
Collapse
Affiliation(s)
- Gemma Tan
- Department of Anatomy and Developmental BiologyMonash University, Building 73, Clayton, Victoria 3800, AustraliaMurdoch Childrens Research InstituteThe Royal Children's Hospital, Flemington Road, Parkville, Victoria 3052, AustraliaDepartment of PaediatricsThe Royal Children's Hospital, University of Melbourne, Flemington Road, Parkville, Victoria 3052, Australia Department of Anatomy and Developmental BiologyMonash University, Building 73, Clayton, Victoria 3800, AustraliaMurdoch Childrens Research InstituteThe Royal Children's Hospital, Flemington Road, Parkville, Victoria 3052, AustraliaDepartment of PaediatricsThe Royal Children's Hospital, University of Melbourne, Flemington Road, Parkville, Victoria 3052, Australia
| | - Andrew G Elefanty
- Department of Anatomy and Developmental BiologyMonash University, Building 73, Clayton, Victoria 3800, AustraliaMurdoch Childrens Research InstituteThe Royal Children's Hospital, Flemington Road, Parkville, Victoria 3052, AustraliaDepartment of PaediatricsThe Royal Children's Hospital, University of Melbourne, Flemington Road, Parkville, Victoria 3052, Australia Department of Anatomy and Developmental BiologyMonash University, Building 73, Clayton, Victoria 3800, AustraliaMurdoch Childrens Research InstituteThe Royal Children's Hospital, Flemington Road, Parkville, Victoria 3052, AustraliaDepartment of PaediatricsThe Royal Children's Hospital, University of Melbourne, Flemington Road, Parkville, Victoria 3052, Australia Department of Anatomy and Developmental BiologyMonash University, Building 73, Clayton, Victoria 3800, AustraliaMurdoch Childrens Research InstituteThe Royal Children's Hospital, Flemington Road, Parkville, Victoria 3052, AustraliaDepartment of PaediatricsThe Royal Children's Hospital, University of Melbourne, Flemington Road, Parkville, Victoria 3052, Australia
| | - Edouard G Stanley
- Department of Anatomy and Developmental BiologyMonash University, Building 73, Clayton, Victoria 3800, AustraliaMurdoch Childrens Research InstituteThe Royal Children's Hospital, Flemington Road, Parkville, Victoria 3052, AustraliaDepartment of PaediatricsThe Royal Children's Hospital, University of Melbourne, Flemington Road, Parkville, Victoria 3052, Australia Department of Anatomy and Developmental BiologyMonash University, Building 73, Clayton, Victoria 3800, AustraliaMurdoch Childrens Research InstituteThe Royal Children's Hospital, Flemington Road, Parkville, Victoria 3052, AustraliaDepartment of PaediatricsThe Royal Children's Hospital, University of Melbourne, Flemington Road, Parkville, Victoria 3052, Australia Department of Anatomy and Developmental BiologyMonash University, Building 73, Clayton, Victoria 3800, AustraliaMurdoch Childrens Research InstituteThe Royal Children's Hospital, Flemington Road, Parkville, Victoria 3052, AustraliaDepartment of PaediatricsThe Royal Children's Hospital, University of Melbourne, Flemington Road, Parkville, Victoria 3052, Australia
| |
Collapse
|
11
|
Mansouri A, Esmaeili F, Nejatpour A, Houshmand F, Shabani L, Ebrahimie E. Differentiation of P19 embryonal carcinoma stem cells into insulin-producing cells promoted by pancreas-conditioned medium. J Tissue Eng Regen Med 2014; 10:600-12. [DOI: 10.1002/term.1927] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 04/25/2014] [Accepted: 05/05/2014] [Indexed: 12/27/2022]
Affiliation(s)
- Akram Mansouri
- Department of Biology, Faculty of Basic Sciences; Shahrekord University; Iran
| | - Fariba Esmaeili
- Research Institute of Biotechnology; Shahrekord University; Iran
- Department of Biology, Faculty of Basic Sciences; University of Isfahan; Iran
| | | | - Fariba Houshmand
- Department of Physiology, Faculty of Medical Sciences; Shahrekord University of Medical Sciences; Iran
| | - Leila Shabani
- Department of Biology, Faculty of Basic Sciences; Shahrekord University; Iran
- Research Institute of Biotechnology; Shahrekord University; Iran
| | - Esmaeil Ebrahimie
- Institute of Biotechnology; Shiraz University; Shiraz Iran
- School of Molecular and Biomedical Science; The University of Adelaide; Adelaide Australia
| |
Collapse
|
12
|
Shahjalal HM, Shiraki N, Sakano D, Kikawa K, Ogaki S, Baba H, Kume K, Kume S. Generation of insulin-producing β-like cells from human iPS cells in a defined and completely xeno-free culture system. J Mol Cell Biol 2014; 6:394-408. [PMID: 24970864 DOI: 10.1093/jmcb/mju029] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Human induced pluripotent stem (hiPS) cells are considered a potential source for the generation of insulin-producing pancreatic β-cells because of their differentiation capacity. In this study, we have developed a five-step xeno-free culture system to efficiently differentiate hiPS cells into insulin-producing cells in vitro. We found that a high NOGGIN concentration is crucial for specifically inducing the differentiation first into pancreatic and duodenal homeobox-1 (PDX1)-positive pancreatic progenitors and then into neurogenin 3 (NGN3)-expressing pancreatic endocrine progenitors, while suppressing the differentiation into hepatic or intestinal cells. We also found that a combination of 3-isobutyl-1-methylxanthine (IBMX), exendin-4, and nicotinamide was important for the differentiation into insulin single-positive cells that expressed various pancreatic β-cell markers. Most notably, the differentiated cells contained endogenous C-peptide pools that were released in response to various insulin secretagogues and high levels of glucose. Therefore, our results demonstrate the feasibility of generating hiPS-derived pancreatic β-cells under xeno-free conditions and highlight their potential to treat patients with type 1 diabetes.
Collapse
Affiliation(s)
- Hussain Md Shahjalal
- Department of Stem Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Honjo 2-2-1, Chuo-Ku, Kumamoto 860-0811, Japan Global-Center of Excellence (G-COE), Kumamoto University, Honjo 2-2-1, Chuo-ku, Kumamoto 860-0811, Japan
| | - Nobuaki Shiraki
- Department of Stem Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Honjo 2-2-1, Chuo-Ku, Kumamoto 860-0811, Japan
| | - Daisuke Sakano
- Department of Stem Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Honjo 2-2-1, Chuo-Ku, Kumamoto 860-0811, Japan Global-Center of Excellence (G-COE), Kumamoto University, Honjo 2-2-1, Chuo-ku, Kumamoto 860-0811, Japan
| | - Kazuhide Kikawa
- Department of Stem Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Honjo 2-2-1, Chuo-Ku, Kumamoto 860-0811, Japan Global-Center of Excellence (G-COE), Kumamoto University, Honjo 2-2-1, Chuo-ku, Kumamoto 860-0811, Japan Department of Pediatrics, Graduate School of Medical Sciences, Kumamoto University, Honjo 1-1-1, Chuo-Ku, Kumamoto 860-8556, Japan
| | - Soichiro Ogaki
- Department of Stem Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Honjo 2-2-1, Chuo-Ku, Kumamoto 860-0811, Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Honjo 1-1-1, Chuo-Ku, Kumamoto 860-8556, Japan
| | - Kazuhiko Kume
- Department of Neuropharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe Street, Mizuho, Nagoya 467-8603, Japan
| | - Shoen Kume
- Department of Stem Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Honjo 2-2-1, Chuo-Ku, Kumamoto 860-0811, Japan Global-Center of Excellence (G-COE), Kumamoto University, Honjo 2-2-1, Chuo-ku, Kumamoto 860-0811, Japan Program for Leading Graduate Schools 'HIGO (Health Life Science; Interdisciplinary and Glocal Oriented) Program,' Kumamoto University, Honjo 2-2-1, Chuo-ku, Kumamoto 860-0811, Japan
| |
Collapse
|
13
|
Lee DH, Chung HM. Differentiation into Endoderm Lineage: Pancreatic differentiation from Embryonic Stem Cells. Int J Stem Cells 2014; 4:35-42. [PMID: 24298332 DOI: 10.15283/ijsc.2011.4.1.35] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2011] [Indexed: 01/22/2023] Open
Abstract
The endoderm gives rise to digestive and respiratory tracts, thyroid, liver, and pancreas. Representative disease of endoderm lineages is type 1 diabetes resulting from destruction of the insulin-producing β cells. Generation of functional β cells from human embryonic stem (ES) cells in vitro can be practical, renewable cell source for replacement cell therapy for type 1 diabetes. It has been achieved by progressive instructive differentiation through each of the developmental stages. In this article, important studies of differentiation into pancreatic β cells from ES cells are reviewed through pancreatic developmental stages as definitive endoderm, primitive gut tube/foregut, and pancreatic cells. The investigation of differentiating ES cells from definitive endoderm to pancreas using signaling, arrays, and proteomics is also introduced.
Collapse
Affiliation(s)
- Dong Hyeon Lee
- Department of Physiology, School of Medicine, CHA University, Seongnam
| | | |
Collapse
|
14
|
Stem cells for pancreatic β-cell replacement in diabetes mellitus: actual perspectives. Curr Opin Organ Transplant 2014; 19:162-8. [PMID: 24553500 DOI: 10.1097/mot.0000000000000055] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
PURPOSE OF REVIEW Type 1 and type 2 diabetes mellitus represent a widespread metabolic disorder, related to autoimmune β-cell destruction and insulin resistance, leading to β-cell dysfunction, respectively, that are associated with severe chronic complications with irreversible multiorgan morphological and functional damage. Conventional treatment, based on exogenous insulin or oral agents may control and delay but not prevent the disease complications, which has lead, so far, to a steady increase in mortality and morbidity. β-Cell substitution cell therapy, initially pursued by whole pancreatic and isolated islet transplantation, with scarce and limited efficiency, now is looking at the new technologies for cell and molecular therapy for diabetes, based on stem cells. RECENT FINDINGS Pancreatic endocrine cells regeneration might replenish the destroyed β-cell pool, with neogenerated β-cell derived from pancreatic and extrapancreatic stem cell sources. Additionally, embryonic or adult stem cells derived from different cell lineages, and able to differentiate into β-like cell elements, may not only restore the original insulin secretory patterns but also exert the immunomodulatory effects aimed at interrupting the β-cell-directed autoimmune destruction vicious cycle. SUMMARY These new strategies may, one day, provide for the final cure of diabetes mellitus.
Collapse
|
15
|
Sheik Abdulazeez S. Diabetes treatment: A rapid review of the current and future scope of stem cell research. Saudi Pharm J 2013; 23:333-40. [PMID: 27134533 PMCID: PMC4834680 DOI: 10.1016/j.jsps.2013.12.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 12/14/2013] [Indexed: 12/13/2022] Open
Abstract
Diabetes mellitus is a major health concern of the developing and developed nations across the globe. This devastating disease accounts for the 5% deaths around the world annually. The current treatment methods do not address the underlying causes of the disease and have severe limitations. Stem cells are unique cells with the potential to differentiate into any type of specialized cells. This feature of both adult and embryonic stem cells was explored in great detail by the scientists around the world and are successful in producing insulin secreting cells. The different type of stem cells (induced pluripotent stem cells (iPSCs), embryonic stem cells (ESCs) and adult stem cells) proves to be potent in treating diabetes with certain limitations. This article precisely reviews the resources and progress made in the field of stem cell research for diabetic treatment.
Collapse
Affiliation(s)
- Sheriff Sheik Abdulazeez
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Shaqra University, Shaqra 11961, Kingdom of Saudi Arabia
| |
Collapse
|
16
|
Raikwar SP, Zavazava N. PDX1-engineered embryonic stem cell-derived insulin producing cells regulate hyperglycemia in diabetic mice. Transplant Res 2012; 1:19. [PMID: 23369186 PMCID: PMC3560994 DOI: 10.1186/2047-1440-1-19] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Accepted: 09/24/2012] [Indexed: 01/09/2023] Open
Abstract
Background Type 1 diabetes can be treated by the transplantation of cadaveric whole pancreata or isolated pancreatic islets. However, this form of treatment is hampered by the chronic shortage of cadaveric donors. Embryonic stem (ES) cell-derived insulin producing cells (IPCs) offer a potentially novel source of unlimited cells for transplantation to treat type 1 and possibly type 2 diabetes. However, thus far, the lack of a reliable protocol for efficient differentiation of ES cells into IPCs has hindered the clinical exploitation of these cells. Methods To efficiently generate IPCs using ES cells, we have developed a double transgenic ES cell line R1Pdx1AcGFP/RIP-Luc that constitutively expresses pancreatic β-cell-specific transcription factor pancreatic and duodenal homeobox gene 1 (Pdx1) as well as rat insulin promoter (RIP) driven luciferase reporter. We have established several protocols for the reproducible differentiation of ES cells into IPCs. The differentiation of ES cells into IPCs was monitored by immunostaining as well as real-time quantitative RT-PCR for pancreatic β-cell-specific markers. Pancreatic β-cell specific RIP became transcriptionally active following the differentiation of ES cells into IPCs and induced the expression of the luciferase reporter. Glucose stimulated insulin secretion by the ES cell-derived IPCs was measured by ELISA. Further, we have investigated the therapeutic efficacy of ES cell-derived IPCs to correct hyperglycemia in syngeneic streptozotocin (STZ)-treated diabetic mice. The long term fate of the transplanted IPCs co-expressing luciferase in syngeneic STZ-induced diabetic mice was monitored by real time noninvasive in vivo bioluminescence imaging (BLI). Results We have recently demonstrated that spontaneous in vivo differentiation of R1Pdx1AcGFP/RIP-Luc ES cell-derived pancreatic endoderm-like cells (PELCs) into IPCs corrects hyperglycemia in diabetic mice. Here, we investigated whether R1Pdx1AcGFP/RIP-Luc ES cells can be efficiently differentiated in vitro into IPCs. Our new data suggest that R1Pdx1AcGFP/RIP-Luc ES cells efficiently differentiate into glucose responsive IPCs. The ES cell differentiation led to pancreatic lineage commitment and expression of pancreatic β cell-specific genes, including Pax4, Pax6, Ngn3, Isl1, insulin 1, insulin 2 and PC2/3. Transplantation of the IPCs under the kidney capsule led to sustained long-term correction of hyperglycemia in diabetic mice. Although these newly generated IPCs effectively rescued hyperglycemic mice, an unexpected result was teratoma formation in 1 out of 12 mice. We attribute the development of the teratoma to the presence of either non-differentiated or partially differentiated stem cells. Conclusions Our data show the potential of Pdx1-engineered ES cells to enhance pancreatic lineage commitment and to robustly drive the differentiation of ES cells into glucose responsive IPCs. However, there is an unmet need for eliminating the partially differentiated stem cells.
Collapse
Affiliation(s)
- Sudhanshu P Raikwar
- Department of Internal Medicine, Division of Immunology, Roy J, and Lucille A, Carver College of Medicine, University of Iowa and Iowa City Veterans Affairs Medical Center, Building 41, Room #128, 601 Highway 6W, Iowa City, IA 52246, USA.
| | | |
Collapse
|
17
|
Iskovich S, Goldenberg-Cohen N, Stein J, Yaniv I, Fabian I, Askenasy N. Elutriated Stem Cells Derived from the Adult Bone Marrow Differentiate into Insulin-Producing Cells In Vivo and Reverse Chemical Diabetes. Stem Cells Dev 2012; 21:86-96. [DOI: 10.1089/scd.2011.0057] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Svetlana Iskovich
- Frankel Laboratory, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| | - Nitza Goldenberg-Cohen
- Krieger Laboratory, Center for Stem Cell Research, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| | - Jerry Stein
- Department of Pediatric Hematology-Oncology, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| | - Isaac Yaniv
- Department of Pediatric Hematology-Oncology, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| | - Ina Fabian
- Department of Cell Biology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nadir Askenasy
- Frankel Laboratory, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| |
Collapse
|
18
|
Saito H, Takeuchi M, Chida K, Miyajima A. Generation of glucose-responsive functional islets with a three-dimensional structure from mouse fetal pancreatic cells and iPS cells in vitro. PLoS One 2011; 6:e28209. [PMID: 22145030 PMCID: PMC3228734 DOI: 10.1371/journal.pone.0028209] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Accepted: 11/03/2011] [Indexed: 11/18/2022] Open
Abstract
Islets of Langerhans are a pancreatic endocrine compartment consisting of insulin-producing β cells together with several other hormone-producing cells. While some insulin-producing cells or immature pancreatic cells have been generated in vitro from ES and iPS cells, islets with proper functions and a three-dimensional (3D) structure have never been successfully produced. To test whether islets can be formed in vitro, we first examined the potential of mouse fetal pancreatic cells. We found that E16.5 pancreatic cells, just before forming islets, were able to develop cell aggregates consisting of β cells surrounded by glucagon-producing α cells, a structure similar to murine adult islets. Moreover, the transplantation of these cells improved blood glucose levels in hyperglycemic mice. These results indicate that functional islets are formed in vitro from fetal pancreatic cells at a specific developmental stage. By adopting these culture conditions to the differentiation of mouse iPS cells, we developed a two-step system to generate islets, i.e. immature pancreatic cells were first produced from iPS cells, and then transferred to culture conditions that allowed the formation of islets from fetal pancreatic cells. The islets exhibited distinct 3D structural features similar to adult pancreatic islets and secreted insulin in response to glucose concentrations. Transplantation of the islets improved blood glucose levels in hyperglycemic mice. In conclusion, the two-step culture system allows the generation of functional islets with a 3D structure from iPS cells.
Collapse
Affiliation(s)
- Hiroki Saito
- Institute of Molecular and Cellular Biosciences, University of Tokyo, Tokyo, Japan
- Graduate School of Agricultural and Life Sciences, University of Tokyo, Tokyo, Japan
- * E-mail: (HS); (AM)
| | - Masaki Takeuchi
- Institute of Molecular and Cellular Biosciences, University of Tokyo, Tokyo, Japan
| | - Kazuhiro Chida
- Graduate School of Agricultural and Life Sciences, University of Tokyo, Tokyo, Japan
| | - Atsushi Miyajima
- Institute of Molecular and Cellular Biosciences, University of Tokyo, Tokyo, Japan
- * E-mail: (HS); (AM)
| |
Collapse
|
19
|
Schroeder IS, Sulzbacher S, Nolden T, Fuchs J, Czarnota J, Meisterfeld R, Himmelbauer H, Wobus AM. Induction and Selection of Sox17-Expressing Endoderm Cells Generated from Murine Embryonic Stem Cells. Cells Tissues Organs 2011; 195:507-23. [DOI: 10.1159/000329864] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/07/2011] [Indexed: 01/16/2023] Open
|
20
|
Cheng H, Zhang YC, Wolfe S, Valencia V, Qian K, Shen L, Tang YL, Hsu WH, Atkinson MA, Phillips MI. Combinatorial treatment of bone marrow stem cells and stromal cell-derived factor 1 improves glycemia and insulin production in diabetic mice. Mol Cell Endocrinol 2011; 345:88-96. [PMID: 21801807 PMCID: PMC3171644 DOI: 10.1016/j.mce.2011.07.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2011] [Revised: 07/06/2011] [Accepted: 07/12/2011] [Indexed: 01/16/2023]
Abstract
Transdifferentiation of stem cells into insulin-producing cells for the treatment of diabetes have shown promising but inconsistent results. We examined the potential for attracting bone marrow stem cells (BMSCs) to the pancreas using a chemokine, stromal cell-derived factor 1 (SDF-1). SDF-1 treatment markedly increased the number of GFP labeled BMSCs in the pancreas, but surprisingly, the majority was observed in liver. The liver cells had typical pancreatic endocrine cell gene expression including insulin I, insulin II, PDX-1, somatostatin, and glucagon. Combined treatment with SDF-1 and BMSC transplant reduced hyperglycemia and prolonged the long-term survival of diabetic mice, and a sub group had complete normoglycemia (<150 mg/dl), restored blood insulin levels, and normal glucose tolerance. Our results suggest that SDF-1 could potentially be used to improve the homing of stem cells and β-cell regeneration. The mechanism appears to involve an increase in insulin producing cells mainly in the liver.
Collapse
Affiliation(s)
- H Cheng
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, 70803, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Anzalone R, Lo Iacono M, Loria T, Di Stefano A, Giannuzzi P, Farina F, La Rocca G. Wharton's jelly mesenchymal stem cells as candidates for beta cells regeneration: extending the differentiative and immunomodulatory benefits of adult mesenchymal stem cells for the treatment of type 1 diabetes. Stem Cell Rev Rep 2011; 7:342-63. [PMID: 20972649 DOI: 10.1007/s12015-010-9196-4] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSC) are uniquely capable of crossing germinative layers borders (i.e. are able to differentiate towards ectoderm-, mesoderm- and endoderm-derived cytotypes) and are viewed as promising cells for regenerative medicine approaches in several diseases. Type I diabetes therapy should potentially benefit from such differentiated cells: the search for alternatives to organ/islet transplantation strategies via stem cells differentiation is an ongoing task, significant goals having been achieved in most experimental settings (e.g. insulin production and euglycaemia restoration), though caution is still needed to ensure safe and durable effects in vivo. MSC are obtainable in high numbers via ex vivo culture and can be differentiated towards insulin-producing cells (IPC). Moreover, recent reports evidenced that MSC possess immunomodulatory activities (acting on both innate and acquired immunity effectors) which should result in a reduction of the immunogenicity of transplanted cells, thus limiting rejection. Moreover it has been proposed that MSC administration should be used to attenuate the autoimmune processes which lead to the destruction of beta cells. This review illustrates the recent advances made in differentiating human MSC to IPC. In particular, we compare the effectiveness of the differentiation protocols applied, the markers and functional assays used to characterize differentiated progeny, and the in vivo controls. We further speculate on how MSC derived from Wharton's jelly of human umbilical cord may represent a more promising regenerative medicine tool, as recently demonstrated for endoderm-derived organs (as liver) in human subjects, also considering their peculiar immunomodulatory features compared to other MSC populations.
Collapse
Affiliation(s)
- Rita Anzalone
- Sezione di Anatomia Umana, Dipartimento di Biomedicina Sperimentale e Neuroscienze Cliniche, Università degli Studi di Palermo, Via del Vespro 129, Palermo, PA 90127, Italy
| | | | | | | | | | | | | |
Collapse
|
22
|
Dimayuga VM, Rodriguez-Porcel M. Molecular imaging of cell therapy for gastroenterologic applications. Pancreatology 2011; 11:414-27. [PMID: 21912197 DOI: 10.1159/000327395] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Stem cell therapy has appeared as a possible therapeutic alternative for numerous diseases. Furthermore, cancer stem cells are a focus of significant interest as they may allow for a better understanding of the genesis of different malignancies. The ultimate goal of stem cell therapeutics is to ensure the viability and functionality of the transplanted cells. Similarly, the ultimate goal of understanding cancer stem cells is to understand how they behave in the living subject. Until recently, the efficacy of stem cell therapies has been assessed by overall organ function recovery. Understanding the behavior and biology of stem cells directly in the living subject can also lead to therapy optimization. Thus, there is a critical need for reliable and accurate methods to understand stem cell biology in vivo. Recent advances in both imaging and molecular biology have enabled transplanted stem cells to be successfully monitored in the living subject. The use of molecular imaging modalities has the capability to answer these questions and may one day be translated to patients. In this review, we will discuss the potential imaging strategies and how they can be utilized, depending on the questions that need to be answered.
Collapse
|
23
|
Insulin-producing surrogate β-cells from embryonic stem cells: are we there yet? Mol Ther 2011; 19:1759-68. [PMID: 21829172 DOI: 10.1038/mt.2011.165] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Embryonic stem cells (ESCs) harbor the potential to generate every cell type of the body by differentiation. The use of hESCs holds great promise for potential cell replacement therapies for degenerative diseases including diabetes mellitus. The recently discovered induced pluripotent stem cells (iPSCs) exhibit immense potential for regenerative medicine as they allow the generation of autologous cells tailored to the patients' immune system. Research for insulin-producing surrogate cells from ESCs has yielded highly controversial results, because many steps and factors in the differentiation process are currently still unknown. Thus, there is no consensus on common standard protocols. The protocols presently used established the differentiation from pluripotent cells toward pancreatic progenitor cells. However, none of the differentiation protocols reported to date have generated by exclusive in vitro differentiation sufficient numbers of insulin-producing cells meeting all essential criteria of a β-cell. The cells often lack the crucial function of regulated insulin secretion upon glucose stimulation. This review focuses on past and current approaches to the generation of insulin-producing cells from pluripotent sources, such as ESCs and iPSCs, and critically discusses the hurdles to be taken before insulin-secreting surrogate cells derived from these stem cells will be of clinical use in humans.
Collapse
|
24
|
Kelly C, Flatt CCS, McClenaghan NH. Stem cell-based approaches for the treatment of diabetes. Stem Cells Int 2011; 2011:424986. [PMID: 21716654 PMCID: PMC3116622 DOI: 10.4061/2011/424986] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Accepted: 03/18/2011] [Indexed: 01/10/2023] Open
Abstract
The incidence of diabetes and the associated debilitating complications are increasing at an alarming rate worldwide. Current therapies for type 1 diabetes focus primarily on administration of exogenous insulin to help restore glucose homeostasis. However, such treatment rarely prevents the long-term complications of this serious metabolic disorder, including neuropathy, nephropathy, retinopathy, and cardiovascular disease. Whole pancreas or islet transplantations have enjoyed limited success in some individuals, but these approaches are hampered by the shortage of suitable donors and the burden of lifelong immunosuppression. Here, we review current approaches to differentiate nonislet cell types towards an islet-cell phenotype which may be used for larger-scale cell replacement strategies. In particular, the differentiation protocols used to direct embryonic stem cells, progenitor cells of both endocrine and nonendocrine origin, and induced pluripotent stem cells towards an islet-cell phenotype are discussed.
Collapse
Affiliation(s)
- Catriona Kelly
- SAAD Centre for Pharmacy & Diabetes, Biomedical Sciences Research Institute, School of Biomedical Sciences, University of Ulster, Coleraine BT52 1SA, UK
| | | | | |
Collapse
|
25
|
Effects of histocompatibility and host immune responses on the tumorigenicity of pluripotent stem cells. Semin Immunopathol 2011; 33:573-91. [PMID: 21461989 PMCID: PMC3204002 DOI: 10.1007/s00281-011-0266-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2011] [Accepted: 03/16/2011] [Indexed: 12/11/2022]
Abstract
Pluripotent stem cells hold great promises for regenerative medicine. They might become useful as a universal source for a battery of new cell replacement therapies. Among the major concerns for the clinical application of stem cell-derived grafts are the risks of immune rejection and tumor formation. Pluripotency and tumorigenicity are closely linked features of pluripotent stem cells. However, the capacity to form teratomas or other tumors is not sufficiently described by inherited features of a stem cell line or a stem cell-derived graft. The tumorigenicity always depends on the inability of the recipient to reject the tumorigenic cells. This review summarizes recent data on the tumorigenicity of pluripotent stem cells in immunodeficient, syngeneic, allogeneic, and xenogeneic hosts. The effects of immunosuppressive treatment and cell differentiation are discussed. Different immune effector mechanisms appear to be involved in the rejection of undifferentiated and differentiated cell populations. Elements of the innate immune system, such as natural killer cells and the complement system, which are active also in syngeneic recipients, appear to preferentially reject undifferentiated cells. This effect could reduce the risk of tumor formation in immunocompetent recipients. Cell differentiation apparently increases susceptibility to rejection by the adaptive immune system in allogeneic hosts. The current data suggest that the immune system of the recipient has a major impact on the outcome of pluripotent stem cell transplantation, whether it is rejection, engraftment, or tumor development. This has to be considered when the results of experimental transplantation models are interpreted and even more when translation into clinics is planned.
Collapse
|
26
|
Gillard P, Mathieu C. Immune and cell therapy in type 1 diabetes: too little too late? Expert Opin Biol Ther 2011; 11:609-21. [PMID: 21406028 DOI: 10.1517/14712598.2011.560568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Type 1 diabetes is caused by autoimmune destruction of insulin-producing β-cells. Intensive insulin therapy protects most patients against chronic complications of diabetes, but exposes patients to acute complications like hypoglycaemia and impacts on quality of life. Therapies that aim at protecting or restoring endogenous insulin secretion might help in decreasing the risk of severe hypoglycemia and long-term complications. AREAS COVERED This article reviews the literature of clinical immunotherapy and β-cell transplantation in treatment of type 1 diabetes with specific focus on the effect on preserving and restoring β-cell mass. EXPERT OPINION Several studies in recent-onset type 1 diabetic patients have provided proof of principle that immunotherapy can preserve residual functional β-cell mass. The observation that this strategy is most effective early in the disease process opens possibilities of arresting and even preventing type 1 diabetes. In patients with too few or no surviving β-cells, current protocols of β-cell transplantation can restore functional β-cell mass up to 25% of levels in healthy controls. Unfortunately, both strategies to date are followed by progressive decline of endogenous insulin secretion later on. Strategies to restore functional β-cell mass to a higher level and to restore immune tolerance are thus needed.
Collapse
Affiliation(s)
- Pieter Gillard
- University Hospital Leuven, Department of Experimental Medicine and Endocrinology, KULeuven, Herestraat 49, B-3000 Leuven, Belgium
| | | |
Collapse
|
27
|
Vicente-Salar N, Santana A, Reig JA, Roche E. Differentiation of Embryonic Stem Cells Using Pancreatic Bud-Conditioned Medium Gives Rise to Neuroectoderm-Derived Insulin-Secreting Cells. Cell Reprogram 2011; 13:77-84. [DOI: 10.1089/cell.2010.0054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Nestor Vicente-Salar
- Research Foundation of Alicante Universitary General Hospital, Hepatology Unit, Alicante, Spain
| | - Alfredo Santana
- Research Unit, Gran Canaria Hospital Dr. Negrin and Genetic Unit, Childhood Hospital Complex, Las Palmas, Canary Islands, Spain
| | - Juan A. Reig
- Institute of Bioengineering, University Miguel Hernandez, Elche, Spain
| | - Enrique Roche
- Institute of Bioengineering, University Miguel Hernandez, Elche, Spain
| |
Collapse
|
28
|
Wen Y, Chen B, Ildstad ST. Stem cell-based strategies for the treatment of type 1 diabetes mellitus. Expert Opin Biol Ther 2010; 11:41-53. [PMID: 21110785 DOI: 10.1517/14712598.2011.540235] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
IMPORTANCE OF THE FIELD β-Cell regeneration and β-cell preservation are two promising therapeutic approaches for the management of patients with type 1 diabetes (T1D). Stem cell-based strategies to address the problems of shortage in β cells, autoimmune and alloimmune responses have become an area of intense study. AREAS COVERED IN THIS REVIEW This review focuses on the progress that has been made in obtaining functional, insulin-producing cells from various types of stem/progenitor cells, including the current knowledge on the immunomodulatory roles of hematopoietic stem cell and multipotent stromal cell in the therapies for T1D. WHAT THE READER WILL GAIN A broad overview of recent advancements in this field is provided. The hurdles that remain in the path of using stem cell-based strategies for the treatment of T1D and possible approaches to overcome these challenges are discussed. TAKE HOME MESSAGE Stem cell-based strategies hold great promise for the treatment of T1D. In spite of the progress that has been made over the last decade, a number of obstacles and concerns need to be cleared before widespread clinical application is possible. In particular, the mechanism of ESC and iPSC-derived β-cell maturation in vivo is poorly understood.
Collapse
Affiliation(s)
- Yujie Wen
- University of Louisville, Institute for Cellular Therapeutics, Louisville, KY 40202-1760, USA
| | | | | |
Collapse
|
29
|
Banerjee I, Sharma N, Yarmush M. Impact of co-culture on pancreatic differentiation of embryonic stem cells. J Tissue Eng Regen Med 2010; 5:313-23. [PMID: 20717889 DOI: 10.1002/term.317] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2010] [Accepted: 04/16/2010] [Indexed: 02/06/2023]
Abstract
Promise of cellular therapy for type 1 diabetes has inspired the search for transplantable cell sources, and embryonic stem cells (ESCs) have emerged as strong candidates. We have developed a directed differentiation protocol to obtain insulin-producing cells from ESCs. The ESCs are first induced towards a homogeneous monolayer of definitive endoderm-like cells by co-culture with primary hepatocytes. Pancreatic commitment is induced by plating the ESC-derived endoderms on Matrigel, along with Sonic hedgehog inhibition and retinoid induction. More than 70% of differentiated cells positively upregulated Pdx-1, along with pro-endocrine transcription factors Ngn3, β2/neroD1, Nkx2.2 and Nkx6.1. Final maturation to islet-specific cells is achieved by co-culturing the ESC-derived pancreatic endocrine cells with endothelial cells, which resulted in Insulin 1 upregulation in 60% of the cell population, along with high levels of IAPP and Glut2. The differentiated cell population also secreted high levels of insulin. Our findings illustrate the significant effect of co-culture in different stages of differentiation and maturation of ESCs in vitro. Such a high yield of pancreatic islet cells has not yet been reported. Our findings establish a robust protocol for islet differentiation.
Collapse
Affiliation(s)
- Ipsita Banerjee
- Center for Engineering in Medicine, Harvard Medical School, Massachusetts General Hospital, Boston, MA 02114, USA.
| | | | | |
Collapse
|
30
|
Li H, Lam A, Xu AM, Sl Lam K, Kim Chung S. High dosage of Exendin-4 increased early insulin secretion in differentiated beta cells from mouse embryonic stem cells. Acta Pharmacol Sin 2010; 31:570-7. [PMID: 20418895 DOI: 10.1038/aps.2010.38] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
AIM To investigate early insulin release (EIR) and late insulin release (LIR) upon glucose challenge as well as important insulin signaling factors in differentiated insulin-producing cells from embryonic stem cells(ESCs). METHODS A recently published protocol was modified by increasing the concentration of Exendin-4 (from 0.1 nmol/L to 10 nmol/L) together with an additional 5-day culture in low glucose (5.5 mmol/L) medium after differentiation. Gene expression profile, insulin content, C-peptide, EIR and LIR were determined. RESULTS Compared to a lower concentration of Exendin-4 (0.1 nmol/L), a higher concentration of Exendin-4 (10 nmol/L) increased glucose-responsive insulin secretion, especially EIR. Moreover, 10 nmol/L Exendin-4 increased the expression of the following genes: insulin 1, Pdx-1 (an important transcription factor, newly recognized insulin signaling factors), Epac1 and Epac2 (exchange proteins directly activated by cAMP 1 and 2), and sulfonylurea receptor 1 (SUR1, the subunit of the K(ATP) channel). CONCLUSION According to current knowledge, our modified protocol with a higher concentration of Exendin-4 (10 nmol/L) together with an additional 5-day 5.5 mmol/L glucose culture after differentiation improved the efficiency of differentiation toward the beta cell phenotype, which was possibly the result of stimulated expression of Pdx-1, Epac 1, and Epac 2, which in turn inhibited the K(ATP) channel through combination with SUR1, leading to increased EIR upon glucose challenge.
Collapse
|
31
|
Glucagon like peptide-1-directed human embryonic stem cells differentiation into insulin-producing cells via hedgehog, cAMP, and PI3K pathways. Pancreas 2010; 39:315-22. [PMID: 19924023 DOI: 10.1097/mpa.0b013e3181bc30dd] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVES That glucagonlike peptide-1 (GLP-1) induces differentiation of primate embryonic stem (ES) cells into insulin-producing cells has been reported by several groups and also confirmed with our observations. METHODS To further elucidate the process in detail and the signaling pathways involved in this differentiation, we induced human ES cells HUES1 differentiated into insulin secretion cells by GLP-1 treatment. RESULTS A time-dependent pattern of down expression of the stem cell markers (human telomerase reverse transcriptase and octamer-4), and the appearance of multiple beta-cell-specific proteins (insulin, glucokinase, glucose transporter, type 2, and islet duodenal homeobox 1) and hedgehog signal molecules (Indian hedgehog, sonic hedgehog, and hedgehog receptor, patched) have been identified. Cotreatment with hedgehog signal inhibitor cytopamine was able to block this differentiation, providing evidence of the involvement of the hedgehog signaling pathway in GLP-1-induced differentiation. We also observed increased transcripts of the transcription factors of activator protein 1, serum response element-1, DNA-binding transcription factors, and cAMP response element in GLP-1-induced ES cell differentiation. Inhibition profile by its specific inhibitors indicated that the cyclic adenosine monophosphate and phosphatidylinositol-3-kinase pathways, but not the mitogen-activated protein kinase pathway, were required for the induced differentiation of ES cells. CONCLUSIONS These data support that GLP-1 directs human ES cell differentiation into insulin-producing cells via hedgehog, cyclic adenosine monophosphate, and phosphatidylinositol-3-kinase pathways.
Collapse
|
32
|
Halban PA, German MS, Kahn SE, Weir GC. Current status of islet cell replacement and regeneration therapy. J Clin Endocrinol Metab 2010; 95:1034-43. [PMID: 20061422 PMCID: PMC2841538 DOI: 10.1210/jc.2009-1819] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
CONTEXT Beta cell mass and function are decreased to varying degrees in both type 1 and type 2 diabetes. In the future, islet cell replacement or regeneration therapy may thus offer therapeutic benefit to people with diabetes, but there are major challenges to be overcome. EVIDENCE ACQUISITION A review of published peer-reviewed medical literature on beta-cell development and regeneration was performed. Only publications considered most relevant were selected for citation, with particular attention to the period 2000-2009 and the inclusion of earlier landmark studies. EVIDENCE SYNTHESIS Islet cell regenerative therapy could be achieved by in situ regeneration or implantation of cells previously derived in vitro. Both approaches are being explored, and their ultimate success will depend on the ability to recapitulate key events in the normal development of the endocrine pancreas to derive fully differentiated islet cells that are functionally normal. There is also debate as to whether beta-cells alone will assure adequate metabolic control or whether it will be necessary to regenerate islets with their various cell types and unique integrated function. Any approach must account for the potential dangers of regenerative therapy. CONCLUSIONS Islet cell regenerative therapy may one day offer an improved treatment of diabetes and potentially a cure. However, the various approaches are at an early stage of preclinical development and should not be offered to patients until shown to be safe as well as more efficacious than existing therapy.
Collapse
Affiliation(s)
- Philippe A Halban
- Department of Genetic Medicine and Development, University of Geneva, University Medical Center, 1 Rue Michel-Servet, 1211 Geneva 4, Switzerland.
| | | | | | | |
Collapse
|
33
|
Notch signaling in pancreatic endocrine cell and diabetes. Biochem Biophys Res Commun 2009; 392:247-51. [PMID: 20035712 DOI: 10.1016/j.bbrc.2009.12.115] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2009] [Accepted: 12/21/2009] [Indexed: 12/21/2022]
Abstract
Recent studies have improved our understanding of the physiological function of Notch signaling pathway and now there is compelling evidence demonstrating that Notch is a key regulator of embryonic development and tissue homeostasis. Although further extensive studies are necessary to illustrate the molecular mechanisms, new insights into the role of Notch signaling in pancreas development and diabetes have been achieved. Importantly, the ability to regulate Notch signaling intensity both positively and negatively may have therapeutic relevance for diabetes. Thus, this paper reviews the current knowledge of the roles of Notch signaling in the pancreatic endocrine cell system.
Collapse
|
34
|
Developmental Engineering: A New Paradigm for the Design and Manufacturing of Cell-Based Products. Part I: From Three-Dimensional Cell Growth to Biomimetics ofIn VivoDevelopment. TISSUE ENGINEERING PART B-REVIEWS 2009; 15:381-94. [DOI: 10.1089/ten.teb.2008.0575] [Citation(s) in RCA: 167] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
35
|
Wang X, Ye K. Three-dimensional differentiation of embryonic stem cells into islet-like insulin-producing clusters. Tissue Eng Part A 2009; 15:1941-52. [PMID: 19196138 DOI: 10.1089/ten.tea.2008.0181] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The production of mature pancreatic cells that function similarly to primary islets is the premise of cell therapy for diabetes. Here, we describe a novel approach to generating more mature insulin-producing cells from embryonic stem (ES) cells. A three-dimensional (3D) ES cell pancreatic differentiation system was developed and used to direct the ES cell differentiation into glucose-responsive, insulin-secreting cells. Using mouse ES cells as a model, we demonstrate that more mature insulin-producing cells can be generated from ES cells in 3D cultures. The 3D differentiated pancreatic endocrine cells can assemble into an islet-like tissue structure that displays greater similarities in phenotype and gene expression profile to adult mouse pancreatic islets, that is, with beta cells in the core and non-beta cells forming the mantel, leading to a significant improvement of the maturity of the insulin-producing cells. Our findings show that nearly 50-60% of the cells in 3D formed cell clusters express insulin. More importantly, those cells exhibit a high level of glucose-responsive insulin and C-peptide syntheses and release. A high level of expression of glucose transporter-2 was also detected in these cells. Compared to two-dimensional ES cell-derived insulin-producing cells, the insulin release from 3D ES cell-derived insulin-producing cells showed a nearly fivefold (p<0.05) increase when exposed to a high glucose (27.7 mM) medium. This 3D culture model provides an excellent system to study pancreatic endocrine morphogenesis and tissue organization. This study also demonstrates the feasibility of producing clinically relevant beta cells from ES cells in a 3D environment.
Collapse
Affiliation(s)
- Xiuli Wang
- Biomedical Engineering Program, College of Engineering, University of Arkansas, Fayetteville, AR 72701, USA
| | | |
Collapse
|
36
|
Abstract
With the already heightened demand placed on organ donation, stem cell therapy has become a tantalizing idea to provide glucose-responsive insulin-producing cells to Type 1 diabetic patients as an alternative to islet transplantation. Multiple groups have developed varied approaches to create a population of cells with the appropriate characteristics. Both adult and embryonic stem cells have received an enormous amount of attention as possible sources of insulin-producing cells. Although adult stem cells lack the pluripotent nature of their embryonic counterparts, they appear to avoid the ethical debate that has centred around the latter. This may limit the eventual application of embryonic stem cells, which have already shown promise in early mouse models. One must also consider the potential of stem cells to form teratomas, a complication which would prove devastating in an immunologically compromised transplant recipient. The present review looks at the progress to date in both the adult and embryonic stem cells fields as potential treatments for diabetes. We also consider some of the limitations of stem cell therapy and the potential complications that may develop with their use.
Collapse
|
37
|
Derivation of insulin-producing cells from human embryonic stem cells. Stem Cell Res 2009; 3:73-87. [DOI: 10.1016/j.scr.2009.08.003] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2009] [Revised: 08/01/2009] [Accepted: 08/18/2009] [Indexed: 12/21/2022] Open
|
38
|
Naujok O, Francini F, Picton S, Bailey CJ, Lenzen S, Jörns A. Changes in gene expression and morphology of mouse embryonic stem cells on differentiation into insulin-producing cells in vitro and in vivo. Diabetes Metab Res Rev 2009; 25:464-76. [PMID: 19425055 DOI: 10.1002/dmrr.965] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
BACKGROUND Embryonic stem (ES) cells have the potential to produce unlimited numbers of surrogate insulin-producing cells for cell replacement therapy of type 1 diabetes mellitus. The impact of the in vivo environment on mouse ES cell differentiation towards insulin-producing cells was analysed morphologically after implantation. METHODS ES cells differentiated in vitro into insulin-producing cells according to the Lumelsky protocol or a new four-stage differentiation protocol were analysed morphologically before and after implantation for gene expression by in situ reverse transcription polymerase chain reaction and protein expression by immunohistochemistry and ultrastructural analysis. RESULTS In comparison with nestin positive ES cells developed according to the reference protocol, the number of ES cells differentiated with the four-stage protocol increased under in vivo conditions upon morphological analysis. The cells exhibited, in comparison to the in vitro situation, increased gene and protein expression of Pdx1, insulin, islet amyloid polypeptide (IAPP), the GLUT2 glucose transporter and glucokinase, which are functional markers for glucose-induced insulin secretion of pancreatic beta cells. Renal sub-capsular implantation of ES cells with a higher degree of differentiation achieved by in vitro differentiation with a four-stage protocol enabled further significant maturation for the beta-cell-specific markers, insulin and the co-stored IAPP as well as the glucose recognition structures. In contrast, further in vivo differentiation was not achieved with cells differentiated in vitro by the reference protocol. CONCLUSIONS A sufficient degree of in vitro differentiation is an essential prerequisite for further substantial maturation in a beta-cell-specific way in vivo, supported by cell-cell contacts and vascularisation.
Collapse
Affiliation(s)
- Ortwin Naujok
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | | | | | | | | | | |
Collapse
|
39
|
Ben-Yehudah A, White C, Navara CS, Castro CA, Ize-Ludlow D, Shaffer B, Sukhwani M, Mathews CE, Chaillet JR, Witchel SF. Evaluating protocols for embryonic stem cell differentiation into insulin-secreting beta-cells using insulin II-GFP as a specific and noninvasive reporter. CLONING AND STEM CELLS 2009; 11:245-57. [PMID: 19508115 PMCID: PMC2996248 DOI: 10.1089/clo.2008.0074] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Stable and full differentiation of pluripotent stem cells into functional beta-cells offers the potential to treat type I diabetes with a theoretically inexhaustible source of replacement cells. In addition to the difficulties in directed differentiation, progress toward an optimized and reliable protocol has been hampered by the complication that cultured cells will concentrate insulin from the media, thus making it difficult to tell which, if any, cells are producing insulin. To address this, we utilized a novel murine embryonic stem cell (mESC) research model, in which the green fluorescent protein (GFP) has been inserted within the C-peptide of the mouse insulinII gene (InsulinII-GFP). Using this method, cells producing insulin are easily identified. We then compared four published protocols for differentiating mESCs into beta-cells to evaluate their relative efficiency by assaying intrinsic insulin production. Cells differentiated using each protocol were easily distinguished based on culture conditions and morphology. This comparison is strengthened because all testing is performed within the same laboratory by the same researchers, thereby removing interlaboratory variability in culture, cells, or analysis. Differentiated cells were analyzed and sorted based on GFP fluorescence as compared to wild type cells. Each differentiation protocol increased GFP fluorescence but only modestly. None of these protocols yielded more than 3% of cells capable of insulin biosynthesis indicating the relative inefficiency of all analyzed protocols. Therefore, improved beta-cells differentiation protocols are needed, and these insulin II GFP cells may prove to be an important tool to accelerate this process.
Collapse
Affiliation(s)
- Ahmi Ben-Yehudah
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Pittsburgh Development Center, Magee-Womens Research Institute and Foundation, University of Pittsburgh School of Medicine , Pittsburgh, PA, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Activin A-Induced Differentiation of Embryonic Stem Cells into Endoderm and Pancreatic Progenitors—The Influence of Differentiation Factors and Culture Conditions. Stem Cell Rev Rep 2009; 5:159-73. [DOI: 10.1007/s12015-009-9061-5] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2009] [Accepted: 02/19/2009] [Indexed: 02/07/2023]
|
41
|
Naujok O, Francini F, Picton S, Jörns A, Bailey CJ, Lenzen S. A new experimental protocol for preferential differentiation of mouse embryonic stem cells into insulin-producing cells. Cell Transplant 2009; 17:1231-42. [PMID: 19181217 DOI: 10.3727/096368908787236549] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Mouse embryonic stem (ES) cells have the potential to differentiate into insulin-producing cells, but efficient protocols for in vitro differentiation have not been established. Here we have developed a new optimized four-stage differentiation protocol and compared this with an established reference protocol. The new protocol minimized differentiation towards neuronal progeny, resulting in a population of insulin-producing cells with beta-cell characteristics but lacking neuronal features. The yield of glucagon and somatostatin cells was negligible. Crucial for this improved yield was the removal of a nestin selection step as well as removal of culture supplements that promote differentiation towards the neuronal lineage. Supplementation of the differentiation medium with insulin and fetal calf serum was beneficial for differentiation towards monohormonal insulin-positive cells. After implantation into diabetic mice these insulin-producing cells produced a time-dependent improvement of the diabetic metabolic state, in contrast to cells differentiated according to the reference protocol. Using a spinner culture instead of an adherent culture of ES cells prevented the differentiation towards insulin-producing cells. Thus, prevention of cell attachment in a spinner culture represents a means to keep ES cells in an undifferentiated state and to inhibit differentiation. In conclusion, this study describes a new optimized four-stage protocol for differentiating ES cells to insulin-producing cells with minimal neuronal cell formation.
Collapse
Affiliation(s)
- Ortwin Naujok
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | | | | | | | | | | |
Collapse
|
42
|
Evans R, Kotchetkova I, Langer S. Just around the corner: rhetorics of progress and promise in genetic research. PUBLIC UNDERSTANDING OF SCIENCE (BRISTOL, ENGLAND) 2009; 18:43-59. [PMID: 19579534 DOI: 10.1177/0963662507078016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
The emerging "diabetes epidemic" threatens to affect 366 million people worldwide by 2030. In the UK, almost 2 million people (about 3.9 percent of the population) are currently diagnosed with diabetes and it is estimated that a further 1 million people have the disease but do not realize it. The prevalence of diabetes, its complications and their effects on the lives of those living with diabetes mean that diabetes research has the potential to bring significant benefits. In this paper, we are concerned with the research involving human embryonic stem (HES) cells that sees diabetes as a potential therapeutic location. Drawing on the idea of the "certainty trough" we examine how the hopes and uncertainties associated with this complex research agenda are understood. We show that those at the research front and those most opposed to the research agenda appear to be the most aware of the uncertainties that need to be resolved. In contrast, funders, typically one-step removed from the research work, see the promise of the research as more real and more likely to be achieved. Significantly, these optimistic funders are supported in their beliefs by the research scientists as constitutive claims are reproduced within the contingent forum. The effect is a collaborative project in which the promise of a technical solution "just around the corner" is sustained whilst concerns about the future difficulties are marginalized.
Collapse
|
43
|
Raikwar SP, Zavazava N. Insulin producing cells derived from embryonic stem cells: are we there yet? J Cell Physiol 2008; 218:256-63. [PMID: 18932230 DOI: 10.1002/jcp.21615] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Derivation of insulin producing cells (IPCs) from embryonic stem (ES) cells provides a potentially innovative form of treatment for type 1 diabetes. Here, we discuss the current state of the art, unique challenges, and future directions on generating IPCs.
Collapse
Affiliation(s)
- Sudhanshu P Raikwar
- Division of Allergy and Immunology, Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | | |
Collapse
|
44
|
Eshpeter A, Jiang J, Au M, Rajotte RV, Lu K, Lebkowski JS, Majumdar AS, Korbutt GS. In vivo characterization of transplanted human embryonic stem cell-derived pancreatic endocrine islet cells. Cell Prolif 2008; 41:843-858. [PMID: 19040565 PMCID: PMC6495805 DOI: 10.1111/j.1365-2184.2008.00564.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2007] [Accepted: 02/29/2008] [Indexed: 11/29/2022] Open
Abstract
OBJECTIVES Islet-like clusters (ILCs), differentiated from human embryonic stem cells (hESCs), were characterized both before and after transplantation under the kidney capsule of streptozotocin-induced diabetic immuno-incompetent mice. MATERIALS AND METHODS Multiple independent ILC preparations (n = 8) were characterized by immunohistochemistry, flow cytometry and cell insulin content, with six preparations transplanted into diabetic mice (n = 42), compared to controls, which were transplanted with either a human fibroblast cell line or undifferentiated hESCs (n = 28). RESULTS Prior to transplantation, ILCs were immunoreactive for the islet hormones insulin, C-peptide and glucagon, and for the ductal epithelial marker cytokeratin-19. ILCs also had cellular insulin contents similar to or higher than human foetal islets. Expression of islet and pancreas-specific cell markers was maintained for 70 days post-transplantation. The mean survival of recipients was increased by transplanted ILCs as compared to transplanted human fibroblast cells (P < 0.0001), or undifferentiated hESCs (P < 0.042). Graft function was confirmed by secretion of human C-peptide in response to an oral bolus of glucose. CONCLUSIONS hESC-derived ILC grafts continued to contain cells that were positive for islet endocrine hormones and were shown to be functional by their ability to secrete human C-peptide. Further enrichment and maturation of ILCs could lead to generation of a sufficient source of insulin-producing cells for transplantation into patients with type 1 diabetes.
Collapse
Affiliation(s)
- A. Eshpeter
- Alberta Diabetes Institute and
- Department of Surgery, University of Alberta, Edmonton, Canada, and
| | - J. Jiang
- Geron Corporation, Menlo Park, CA, USA
| | - M. Au
- Geron Corporation, Menlo Park, CA, USA
| | - R. V. Rajotte
- Alberta Diabetes Institute and
- Department of Surgery, University of Alberta, Edmonton, Canada, and
| | - K. Lu
- Geron Corporation, Menlo Park, CA, USA
| | | | | | - G. S. Korbutt
- Alberta Diabetes Institute and
- Department of Surgery, University of Alberta, Edmonton, Canada, and
| |
Collapse
|
45
|
Milewski WM, Temple KA, Wesselschmidt RL, Hara M. Generation of embryonic stem cells from mouse insulin I promoter-green fluorescent protein transgenic mice and characterization in a teratoma model. In Vitro Cell Dev Biol Anim 2008; 45:1-5. [PMID: 18855079 DOI: 10.1007/s11626-008-9142-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2008] [Accepted: 08/25/2008] [Indexed: 11/29/2022]
Abstract
Insulin-secreting pancreatic beta cells play a key role in the pathogenesis of diabetes mellitus. Potential new treatments for this disease include cell-replacement therapies using embryonic stem cells (ESCs). We have generated ESCs from a transgenic mouse model, mouse insulin 1 promoter (MIP) green fluorescent protein (GFP) mice, in which embryonic and adult beta cells are genetically tagged with GFP. The aim of the present study is to examine the differentiation potential of MIP-GFP ESCs in the microenvironment of the kidney capsule. The ESCs grew rapidly and formed a teratoma with GFP-expressing beta-like cells present in clusters that formed a cord-like structure similar to what is seen in the embryonic pancreas. These structures also included glucagon-expressing alpha cells and amylase-expressing acinar cells. Electron microscopic analysis showed insulin-like granules in columnar epithelium with microvilli adjacent to exocrine-like granule-containing cells. The MIP-GFP ESCs should be a useful research tool to study the differentiation capacity of ESCs toward pancreatic lineages.
Collapse
Affiliation(s)
- Wieslawa M Milewski
- Department of Medicine, The University of Chicago, 5841 South Maryland Avenue, MC1027, Chicago, IL 60637, USA
| | | | | | | |
Collapse
|
46
|
Stammzellforschung – Status, Ausblick und bioethischer Aspekt. Wien Med Wochenschr 2008; 158:493-502. [DOI: 10.1007/s10354-008-0551-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2008] [Accepted: 03/11/2008] [Indexed: 12/22/2022]
|
47
|
Singh P, Williams DJ. Cell therapies: realizing the potential of this new dimension to medical therapeutics. J Tissue Eng Regen Med 2008; 2:307-19. [DOI: 10.1002/term.108] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
48
|
Abstract
Type 1 diabetes is characterized by the selective destruction of pancreatic β-cells caused by an autoimmune attack. Type 2 diabetes is a more complex pathology which, in addition to β-cell loss caused by apoptotic programs, includes β-cell dedifferentiation and peripheric insulin resistance. β-Cells are responsible for insulin production, storage and secretion in accordance to the demanding concentrations of glucose and fatty acids. The absence of insulin results in death and therefore diabetic patients require daily injections of the hormone for survival. However, they cannot avoid the appearance of secondary complications affecting the peripheral nerves as well as the eyes, kidneys and cardiovascular system. These afflictions are caused by the fact that external insulin injection does not mimic the tight control that pancreaticderived insulin secretion exerts on the body’s glycemia. Restoration of damaged β-cells by transplantation from exogenous sources or by endocrine pancreas regeneration would be ideal therapeutic options. In this context, stem cells of both embryonic and adult origin (including β-cell/islet progenitors) offer some interesting alternatives, taking into account the recent data indicating that these cells could be the building blocks from which insulin secreting cells could be generated in vitro under appropriate culture conditions. Although in many cases insulin-producing cells derived from stem cells have been shown to reverse experimentally induced diabetes in animal models, several concerns need to be solved before finding a definite medical application. These refer mainly to the obtainment of a cell population as similar as possible to pancreatic β-cells, and to the problems related with the immune compatibility and tumor formation. This review will summarize the different approaches that have been used to obtain insulin-producing cells from embryonic and adult stem cells, and the main problems that hamper the clinical applications of this technology.
Collapse
|
49
|
Naujok O, Francini F, Jörns A, Lenzen S. An efficient experimental strategy for mouse embryonic stem cell differentiation and separation of a cytokeratin-19-positive population of insulin-producing cells. Cell Prolif 2008; 41:607-24. [PMID: 18616698 DOI: 10.1111/j.1365-2184.2008.00541.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
OBJECTIVES Embryonic stem cells are a potential source for insulin-producing cells, but existing differentiation protocols are of limited efficiency. Here, the aim has been to develop a new one, which drives development of embryonic stem cells towards insulin-producing cells rather than to neuronal cell types, and to combine this with a strategy for their separation from insulin-negative cells. MATERIALS AND METHODS The cytokeratin-19 (CK19) promoter was used to control the expression of enhanced yellow fluorescence protein in mouse embryonic stem cells during their differentiation towards insulin-producing cells, using a new optimized four-stage protocol. Two cell populations, CK19(+) and CK19(-) cells, were successfully fluorescence sorted and analysed. RESULTS The new method reduced neuronal progeny and suppressed differentiation into glucagon- and somatostatin-producing cells. Concomitantly, beta-cell like characteristics of insulin-producing cells were strengthened, as documented by high gene expression of the Glut2 glucose transporter and the transcription factor Pdx1. This novel protocol was combined with a cell-sorting technique. Through the combined procedure, a fraction of glucose-responsive insulin-secreting CK19(+) cells was obtained with 40-fold higher insulin gene expression and 50-fold higher insulin content than CK19(-) cells. CK19(+) cells were immunoreactive for C-peptide and had ultrastructural characteristics of an insulin-secretory cell. CONCLUSION Differentiated CK19(+) cells reflect an endocrine precursor cell type of ductal origin, potentially suitable for insulin replacement therapy in diabetes.
Collapse
Affiliation(s)
- O Naujok
- Institute of Clinical Biochemistry, Hannover Medical School, D-30623 Hannover, Germany
| | | | | | | |
Collapse
|
50
|
Rovira M, Jané-Valbuena J, Marchand M, Savatier P, Real FX, Skoudy A. Viral-mediated coexpression of Pdx1 and p48 regulates exocrine pancreatic differentiation in mouse ES cells. CLONING AND STEM CELLS 2008; 9:327-38. [PMID: 17907943 DOI: 10.1089/clo.2006.0064] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Embryonic stem cells (ES) can spontaneously activate a pancreatic differentiation program in vitro, although with low efficiency. The aim was to improve such process by using viral mediated gene transduction. In this study, we have examined the suitability of using viral vectors to express key transcriptional factors involved in pancreatic development. ES cell lines that constitutively express Pdx1, a homeodomain protein involved in both exocrine and endocrine pancreatic development and differentiation, were established using a lentiviral vector. These cells were additionally infected with an adenovirus expressing p48, a bHLH factor that is also crucial for pancreatic development and acinar differentiation. Quantitative RT-PCR analysis demonstrated an increase in the expression of exocrine genes, including those coding for both digestive enzymes and transcription factors. Immunocytochemical staining also revealed an increase in the number of amylase-expressing cell clusters. However, other important genes involved in acinar cell maturation (i.e., Mist1) were not modulated under these conditions, suggesting that the cells display features of immature exocrine cells or because of an uncoupled gene expression of the exocrine differentiation program. Importantly, this effect was selective for the acinar lineage as the expression of a large set of endocrine markers remained unchanged. Therefore, combined expression of key genes involved in pancreatic development may be a promising approach to generate mature pancreatic exocrine cells.
Collapse
Affiliation(s)
- Meritxell Rovira
- Cell and Molecular Biology Unit, Institut Municipal d'Investigació Mèdica (IMIM), Dr Aiguader 88, Barcelona, Spain
| | | | | | | | | | | |
Collapse
|