1
|
Hawash M, Jaradat N, Abualhasan M, Jadallah J, Fashafsheh L, Zaid S, Qamhia N, Qneibi M, Qaoud MT, Tari O, Merski M, Boşnak AS, Mousa A, Issa L, Eid AM. Integrative bioinformatic and experimental analysis of benzoylbenzodioxol derivatives: hypoglycemic potential in diabetic mice. 3 Biotech 2024; 14:255. [PMID: 39350934 PMCID: PMC11438745 DOI: 10.1007/s13205-024-04103-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 09/19/2024] [Indexed: 10/04/2024] Open
Abstract
We investigated the hypoglycemic activity and pharmacokinetic study of two synthesized benzoyl benzodioxol derivatives, compound I (methyl 2-(6-(2-bromobenzoyl)benzo[d][1,3]dioxol-5-yl)acetate), and compound II, 2-(6-benzoylbenzo[d][1,3]dioxol-5-yl)acetic acid, which showed very strong α-amylase inhibiting activity in our previous study. Then, diabetes was induced by the injection of streptozotocin to mice. The molecular docking simulations and analyses of density functional theory analyses were conducted to study the binding interactions with human pancreatic alpha-amylase, and their pharmacokinetic properties were further evaluated by ADMET profiling. Compound I showed the most important hypoglycemic effect, decreasing the blood glucose by 32.4%, higher than that of compound II by 14.8% and even the positive control acarbose by 22.9%. Histopathological examination revealed that diabetic livers showed portal inflammation with some apoptotic hepatocytes due to streptozotocin treatment, whereas controls without any treatment maintained normal liver architecture. Molecular docking studies gave results for the best binding affinity of the compound I, through its strong water bridges and π-π interactions, and also through analysis with density functional theory, was more stable and reactive when compared to compound II. Further ADMET analysis showed that both compounds shared a promising pharmacokinetic profile, and compound I had the potential for CNS penetration. Thus, compound I was selected as the best candidate for developing new hypoglycemic agents with potent efficacy, good binding interactions, and excellent pharmacokinetic properties.
Collapse
Affiliation(s)
- Mohammed Hawash
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Nidal Jaradat
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Murad Abualhasan
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Jazeel Jadallah
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Lama Fashafsheh
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Salsabeela Zaid
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Naim Qamhia
- Department of Pathology, An-Najah National University Hospital, Nablus, 44839 Palestine
| | - Mohammad Qneibi
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, 44839 Palestine
| | - Mohammed T. Qaoud
- Department of Pharmacy, Faculty of Pharmacy, Cyprus International University, Northern Cyprus, Mersin 10, 99258 Nicosia, Türkiye
| | - Ozden Tari
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cukurova University, 01330 Adana, Türkiye
| | - Matthew Merski
- Instituto de Investigação E Inovação Em Saúde, Universidade Do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Ahmet S. Boşnak
- Department of Pharmacy, Faculty of Pharmacy, Cyprus International University, Northern Cyprus, Mersin 10, 99258 Nicosia, Türkiye
| | - Ahmed Mousa
- Department of Pathology, An-Najah National University Hospital, Nablus, 44839 Palestine
| | - Linda Issa
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Ahmad M. Eid
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| |
Collapse
|
2
|
Liu J, Yang Y, Qi Y. Efficacy of mesenchymal stromal cells in the treatment of type 1 diabetes: a systematic review. Cell Tissue Bank 2024; 25:663-676. [PMID: 38383908 PMCID: PMC11143029 DOI: 10.1007/s10561-024-10128-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 01/16/2024] [Indexed: 02/23/2024]
Abstract
To investigate the efficacy of mesenchymal stromal cells in the treatment of type 1 diabetes. Articles about the effects of mesenchymal stromal cells for T1D were retrieved in PubMed, Web of Science, Embase, and the Cochrane Library databases up to July 2023. Additional relevant studies were manually searched through citations. HbA1c, FBG, PBG, insulin requirement and C-peptide were assessed. The risk of bias was evaluated with the ROB 2.0 and ROBINS-I tools. Six RCTs and eight nRCTs were included. Of the 14 studies included, two evaluated BM-MSCs, three evaluated UC-MSCs, five evaluated AHSCT, two evaluated CB-SCs, and two evaluated UC-SCs plus aBM-MNCs. At the end of follow-up, ten studies found that mesenchymal stromal cells improved glycemic outcomes in T1D, while the remaining four studies showed no significant improvement. Findings support the positive impacts observed from utilizing mesenchymal stromal cells in individuals with T1D. However, the overall methodological quality of the identified studies and findings is heterogeneous, limiting the interpretation of the therapeutic benefits of mesenchymal stromal cells in T1D. Methodically rigorous research is needed to further increase credibility.
Collapse
Affiliation(s)
- Jiaxin Liu
- Department of Endocrinology, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, 430034, China
| | - Yang Yang
- Department of Endocrinology, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, 430034, China.
| | - Yun Qi
- Department of Endocrinology, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, 430034, China.
| |
Collapse
|
3
|
Chen J, Lu J, Wang SN, Miao CY. Application and challenge of pancreatic organoids in therapeutic research. Front Pharmacol 2024; 15:1366417. [PMID: 38855754 PMCID: PMC11157021 DOI: 10.3389/fphar.2024.1366417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 05/06/2024] [Indexed: 06/11/2024] Open
Abstract
The in-vivo non-human primate animal and in-vitro cell disease models play a crucial part in the study of the mechanisms underlying the occurrence and development of pancreatic diseases, but with increasingly prominent limitations with in-depth research. Organoids derived from human pluripotent and adult stem cells resemble human in-vivo organs in their cellular composition, spatial tissue structure and physiological function, making them as an advantageous research tool. Up until now, numerous human organoids, including pancreas, have been effectively developed, demonstrating significant potential for research in organ development, disease modeling, drug screening, and regenerative medicine. However, different from intestine, liver and other organs, the pancreas is the only special organ in the human body, consisting of an exocrine gland and an endocrine gland. Thus, the development of pancreatic organoid technology faces greater challenges, and how to construct a composite pancreatic organoid with exocrine and endocrine gland is still difficult in current research. By reviewing the fundamental architecture and physiological role of the human pancreas, along with the swiftly developing domain of pancreatic organoids, we summarize the method and characteristics of human pancreatic organoids, and its application in modeling pancreatic diseases, as a platform for individualized drug screening and in regenerative medicine study. As the first comprehensive review that focus on the pharmacological study of human pancreatic organoid, the review hopes to help scholars to have a deeper understanding in the study of pancreatic organoid.
Collapse
Affiliation(s)
- Jin Chen
- Department of Endocrinology and Metabolism, Changhai Hospital, Second Military University /Naval Medical University, Shanghai, China
- Department of Pharmacology, Second Military Medical University /Naval Medical University, Shanghai, China
| | - Jin Lu
- Department of Endocrinology and Metabolism, Changhai Hospital, Second Military University /Naval Medical University, Shanghai, China
| | - Shu-Na Wang
- Department of Pharmacology, Second Military Medical University /Naval Medical University, Shanghai, China
| | - Chao-Yu Miao
- Department of Pharmacology, Second Military Medical University /Naval Medical University, Shanghai, China
| |
Collapse
|
4
|
Singh A, Afshan N, Singh A, Singh SK, Yadav S, Kumar M, Sarma DK, Verma V. Recent trends and advances in type 1 diabetes therapeutics: A comprehensive review. Eur J Cell Biol 2023; 102:151329. [PMID: 37295265 DOI: 10.1016/j.ejcb.2023.151329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/12/2023] [Accepted: 06/03/2023] [Indexed: 06/12/2023] Open
Abstract
Type 1 diabetes (T1D) is a chronic autoimmune disease characterized by the destruction of pancreatic β-cells, leading to insulin deficiency. Insulin replacement therapy is the current standard of care for T1D, but it has significant limitations. However, stem cell-based replacement therapy has the potential to restore β-cell function and achieve glycaemic control eradicating the necessity for drugs or injecting insulin externally. While significant progress has been made in preclinical studies, the clinical translation of stem cell therapy for T1D is still in its early stages. In continuation, further research is essentially required to determine the safety and efficacy of stem cell therapies and to develop strategies to prevent immune rejection of stem cell-derived β-cells. The current review highlights the current state of cellular therapies for T1D including, different types of stem cell therapies, gene therapy, immunotherapy, artificial pancreas, and cell encapsulation being investigated, and their potential for clinical translation.
Collapse
Affiliation(s)
- Akash Singh
- Stem Cell Research Centre, Department of Haematology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, India
| | - Noor Afshan
- Stem Cell Research Centre, Department of Haematology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, India
| | - Anshuman Singh
- Stem Cell Research Centre, Department of Haematology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, India
| | - Suraj Kumar Singh
- Stem Cell Research Centre, Department of Haematology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, India
| | - Sudhanshu Yadav
- Stem Cell Research Centre, Department of Haematology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, India
| | - Manoj Kumar
- ICMR-National Institute for Research in Environmental Health, Bhopal, India
| | | | - Vinod Verma
- Stem Cell Research Centre, Department of Haematology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, India.
| |
Collapse
|
5
|
Tatum R, Wong D, Martins PN, Tchantchaleishvili V. Current status and future directions in the development and optimization of thoracic and abdominal artificial organs. Artif Organs 2023; 47:451-458. [PMID: 36421073 DOI: 10.1111/aor.14458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 09/18/2022] [Accepted: 11/06/2022] [Indexed: 11/27/2022]
Abstract
INTRODUCTION Artificial organs are engineered devices with the capacity to be implanted or integrated into a living body to replace a failing organ, or to duplicate or augment one or multiple functions of the diseased organ. AREAS COVERED We evaluate the present landscape and future possibilities of artificial organ engineering by exploring the spectrum of four distinguishable device features: mobility, compatibility, functionality, and material composition. These mechanical and functional differences provide the framework through which we examine the current status and future possibilities of the abdominal and thoracic artificial organs. EXPERT OPINION Transforming the artificial organs landscape in ways that expand the scope of existing device capabilities and improve the clinical utility of artificial organs will require making improvements upon existing technologies and multidisciplinary cooperation to create and discover new capacities.
Collapse
Affiliation(s)
- Robert Tatum
- Division of Cardiac Surgery, Department of Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Daniella Wong
- Division of Cardiac Surgery, Department of Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Paulo N Martins
- Division of Cardiac Surgery, Department of Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Vakhtang Tchantchaleishvili
- Division of Cardiac Surgery, Department of Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
6
|
Importance of multiple endocrine cell types in islet organoids for type 1 diabetes treatment. Transl Res 2022; 250:68-83. [PMID: 35772687 DOI: 10.1016/j.trsl.2022.06.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 06/08/2022] [Accepted: 06/21/2022] [Indexed: 11/21/2022]
Abstract
Almost 50 years ago, scientists developed the bi-hormonal abnormality hypothesis, stating that diabetes is not caused merely by the impaired insulin signaling. Instead, the presence of inappropriate level of glucagon is a prerequisite for the development of type 1 diabetes (T1D). It is widely understood that the hormones insulin and glucagon, secreted by healthy β and α cells respectively, operate in a negative feedback loop to maintain the body's blood sugar levels. Despite this fact, traditional T1D treatments rely solely on exogenous insulin injections. Furthermore, research on cell-based therapies and stem-cell derived tissues tends to focus on the replacement of β cells alone. In vivo, the pancreas is made up of 4 major endocrine cell types, that is, insulin-producing β cells, glucagon-producing α cells, somatostatin-producing δ cells, and pancreatic polypeptide-producing γ cells. These distinct cell types are involved synergistically in regulating islet functions. Therefore, it is necessary to produce a pancreatic islet organoid in vitro consisting of all these cell types that adequately replaces the function of the native islets. In this review, we describe the unique function of each pancreatic endocrine cell type and their interactions contributing to the maintenance of normoglycemia. Furthermore, we detail current sources of whole islets and techniques for their long-term expansion and culture. In addition, we highlight a vast potential of the pancreatic islet organoids for transplantation and diabetes research along with updated new approaches for successful transplantation using stem cell-derived islet organoids.
Collapse
|
7
|
Angelescu MA, Andronic O, Dima SO, Popescu I, Meivar-Levy I, Ferber S, Lixandru D. miRNAs as Biomarkers in Diabetes: Moving towards Precision Medicine. Int J Mol Sci 2022; 23:12843. [PMID: 36361633 PMCID: PMC9655971 DOI: 10.3390/ijms232112843] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 09/27/2022] [Accepted: 10/19/2022] [Indexed: 09/08/2023] Open
Abstract
Diabetes mellitus (DM) is a complex metabolic disease with many specifically related complications. Early diagnosis of this disease could prevent the progression to overt disease and its related complications. There are several limitations to using existing biomarkers, and between 24% and 62% of people with diabetes remain undiagnosed and untreated, suggesting a large gap in current diagnostic practices. Early detection of the percentage of insulin-producing cells preceding loss of function would allow for effective therapeutic interventions that could delay or slow down the onset of diabetes. MicroRNAs (miRNAs) could be used for early diagnosis, as well as for following the progression and the severity of the disease, due to the fact of their pancreatic specific expression and stability in various body fluids. Thus, many studies have focused on the identification and validation of such groups or "signatures of miRNAs" that may prove useful in diagnosing or treating patients. Here, we summarize the findings on miRNAs as biomarkers in diabetes and those associated with direct cellular reprogramming strategies, as well as the relevance of miRNAs that act as a bidirectional switch for cell therapy of damaged pancreatic tissue and the studies that have measured and tracked miRNAs as biomarkers in insulin resistance are addressed.
Collapse
Affiliation(s)
| | - Octavian Andronic
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 050474 Bucharest, Romania
- University Emergency Hospital, 050098 Bucharest, Romania
| | - Simona Olimpia Dima
- Center of Excelence in Translational Medicine (CEMT), Fundeni Clinical Institute, 022328 Bucharest, Romania
- Academy Nicolae Cajal Institute of Medical Scientific Research, Titu Maiorescu University, 040441 Bucharest, Romania
| | - Irinel Popescu
- Center of Excelence in Translational Medicine (CEMT), Fundeni Clinical Institute, 022328 Bucharest, Romania
- Academy Nicolae Cajal Institute of Medical Scientific Research, Titu Maiorescu University, 040441 Bucharest, Romania
| | - Irit Meivar-Levy
- Academy Nicolae Cajal Institute of Medical Scientific Research, Titu Maiorescu University, 040441 Bucharest, Romania
- Orgenesis Ltd., Ness Ziona 7414002, Israel
| | - Sarah Ferber
- Academy Nicolae Cajal Institute of Medical Scientific Research, Titu Maiorescu University, 040441 Bucharest, Romania
- Orgenesis Ltd., Ness Ziona 7414002, Israel
- Department of Human Genetics, Sackler School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Daniela Lixandru
- Center of Excelence in Translational Medicine (CEMT), Fundeni Clinical Institute, 022328 Bucharest, Romania
- Department of Biochemistry, University of Medicine and Pharmacy “Carol Davila”, 050474 Bucharest, Romania
| |
Collapse
|
8
|
Dang Le Q, Rodprasert W, Kuncorojakti S, Pavasant P, Osathanon T, Sawangmake C. In vitro generation of transplantable insulin-producing cells from canine adipose-derived mesenchymal stem cells. Sci Rep 2022; 12:9127. [PMID: 35650303 PMCID: PMC9160001 DOI: 10.1038/s41598-022-13114-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 05/20/2022] [Indexed: 11/27/2022] Open
Abstract
Canine mesenchymal stem cells (cMSCs) have potential applications for regenerative therapy, including the generation of insulin-producing cells (IPCs) for studying and treating diabetes. In this study, we established a useful protocol for generating IPCs from canine adipose mesenchymal stem cells (cAD-MSCs). Subsequently, in vitro preservation of pluronic F127-coated alginate (ALGPA)-encapsulated cAD-MSC-derived IPCs was performed to verify ready-to-use IPCs. IPCs were induced from cAD-MSCs with the modulated three-stepwise protocol. The first step of definitive endoderm (DE) induction showed that the cooperation of Chir99021 and Activin A created the effective production of Sox17-expressed DE cells. The second step for pancreatic endocrine (PE) progenitor induction from DE indicated that the treatment with taurine, retinoic acid, FGF2, EGF, TGFβ inhibitor, dorsomorphin, nicotinamide, and DAPT showed the significant upregulation of the pancreatic endocrine precursor markers Pdx1 and Ngn3. The last step of IPC production, the combination of taurine, nicotinamide, Glp-1, forskolin, PI3K inhibitor, and TGFβ inhibitor, yielded efficiently functional IPCs from PE precursors. Afterward, the maintenance of ALGPA-encapsulated cAD-MSC-derived IPCs with VSCBIC-1, a specialized medium, enhanced IPC properties. Conclusion, the modulated three-stepwise protocol generates the functional IPCs. Together, the encapsulation of cAD-MSC-derived IPCs and the cultivation with VSCBIC-1 enrich the maturation of generated IPCs.
Collapse
Affiliation(s)
- Quynh Dang Le
- International Program of Veterinary Science and Technology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
- Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Veterinary Pharmacology and Stem Cell Research Laboratory, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
- Veterinary Stem Cell and Bioengineering Research Unit, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Watchareewan Rodprasert
- Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Veterinary Pharmacology and Stem Cell Research Laboratory, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Suryo Kuncorojakti
- Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Veterinary Pharmacology and Stem Cell Research Laboratory, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
- Department of Veterinary Science, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya, East Java, Indonesia
| | - Prasit Pavasant
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Regenerative Dentistry (CERD), Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Thanaphum Osathanon
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
- Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Chenphop Sawangmake
- Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Veterinary Pharmacology and Stem Cell Research Laboratory, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand.
- Veterinary Stem Cell and Bioengineering Research Unit, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand.
- Department of Pharmacology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand.
- Center of Excellence in Regenerative Dentistry (CERD), Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand.
| |
Collapse
|
9
|
Differentiation of multipotent stem cells to insulin-producing cells for treatment of diabetes mellitus: bone marrow- and adipose tissue-derived cells comparison. Mol Biol Rep 2022; 49:3539-3548. [PMID: 35107740 DOI: 10.1007/s11033-022-07194-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 01/25/2022] [Indexed: 10/19/2022]
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) from human adipose tissue and bone marrow have a great potential for use in cell therapy due to their ease of isolation, expansion, and differentiation. Our intention was to isolate and promote in vitro expansion and differentiation of MSCs from human adipose and bone marrow tissue into cells with a pancreatic endocrine phenotype and to compare the potency of these cells together. METHODS AND RESULTS MSCs were pre-induced with nicotinamide, mercaptoethanol, B-27 and b-FGF in L-DMEM for 2 days and re-induced again in supplemented H-DMEM for another 3 days. Expression of five genes in differentiated beta cells was evaluated by Real-time PCR and western blotting and the potency of insulin release in response to glucose stimulation was evaluated by insulin and C-peptide ELISA kit. The differentiated cells were evaluated by immunocytochemistry staining for Insulin and PDX-1. Quantitative RT-PCR results showed up-regulation of four genes in differentiated beta-islet cells (Insulin, Ngn-3, Pax-4 and Pdx-1) compared with the control. Western blot analysis showed that MSCs cells mainly produced proinsulin and insulin after differentiation but nestin was more expressed in pre-differentiated stem cells. Glucose and insulin secretion assay showed that insulin levels and C-peptide secretion were significantly increased in response to 10 mM glucose. CONCLUSIONS Our study showed that both adipose and bone marrow stem cells could differentiate into functional beta-islet cells but it seems that adipose stem cells could be a better choice for treatment of diabetes mellitus according to their higher potency.
Collapse
|
10
|
Garrido-Utrilla A, Ayachi C, Friano ME, Atlija J, Balaji S, Napolitano T, Silvano S, Druelle N, Collombat P. Conversion of Gastrointestinal Somatostatin-Expressing D Cells Into Insulin-Producing Beta-Like Cells Upon Pax4 Misexpression. Front Endocrinol (Lausanne) 2022; 13:861922. [PMID: 35573999 PMCID: PMC9103212 DOI: 10.3389/fendo.2022.861922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 03/17/2022] [Indexed: 11/13/2022] Open
Abstract
Type 1 diabetes results from the autoimmune-mediated loss of insulin-producing beta-cells. Accordingly, important research efforts aim at regenerating these lost beta-cells by converting pre-existing endogenous cells. Following up on previous results demonstrating the conversion of pancreatic somatostatin delta-cells into beta-like cells upon Pax4 misexpression and acknowledging that somatostatin-expressing cells are highly represented in the gastrointestinal tract, one could wonder whether this Pax4-mediated conversion could also occur in the GI tract. We made use of transgenic mice misexpressing Pax4 in somatostatin cells (SSTCrePOE) to evaluate a putative Pax4-mediated D-to-beta-like cell conversion. Additionally, we implemented an ex vivo approach based on mice-derived gut organoids to assess the functionality of these neo-generated beta-like cells. Our results outlined the presence of insulin+ cells expressing several beta-cell markers in gastrointestinal tissues of SSTCrePOE animals. Further, using lineage tracing, we established that these cells arose from D cells. Lastly, functional tests on mice-derived gut organoids established the ability of neo-generated beta-like cells to release insulin upon stimulation. From this study, we conclude that the misexpression of Pax4 in D cells appears sufficient to convert these into functional beta-like cells, thus opening new research avenues in the context of diabetes research.
Collapse
Affiliation(s)
- Anna Garrido-Utrilla
- Université Côte d’Azur, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé Et de la Recherche Médicale (INSERM), Institut de Biologie Valrose (iBV), Nice, France
| | - Chaïma Ayachi
- Université Côte d’Azur, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé Et de la Recherche Médicale (INSERM), Institut de Biologie Valrose (iBV), Nice, France
| | - Marika Elsa Friano
- Université Côte d’Azur, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé Et de la Recherche Médicale (INSERM), Institut de Biologie Valrose (iBV), Nice, France
| | - Josipa Atlija
- Department of Cryopreservation, Distribution, Typing and Animal Archiving, Centre National de la Recherche Scientifique-Unité d'Appui à la Recherche (CNRS-UAR) 44 Typage et Archivage d’Animaux Modèles (TAAM), Orléans, France
| | - Shruti Balaji
- PlantaCorp Gesellschaft mit beschränkter Haftung (GmbH), Hamburg, Germany
| | - Tiziana Napolitano
- Université Côte d’Azur, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé Et de la Recherche Médicale (INSERM), Institut de Biologie Valrose (iBV), Nice, France
| | - Serena Silvano
- Université Côte d’Azur, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé Et de la Recherche Médicale (INSERM), Institut de Biologie Valrose (iBV), Nice, France
| | - Noémie Druelle
- Columbia University College of Physicians & Surgeons, Department of Medicine, New York, NY, United States
- *Correspondence: Noémie Druelle, ; Patrick Collombat,
| | - Patrick Collombat
- Université Côte d’Azur, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé Et de la Recherche Médicale (INSERM), Institut de Biologie Valrose (iBV), Nice, France
- *Correspondence: Noémie Druelle, ; Patrick Collombat,
| |
Collapse
|
11
|
Alvarez Fallas ME, Pedraza-Arevalo S, Cujba AM, Manea T, Lambert C, Morrugares R, Sancho R. Stem/progenitor cells in normal physiology and disease of the pancreas. Mol Cell Endocrinol 2021; 538:111459. [PMID: 34543699 PMCID: PMC8573583 DOI: 10.1016/j.mce.2021.111459] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 03/19/2021] [Accepted: 09/13/2021] [Indexed: 02/08/2023]
Abstract
Though embryonic pancreas progenitors are well characterised, the existence of stem/progenitor cells in the postnatal mammalian pancreas has been long debated, mainly due to contradicting results on regeneration after injury or disease in mice. Despite these controversies, sequencing advancements combined with lineage tracing and organoid technologies indicate that homeostatic and trigger-induced regenerative responses in mice could occur. The presence of putative progenitor cells in the adult pancreas has been proposed during homeostasis and upon different stress challenges such as inflammation, tissue damage and oncogenic stress. More recently, single cell transcriptomics has revealed a remarkable heterogeneity in all pancreas cell types, with some cells showing the signature of potential progenitors. In this review we provide an overview on embryonic and putative adult pancreas progenitors in homeostasis and disease, with special emphasis on in vitro culture systems and scRNA-seq technology as tools to address the progenitor nature of different pancreatic cells.
Collapse
Affiliation(s)
- Mario Enrique Alvarez Fallas
- Centre for Stem Cells and Regenerative Medicine, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Sergio Pedraza-Arevalo
- Centre for Stem Cells and Regenerative Medicine, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Ana-Maria Cujba
- Centre for Stem Cells and Regenerative Medicine, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Teodora Manea
- Centre for Stem Cells and Regenerative Medicine, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Christopher Lambert
- Centre for Stem Cells and Regenerative Medicine, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Rosario Morrugares
- Instituto Maimonides de Investigacion Biomedica de Cordoba (IMIBIC), Cordoba, Spain; Departamento de Biologia Celular, Fisiologia e Inmunologia, Universidad de Cordoba, Cordoba, Spain; Hospital Universitario Reina Sofia, Cordoba, Spain
| | - Rocio Sancho
- Centre for Stem Cells and Regenerative Medicine, Faculty of Life Sciences & Medicine, King's College London, London, UK; Department of Medicine III, University Hospital Carl Gustav Carus, Dresden, Germany.
| |
Collapse
|
12
|
Muthu S, Jeyaraman M, Jeyaraman N, Rajendran RL, Gangadaran P. Where Do We Stand in Stem Cell Therapy for the Management of Diabetes Mellitus?-A Scientometric Research Trend Analysis from 1990 to 2020. Bioengineering (Basel) 2021; 8:bioengineering8110159. [PMID: 34821725 PMCID: PMC8615097 DOI: 10.3390/bioengineering8110159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 10/19/2021] [Accepted: 10/24/2021] [Indexed: 02/05/2023] Open
Abstract
Stem cell therapy has been considered a promising strategy in the management of both type I and type II diabetes mellitus (DM) because of its immunomodulatory and regenerative capability to restore the beta cell number and function. Various modalities of cellular therapy like transplantation of pancreatic islet cells, transplantation of pancreatic ductal stem cells, and mesenchymal stromal cell transplantation have been tried, and the modality is undergoing rapid advancements that may become the reality in the near future. In the course of its evolution, it is essential to have a comprehensive summary of the progress for a greater capacity to refine our future directives. With technological developments like data mining, graphic drawing, and information analytics combined with computational statistics, visualization of scientific metrology has become a reality. With a newer perspective, we intend to use scientometric tools including text mining, co-word analysis, word frequency analysis, co-citation analysis, cluster network analysis, to perform a systematic and comprehensive analysis of the research trend in stem cell therapy in the management of DM over the past three decades (1990-2020) and to identify the future research hotspots.
Collapse
Affiliation(s)
- Sathish Muthu
- Department of Orthopaedics, Government Medical College and Hospital, Dindigul 624001, Tamil Nadu, India;
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida 201310, Uttar Pradesh, India
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow 226010, Uttar Pradesh, India;
| | - Madhan Jeyaraman
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida 201310, Uttar Pradesh, India
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow 226010, Uttar Pradesh, India;
- Department of Orthopedics, School of Medical Sciences and Research, Sharda University, Greater Noida 201310, Uttar Pradesh, India
- Correspondence: (M.J.); (P.G.); Tel.: +82-53-420-4914 (P.G.)
| | - Naveen Jeyaraman
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow 226010, Uttar Pradesh, India;
- Department of Orthopedics, School of Medical Sciences and Research, Sharda University, Greater Noida 201310, Uttar Pradesh, India
| | - Ramya Lakshmi Rajendran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea;
| | - Prakash Gangadaran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea;
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu 41944, Korea
- Correspondence: (M.J.); (P.G.); Tel.: +82-53-420-4914 (P.G.)
| |
Collapse
|
13
|
Bittenglova K, Habart D, Saudek F, Koblas T. The Potential of Pancreatic Organoids for Diabetes Research and Therapy. Islets 2021; 13:85-105. [PMID: 34523383 PMCID: PMC8528407 DOI: 10.1080/19382014.2021.1941555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 06/04/2021] [Indexed: 10/20/2022] Open
Abstract
The success of clinical transplantation of pancreas or isolated pancreatic islets supports the concept of cell-based cure for diabetes. One limitation is the shortage of cadaver human pancreata. The demand-supply gap could potentially be bridged by harnessing the self-renewal capacity of stem cells. Pluripotent stem cells and adult pancreatic stem cells have been explored as possible cell sources. Recently, a system for long-term culture of proposed adult pancreatic stem cells in a form of organoids was developed. Generated organoids partially mimic the architecture and cell-type composition of pancreatic tissue. Here, we review the attempts over the past decade, to utilize the organoid cell culture principles in order to identify, expand, and differentiate the adult pancreatic stem cells from different compartments of mouse and human pancreata. The development of the culture conditions, effects of specific growth factors and small molecules is discussed. The potential utility of the adult pancreatic stem cells is considered in the context of other cell sources.
Collapse
Affiliation(s)
- Katerina Bittenglova
- Department of Diabetes, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
- First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - David Habart
- Department of Diabetes, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Frantisek Saudek
- Department of Diabetes, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Tomas Koblas
- Department of Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| |
Collapse
|
14
|
Human umbilical cord mesenchymal stem cells in type 2 diabetes mellitus: the emerging therapeutic approach. Cell Tissue Res 2021; 385:497-518. [PMID: 34050823 DOI: 10.1007/s00441-021-03461-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 04/11/2021] [Indexed: 12/14/2022]
Abstract
The umbilical cord has been proved to be an easy-access, reliable, and useful source of mesenchymal stem cells (MSC) for clinical applications due to its primitive, immunomodulatory, non-immunogenic, secretory and paracrine, migratory, proliferative, and multipotent properties. This set of characteristics has recently attracted great research interest in the fields of nanotechnology and regenerative medicine and cellular therapy. Accumulating evidence supports a pronounced therapeutic potential of MSC in many different pathologies, from hematology to immunology, wound-healing, tissue regeneration, and oncology. Diabetes mellitus, branded the epidemic of the century, is considered a chronic metabolic disorder, representing a major burden for health system sustainability and an important public health challenge to modern societies. The available treatments for type 2 diabetes mellitus (T2DM) still rely mainly on combinations of oral antidiabetic agents with lifestyle and nutritional adjustments. Despite the continuous development of novel and better hypoglycemic drugs, their efficacy is limited in the installment and progression of silent T2DM complications. T2DM comorbidities and mortality rates still make it a serious, common, costly, and long-term manageable disease. Recently, experimental models, preclinical observations, and clinical studies have provided some insights and preliminary promising results using umbilical cord MSCs to treat and manage diabetes. This review focuses on the latest research and applications of human-derived umbilical cord MSC in the treatment and management of T2DM, exploring and systematizing the key effects of both umbilical cord MSC and its factor-rich secretome accordingly with the major complications associated to T2DM.
Collapse
|
15
|
Tootee A, Nikbin B, Ghahary A, Esfahani EN, Arjmand B, Aghayan H, Qorbani M, Larijani B. Immunopathology of Type 1 Diabetes and Immunomodulatory Effects of Stem Cells: A Narrative Review of the Literature. Endocr Metab Immune Disord Drug Targets 2021; 22:169-197. [PMID: 33538679 DOI: 10.2174/1871530321666210203212809] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 10/11/2020] [Accepted: 10/27/2020] [Indexed: 11/22/2022]
Abstract
Type 1 Diabetes (T1D) is a complex autoimmune disorder which occurs as a result of an intricate series of pathologic interactions between pancreatic β-cells and a wide range of components of both the innate and the adaptive immune systems. Stem-cell therapy, a recently-emerged potentially therapeutic option for curative treatment of diabetes, is demonstrated to cause significant alternations to both different immune cells such as macrophages, natural killer (NK) cells, dendritic cells, T cells, and B cells and non-cellular elements including serum cytokines and different components of the complement system. Although there exists overwhelming evidence indicating that the documented therapeutic effects of stem cells on patients with T1D is primarily due to their potential for immune regulation rather than pancreatic tissue regeneration, to date, the precise underlying mechanisms remain obscure. On the other hand, immune-mediated rejection of stem cells remains one of the main obstacles to regenerative medicine. Moreover, the consequences of efferocytosis of stem-cells by the recipients' lung-resident macrophages have recently emerged as a responsible mechanism for some immune-mediated therapeutic effects of stem-cells. This review focuses on the nature of the interactions amongst different compartments of the immune systems which are involved in the pathogenesis of T1D and provides explanation as to how stem cell-based interventions can influence immune system and maintain the physiologic equilibrium.
Collapse
Affiliation(s)
- Ali Tootee
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, . Iran
| | - Behrouz Nikbin
- Research Center of Molecular Immunology, Tehran University of Medical Sciences, Tehran, . Iran
| | - Aziz Ghahary
- British Columbia Professional Firefighters' Burn and Wound Healing Research Laboratory, Department of Surgery, Plastic Surgery, University of British Columbia, Vancouver, . Canada
| | - Ensieh Nasli Esfahani
- Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, . Iran
| | - Babak Arjmand
- Cell therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, . Iran
| | - Hamidreza Aghayan
- Cell therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, . Iran
| | - Mostafa Qorbani
- Non-communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, . Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, . Iran
| |
Collapse
|
16
|
Kh S, Haider KH. Stem Cells: A Renewable Source of Pancreatic β-Cells and Future for Diabetes Treatment. Stem Cells 2021. [DOI: 10.1007/978-3-030-77052-5_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
17
|
Abstract
Traumatic injuries are a leading cause of death and disability in both military and civilian populations. Given the complexity and diversity of traumatic injuries, novel and individualized treatment strategies are required to optimize outcomes. Cellular therapies have potential benefit for the treatment of acute or chronic injuries, and various cell-based pharmaceuticals are currently being tested in preclinical studies or in clinical trials. Cellular therapeutics may have the ability to complement existing therapies, especially in restoring organ function lost due to tissue disruption, prolonged hypoxia or inflammatory damage. In this article we highlight the current status and discuss future directions of cellular therapies for the treatment of traumatic injury. Both published research and ongoing clinical trials are discussed here.
Collapse
|
18
|
Ghoneim MA, Refaie AF, Elbassiouny BL, Gabr MM, Zakaria MM. From Mesenchymal Stromal/Stem Cells to Insulin-Producing Cells: Progress and Challenges. Stem Cell Rev Rep 2020; 16:1156-1172. [PMID: 32880857 PMCID: PMC7667138 DOI: 10.1007/s12015-020-10036-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mesenchymal stromal cells (MSCs) are an attractive option for cell therapy for type 1 diabetes mellitus (DM). These cells can be obtained from many sources, but bone marrow and adipose tissue are the most studied. MSCs have distinct advantages since they are nonteratogenic, nonimmunogenic and have immunomodulatory functions. Insulin-producing cells (IPCs) can be generated from MSCs by gene transfection, gene editing or directed differentiation. For directed differentiation, MSCs are usually cultured in a glucose-rich medium with various growth and activation factors. The resulting IPCs can control chemically-induced diabetes in immune-deficient mice. These findings are comparable to those obtained from pluripotent cells. PD-L1 and PD-L2 expression by MSCs is upregulated under inflammatory conditions. Immunomodulation occurs due to the interaction between these ligands and PD-1 receptors on T lymphocytes. If this function is maintained after differentiation, life-long immunosuppression or encapsulation could be avoided. In the clinical setting, two sites can be used for transplantation of IPCs: the subcutaneous tissue and the omentum. A 2-stage procedure is required for the former and a laparoscopic procedure for the latter. For either site, cells should be transplanted within a scaffold, preferably one from fibrin. Several questions remain unanswered. Will the transplanted cells be affected by the antibodies involved in the pathogenesis of type 1 DM? What is the functional longevity of these cells following their transplantation? These issues have to be addressed before clinical translation is attempted. Graphical Abstract Bone marrow MSCs are isolated from the long bone of SD rats. Then they are expanded and through directed differentiation insulin-producing cells are formed. The differentiated cells are loaded onto a collagen scaffold. If one-stage transplantation is planned, a drug delivery system must be incorporated to ensure immediate oxygenation, promote vascularization and provide some growth factors. Some mechanisms involved in the immunomodulatory function of MSCs. These are implemented either by cell to cell contact or by the release of soluble factors. Collectively, these pathways results in an increase in T-regulatory cells.
Collapse
|
19
|
Yang K, Lee M, Jones PA, Liu SS, Zhou A, Xu J, Sreekanth V, Wu JLY, Vo L, Lee EA, Pop R, Lee Y, Wagner BK, Melton DA, Choudhary A, Karp JM. A 3D culture platform enables development of zinc-binding prodrugs for targeted proliferation of β cells. SCIENCE ADVANCES 2020; 6:eabc3207. [PMID: 33208361 PMCID: PMC7673808 DOI: 10.1126/sciadv.abc3207] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 10/07/2020] [Indexed: 06/11/2023]
Abstract
Advances in treating β cell loss include islet replacement therapies or increasing cell proliferation rate in type 1 and type 2 diabetes, respectively. We propose developing multiple proliferation-inducing prodrugs that target high concentration of zinc ions in β cells. Unfortunately, typical two-dimensional (2D) cell cultures do not mimic in vivo conditions, displaying a markedly lowered zinc content, while 3D culture systems are laborious and expensive. Therefore, we developed the Disque Platform (DP)-a high-fidelity culture system where stem cell-derived β cells are reaggregated into thin, 3D discs within 2D 96-well plates. We validated the DP against standard 2D and 3D cultures and interrogated our zinc-activated prodrugs, which release their cargo upon zinc chelation-so preferentially in β cells. Through developing a reliable screening platform that bridges the advantages of 2D and 3D culture systems, we identified an effective hit that exhibits 2.4-fold increase in β cell proliferation compared to harmine.
Collapse
Affiliation(s)
- Kisuk Yang
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Center for Nanomedicine, Harvard Stem Cell Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139, USA
- Proteomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02142, USA
| | - Miseon Lee
- Chemical Biology and Therapeutics Science, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Peter Anthony Jones
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Center for Nanomedicine, Harvard Stem Cell Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139, USA
| | - Sophie S Liu
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Center for Nanomedicine, Harvard Stem Cell Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Angela Zhou
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Center for Nanomedicine, Harvard Stem Cell Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jun Xu
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Center for Nanomedicine, Harvard Stem Cell Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Vedagopuram Sreekanth
- Chemical Biology and Therapeutics Science, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Jamie L Y Wu
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Center for Nanomedicine, Harvard Stem Cell Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Lillian Vo
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Center for Nanomedicine, Harvard Stem Cell Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Eunjee A Lee
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Center for Nanomedicine, Harvard Stem Cell Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139, USA
| | - Ramona Pop
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Yuhan Lee
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Center for Nanomedicine, Harvard Stem Cell Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139, USA
| | - Bridget K Wagner
- Chemical Biology and Therapeutics Science, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Douglas A Melton
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Amit Choudhary
- Chemical Biology and Therapeutics Science, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
- Divisions of Renal Medicine and Engineering, Brigham and Women's Hospital, Boston, MA 02115, USA
- Chemical Biology Program, Harvard University, Cambridge, MA 02138, USA
| | - Jeffrey M Karp
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
- Center for Nanomedicine, Harvard Stem Cell Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139, USA
- Proteomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| |
Collapse
|
20
|
Xiang X, Xiang Y, Jin S, Wang Z, Xu Y, Su C, Shi Q, Chen C, Yu Q, Song C. The hypoglycemic effect of extract/fractions from Fuzhuan Brick-Tea in streptozotocin-induced diabetic mice and their active components characterized by LC-QTOF-MS/MS. J Food Sci 2020; 85:2933-2942. [PMID: 32794200 DOI: 10.1111/1750-3841.15373] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 05/22/2020] [Accepted: 06/18/2020] [Indexed: 01/07/2023]
Abstract
Fuzhuan Brick-Tea is a postfermented product with the hypoglycemic effect, which is prepared from the leaves of Camellia sinensis var. sinensis. However, the material basis associated with the hypoglycemic effect was not clear. The present research was designed to explore the hypoglycemic effect of extract/fractions from Fuzhuan Brick-Tea in streptozotocin-induced type II diabetic mice. Then an ultra-high pressure liquid chromatography along with quadrupole time of flight mass spectrometry was used to analyze the phytochemicals in Fuzhuan Brick-Tea fractions. As a result, the hypoglycemic and hypolipidemic effects were evidently observed from the serum biochemical indexes and liver pathological examination in type II diabetic mice. In addition, there were total of 20 major components including eight lysophosphatidylcholines (Lyso-PCs), five fatty acids, and seven novel theophylline derivatives tentatively identified in the active fraction from water extract. Therefore, these components were assumed to contribute partly to the hypoglycemic effect of Fuzhuan Brick-Tea. These findings also give the evidence that the Lyso-PCs, fatty acids, and novel theophylline derivatives in Fuzhuan Brick-Tea may provide benefits in ameliorating disorders of glucose and lipid metabolism. PRACTICAL APPLICATION: This study suggests that the Lyso-PCs, fatty acids, and novel theophylline derivatives in Fuzhuan Brick-Tea may provide benefits in ameliorating disorders of glucose and lipid metabolism. It can be taken as a beneficial diet additive or nutraceutical.
Collapse
Affiliation(s)
- Xingliang Xiang
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, China
| | - Yi Xiang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shuna Jin
- State Key Laboratory of Environmental Health School of Public Health Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhiqiang Wang
- Mogon Golden Fiower Tea Technology Co. Ltd., Changsha, Hunan, China
| | - Yiqiang Xu
- Mogon Golden Fiower Tea Technology Co. Ltd., Changsha, Hunan, China
| | - Chao Su
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, China
| | - Qingxin Shi
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, China
| | - Cheng Chen
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, China
| | - Qingsong Yu
- Department of Otolaryngology Union Hospital Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chengwu Song
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei, China
| |
Collapse
|
21
|
Jacob A, Southard S, Rust W. Cell Replacement Therapy for Insulin-Dependent Diabetes: Maintaining Islet Architecture and Distribution After Graft. CURRENT TRANSPLANTATION REPORTS 2020. [DOI: 10.1007/s40472-020-00281-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
22
|
Looking into dental pulp stem cells in the therapy of photoreceptors and retinal degenerative disorders. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2019; 203:111727. [PMID: 31862637 DOI: 10.1016/j.jphotobiol.2019.111727] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 11/13/2019] [Accepted: 12/02/2019] [Indexed: 01/09/2023]
Abstract
Blindness and vision impairment are caused by irremediable retinal degeneration in affected individuals worldwide. Cell therapy for a retinal replacement can potentially rescue their vision, specifically for those who lost the light sensing photoreceptors in the eye. As such, well-characterized retinal cells are required for the replacement purposes. Stem cell-based therapy in photoreceptor and retinal pigment epithelium transplantation is well received, however, the drawbacks of retinal transplantation is the limited clinical protocols development, insufficient number of transplanted cells for recovery, the selection of potential stem cell sources that can be differentiated into the target cells, and the ability of cells to migrate to the host tissue. Dental pulp stem cells (DPSC) belong to a subset of mesenchymal stem cells, and are recently being studied due to its high capability of differentiating into cells of the neuronal lineage. In this review, we look into the potential uses of DPSC in treating retinal degeneration, and also the current data supporting its application.
Collapse
|
23
|
Farooq T, Rehman K, Hameed A, Akash MSH. Stem Cell Therapy and Type 1 Diabetes Mellitus: Treatment Strategies and Future Perspectives. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1084:95-107. [PMID: 29896720 DOI: 10.1007/5584_2018_195] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Type 1 diabetes mellitus (T1DM) is classified as an autoimmune disease which progressively results in the depletion of insulin-secreting β-cells. Consequently, the insulin secretion stops leading to hyperglycemic situations within the body. Under severe conditions, it also causes multi-organ diabetes-associated dysfunctionalities notably hypercoagulability, neuropathy, nephropathy, retinopathy, and sometimes organ failures. The prevalence of this disease has been noticed about 3% that has highlighted the serious concerns for healthcare professionals around the globe. For the treatment of this disease, the cell therapy is considered as an important therapeutic approach for the replacement of damaged β-cells. However, the development of autoantibodies unfortunately reduces their effectiveness with the passage of time and finally with the recurrence of diabetes mellitus. The development of new techniques for extraction and transplantation of islets failed to support this approach due to the issues related to major surgery and lifelong dependence on immunosuppression. For T1DM, such cells are supposed to produce, store, and supply insulin to maintain glucose homeostasis. The urgent need of much-anticipated substitute for insulin-secreting β-cells directed the researchers to focus on stem cells (SCs) to produce insulin-secreting β-cells. For being more specific and targeted therapeutic approaches, SC-based strategies opened up the new horizons to cure T1DM. This cell-based therapy aimed to produce functional insulin-secreting β-cells to cure diabetes on forever basis. The intrinsic regenerative potential along with immunomodulatory abilities of SCs highlights the therapeutic potential of SC-based strategies. In this article, we have comprehensively highlighted the role of SCs to treat diabetes mellitus.
Collapse
Affiliation(s)
- Tahir Farooq
- Department of Applied Chemistry, Government College University, Faisalabad, Pakistan
| | - Kanwal Rehman
- Institute of Pharmacy, Physiology and Pharmacology, University of Agriculture, Faisalabad, Pakistan.
| | - Arruje Hameed
- Department of Biochemistry, Government College University, Faisalabad, Pakistan
| | | |
Collapse
|
24
|
Hoveizi E, Tavakol S, Shirian S, Sanamiri K. Electrospun Nanofibers for Diabetes: Tissue Engineering and Cell-Based Therapies. Curr Stem Cell Res Ther 2019; 14:152-168. [PMID: 30338744 DOI: 10.2174/1574888x13666181018150107] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 03/11/2018] [Accepted: 09/05/2018] [Indexed: 02/08/2023]
Abstract
Diabetes mellitus is an autoimmune disease which causes loss of insulin secretion producing hyperglycemia by promoting progressive destruction of pancreatic β cells. An ideal therapeutic approach to manage diabetes mellitus is pancreatic β cells replacement. The aim of this review article was to evaluate the role of nanofibrous scaffolds and stem cells in the treatment of diabetes mellitus. Various studies have pointed out that application of electrospun biomaterials has considerably attracted researchers in the field of tissue engineering. The principles of cell therapy for diabetes have been reviewed in the first part of this article, while the usability of tissue engineering as a new therapeutic approach is discussed in the second part.
Collapse
Affiliation(s)
- Elham Hoveizi
- Department of Biology, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran.,Stem Cells and Transgenic Technology Research Center (STTRC), Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Shima Tavakol
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Sadegh Shirian
- Department of Pathology, School of Veterinary Medicine, Shahrekord University, Shahrekord, Iran.,Shiraz Molecular Research Center, Dr. Daneshbod Pathology Lab, Shiraz, Iran
| | - Khadije Sanamiri
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| |
Collapse
|
25
|
Rashed S, Gabr M, Abdel-Aziz AA, Zakaria M, Khater S, Ismail A, Fouad A, Refaie A. Differentiation Potential of Nestin (+) and Nestin (-) Cells Derived from Human Bone Marrow Mesenchymal Stem Cells into Functional Insulin Producing Cells. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2019; 8:1-13. [PMID: 32195201 DOI: 10.22088/ijmcm.bums.8.1.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 07/13/2019] [Indexed: 01/09/2023]
Abstract
The feasibility of isolating and manipulating mesenchymal stem cells (MSCs) from human patients provides hope for curing numerous diseases and disorders. Recent phenotypic analysis has shown heterogeneity of MSCs. Nestin progenitor cell is a subpopulation within MSCs which plays a role in pancreas regeneration during embryogenesis. This study aimed to separate nestin (+) cells from human bone marrow MSCs, and differentiate these cells into functional insulin producing cells (IPCs) compared with nestin (-) cells. Manual magnetic separation was performed to obtain nestin (+) cells from MSCs. Approximately 91±3.3% of nestin (+) cells were positive for anti-nestin antibody. Pluripotent genes were overexpressed in nestin (+) cells compared with nestin (-) cells as revealed by quantitative real time-PCR (qRT-PCR). Following in vitro differentiation, flow cytometric analysis showed that 2.7±0.5% of differentiated nestin (+) cells were positive for anti-insulin antibody in comparison with 0.08±0.02% of nestin (-) cells. QRT-PCR showed higher expression of insulin and other endocrine genes in comparison with nestin (-) cells. While immunofluorescence technique showed the presence of insulin and C-peptide granules in nestin (+) cells. Therefore, our results introduced nestin (+) cells as a pluripotent subpopulation within human MSCs which is capable to differentiate and produce functional IPCs.
Collapse
Affiliation(s)
- Sahar Rashed
- Department of Biotechnology, Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| | - Mahmoud Gabr
- Department of Biotechnology, Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| | - Abdel-Aziz Abdel-Aziz
- Biochemistry Division, Chemistry Department, Faculty of Science, Mansoura University, Mansoura, Egypt
| | - Mahmoud Zakaria
- Department of Biotechnology, Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| | - Sherry Khater
- Department of Biotechnology, Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| | - Amani Ismail
- Department of Biotechnology, Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| | - Ali Fouad
- Department of Biotechnology, Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| | - Ayman Refaie
- Nephrology Department, Urology and Nephrology Center, Mansoura University, Mansoura, Egypt
| |
Collapse
|
26
|
NRG1 type I dependent autoparacrine stimulation of Schwann cells in onion bulbs of peripheral neuropathies. Nat Commun 2019; 10:1467. [PMID: 30931926 PMCID: PMC6443727 DOI: 10.1038/s41467-019-09385-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Accepted: 03/07/2019] [Indexed: 01/28/2023] Open
Abstract
In contrast to acute peripheral nerve injury, the molecular response of Schwann cells in chronic neuropathies remains poorly understood. Onion bulb structures are a pathological hallmark of demyelinating neuropathies, but the nature of these formations is unknown. Here, we show that Schwann cells induce the expression of Neuregulin-1 type I (NRG1-I), a paracrine growth factor, in various chronic demyelinating diseases. Genetic disruption of Schwann cell-derived NRG1 signalling in a mouse model of Charcot-Marie-Tooth Disease 1A (CMT1A), suppresses hypermyelination and the formation of onion bulbs. Transgenic overexpression of NRG1-I in Schwann cells on a wildtype background is sufficient to mediate an interaction between Schwann cells via an ErbB2 receptor-MEK/ERK signaling axis, which causes onion bulb formations and results in a peripheral neuropathy reminiscent of CMT1A. We suggest that diseased Schwann cells mount a regeneration program that is beneficial in acute nerve injury, but that overstimulation of Schwann cells in chronic neuropathies is detrimental. Onion bulbs are a hallmark of demyelinating peripheral neuropathies. Here the authors identify Neuregulin-1 type I expression in Schwann cells as an essential mechanism involved in the formation of these characteristic structures.
Collapse
|
27
|
Rajab TK, Tchantchaleishvili V. Can tissue engineering produce bioartificial organs for transplantation? Artif Organs 2019; 43:536-541. [PMID: 30891801 DOI: 10.1111/aor.13443] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 02/22/2019] [Indexed: 12/21/2022]
Affiliation(s)
- Taufiek Konrad Rajab
- Division of Cardiac Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | | |
Collapse
|
28
|
Navigating Two Roads to Glucose Normalization in Diabetes: Automated Insulin Delivery Devices and Cell Therapy. Cell Metab 2019; 29:545-563. [PMID: 30840911 DOI: 10.1016/j.cmet.2019.02.007] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 02/12/2019] [Accepted: 02/13/2019] [Indexed: 12/23/2022]
Abstract
Incredible strides have been made since the discovery of insulin almost 100 years ago. Insulin formulations have improved dramatically, glucose levels can be measured continuously, and recently first-generation biomechanical "artificial pancreas" systems have been approved by regulators around the globe. However, still only a small fraction of patients with diabetes achieve glycemic goals. Replacement of insulin-producing cells via transplantation shows significant promise, but is limited in application due to supply constraints (cadaver-based) and the need for chronic immunosuppression. Over the past decade, significant progress has been made to address these barriers to widespread implementation of a cell therapy. Can glucose levels in people with diabetes be normalized with artificial pancreas systems or via cell replacement approaches? Here we review the road ahead, including the challenges and opportunities of both approaches.
Collapse
|
29
|
Beikmohammadi L, Bandehpour M, Hashemi SM, Kazemi B. Generation of insulin-producing hepatocyte-like cells from human Wharton's jelly mesenchymal stem cells as an alternative source of islet cells. J Cell Physiol 2019; 234:17326-17336. [PMID: 30790280 DOI: 10.1002/jcp.28352] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 01/24/2019] [Accepted: 01/28/2019] [Indexed: 12/11/2022]
Abstract
Islet cell transplantation, as a treatment of type 1 diabetes, has a lot of complexity such as allograft rejections and an insufficient number of donors. The liver can be used as a replacement for endogenous insulin production. Hepatocytes can inherently respond to glucose levels and secrete proteins. Utilization of mesenchymal stem cells for curing diabetes represents a major focus of recent investigations. As a new choice for transplantation, we have proposed glucose-regulated insulin-producing hepatocyte-like cells, which produce insulin dependent on glucose levels. We have transfected human Wharton's jelly mesenchymal stem cells with the special construct, which included homology arms and glucose-responsive elements upstream of the minimum liver-type pyruvate kinase promoter-directed insulin gene. Then, we have differentiated these transfected cells to hepatocyte-like cells by using serial exposure of different inducing material and exogenous growth factors. Immunofluorescence analyses have demonstrated the expression of albumin, cytokeratin-18, Hep-Par1, α-fetoprotein, and insulin. The expression of hepatocyte marker genes in the differentiated cells was confirmed by reverse-transcription polymerase chain reaction. Interestingly, flow cytometry results showed that approximately 60% of the insulin-producing hepatocyte-like cells were simultaneously cytochrome P450 3A4 (CYP3A4) and insulin positive. CYP3A4 is a significant enzyme found in mature liver tissue. This confirmed that the differentiation and the transfection procedures were done correctly. They were functionally active by releasing insulin in response to elevated glucose concentrations in vitro. These applicable cells could be used in the liver for cell therapy of diabetes.
Collapse
Affiliation(s)
- Leila Beikmohammadi
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mojgan Bandehpour
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Mahmoud Hashemi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Tissue Engineering and Applied Cell Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bahram Kazemi
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
30
|
Horie S, Gonzalez HE, Laffey JG, Masterson CH. Cell therapy in acute respiratory distress syndrome. J Thorac Dis 2018; 10:5607-5620. [PMID: 30416812 DOI: 10.21037/jtd.2018.08.28] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Acute respiratory distress syndrome (ARDS) is driven by a severe pro-inflammatory response resulting in lung damage, impaired gas exchange and severe respiratory failure. There is no specific treatment that effectively improves outcome in ARDS. However, in recent years, cell therapy has shown great promise in preclinical ARDS studies. A wide range of cells have been identified as potential candidates for use, among these are mesenchymal stromal cells (MSCs), which are adult multi-lineage cells that can modulate the immune response and enhance repair of damaged tissue. The therapeutic potential of MSC therapy for sepsis and ARDS has been demonstrated in multiple in vivo models. The therapeutic effect of these cells seems to be due to two different mechanisms; direct cellular interaction, and paracrine release of different soluble products such as extracellular vesicles (EVs)/exosomes. Different approaches have also been studied to enhance the therapeutic effect of these cells, such as the over-expression of anti-inflammatory or pro-reparative molecules. Several clinical trials (phase I and II) have already shown safety of MSCs in ARDS and other diseases. However, several translational issues still need to be addressed, such as the large-scale production of cells, and their potentiality and variability, before the therapeutic potential of stem cells therapies can be realized.
Collapse
Affiliation(s)
- Shahd Horie
- Regenerative Medicine Institute (REMEDI), CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, National University of Ireland Galway, Galway, Ireland
| | - Hector Esteban Gonzalez
- Regenerative Medicine Institute (REMEDI), CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, National University of Ireland Galway, Galway, Ireland
| | - John G Laffey
- Regenerative Medicine Institute (REMEDI), CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, National University of Ireland Galway, Galway, Ireland.,Department of Anesthesia and Intensive Care Medicine, Galway University Hospitals, SAOLTA Hospital Group, Ireland
| | - Claire H Masterson
- Regenerative Medicine Institute (REMEDI), CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, National University of Ireland Galway, Galway, Ireland
| |
Collapse
|
31
|
Peloso A, Citro A, Zoro T, Cobianchi L, Kahler-Quesada A, Bianchi CM, Andres A, Berishvili E, Piemonti L, Berney T, Toso C, Oldani G. Regenerative Medicine and Diabetes: Targeting the Extracellular Matrix Beyond the Stem Cell Approach and Encapsulation Technology. Front Endocrinol (Lausanne) 2018; 9:445. [PMID: 30233489 PMCID: PMC6127205 DOI: 10.3389/fendo.2018.00445] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 07/18/2018] [Indexed: 12/20/2022] Open
Abstract
According to the Juvenile Diabetes Research Foundation (JDRF), almost 1. 25 million people in the United States (US) have type 1 diabetes, which makes them dependent on insulin injections. Nationwide, type 2 diabetes rates have nearly doubled in the past 20 years resulting in more than 29 million American adults with diabetes and another 86 million in a pre-diabetic state. The International Diabetes Ferderation (IDF) has estimated that there will be almost 650 million adult diabetic patients worldwide at the end of the next 20 years (excluding patients over the age of 80). At this time, pancreas transplantation is the only available cure for selected patients, but it is offered only to a small percentage of them due to organ shortage and the risks linked to immunosuppressive regimes. Currently, exogenous insulin therapy is still considered to be the gold standard when managing diabetes, though stem cell biology is recognized as one of the most promising strategies for restoring endocrine pancreatic function. However, many issues remain to be solved, and there are currently no recognized treatments for diabetes based on stem cells. In addition to stem cell resesarch, several β-cell substitutive therapies have been explored in the recent era, including the use of acellular extracellular matrix scaffolding as a template for cellular seeding, thus providing an empty template to be repopulated with β-cells. Although this bioengineering approach still has to overcome important hurdles in regards to clinical application (including the origin of insulin producing cells as well as immune-related limitations), it could theoretically provide an inexhaustible source of bio-engineered pancreases.
Collapse
Affiliation(s)
- Andrea Peloso
- Division of Abdominal Surgery, Department of Surgery, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- HepatoPancreato-Biliary Centre, Geneva University Hospitals, Geneva, Switzerland
| | - Antonio Citro
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Tamara Zoro
- Department of General Surgery, IRCCS Policlinico San Matteo, Pavia, Italy
- Department of Clinical, Surgical, Diagnostic and Paediatric Sciences, University of Pavia, Pavia, Italy
| | - Lorenzo Cobianchi
- Department of General Surgery, IRCCS Policlinico San Matteo, Pavia, Italy
- Department of Clinical, Surgical, Diagnostic and Paediatric Sciences, University of Pavia, Pavia, Italy
| | - Arianna Kahler-Quesada
- Division of Abdominal Surgery, Department of Surgery, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Carlo M. Bianchi
- Department of General Surgery, IRCCS Policlinico San Matteo, Pavia, Italy
- Department of Clinical, Surgical, Diagnostic and Paediatric Sciences, University of Pavia, Pavia, Italy
| | - Axel Andres
- Division of Abdominal Surgery, Department of Surgery, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- HepatoPancreato-Biliary Centre, Geneva University Hospitals, Geneva, Switzerland
| | - Ekaterine Berishvili
- Cell Isolation and Transplantation Center, University of Geneva, Geneva, Switzerland
- Institute of Medical Research, Ilia State University, Tbilisi, Georgia
| | - Lorenzo Piemonti
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Thierry Berney
- Cell Isolation and Transplantation Center, University of Geneva, Geneva, Switzerland
| | - Christian Toso
- Division of Abdominal Surgery, Department of Surgery, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- HepatoPancreato-Biliary Centre, Geneva University Hospitals, Geneva, Switzerland
| | - Graziano Oldani
- Division of Abdominal Surgery, Department of Surgery, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- HepatoPancreato-Biliary Centre, Geneva University Hospitals, Geneva, Switzerland
| |
Collapse
|
32
|
Elham H, Mahmoud H. The Effect of Pancreas Islet-Releasing Factors on the Direction of Embryonic Stem Cells Towards Pdx1 Expressing Cells. Appl Biochem Biotechnol 2018; 186:371-383. [DOI: 10.1007/s12010-018-2733-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Accepted: 03/12/2018] [Indexed: 11/24/2022]
|
33
|
Salguero-Aranda C, Tapia-Limonchi R, Cahuana GM, Hitos AB, Diaz I, Hmadcha A, Fraga M, Martín F, Soria B, Tejedo JR, Bedoya FJ. Differentiation of Mouse Embryonic Stem Cells toward Functional Pancreatic β-Cell Surrogates through Epigenetic Regulation of Pdx1 by Nitric Oxide. Cell Transplant 2018; 25:1879-1892. [PMID: 26980118 DOI: 10.3727/096368916x691178] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Pancreatic and duodenal homeobox 1 (Pdx1) is a transcription factor that regulates the embryonic development of the pancreas and the differentiation toward β cells. Previously, we have shown that exposure of mouse embryonic stem cells (mESCs) to high concentrations of diethylenetriamine nitric oxide adduct (DETA-NO) triggers differentiation events and promotes the expression of Pdx1. Here we report evidence that Pdx1 expression is associated with release of polycomb repressive complex 2 (PRC2) and P300 from its promoter region. These events are accompanied by epigenetic changes in bivalent markers of histones trimethylated histone H3 lysine 27 (H3K27me3) and H3K4me3, site-specific changes in DNA methylation, and no change in H3 acetylation. On the basis of these findings, we developed a protocol to differentiate mESCs toward insulin-producing cells consisting of sequential exposure to DETA-NO, valproic acid, and P300 inhibitor (C646) to enhance Pdx1 expression and a final maturation step of culture in suspension to form cell aggregates. This small molecule-based protocol succeeds in obtaining cells that express pancreatic β-cell markers such as PDX1, INS1, GCK, and GLUT2 and respond in vitro to high glucose and KCl.
Collapse
Affiliation(s)
- Carmen Salguero-Aranda
- Andalusian Center for Molecular Biology and Regenerative Medicine, Seville, Spain.,Center for Network Research in Diabetes and Metabolic Diseases (CIBERDEM) Instituto de Salud Carlos III, Madrid, Spain.,Cell Therapy Network, Madrid (RED-TERCEL), Instituto de Salud Carlos III, Madrid, Spain.,Fundación Progreso y Salud, Seville, Spain
| | - Rafael Tapia-Limonchi
- Andalusian Center for Molecular Biology and Regenerative Medicine, Seville, Spain.,Pablo de Olavide University, Seville, Spain
| | - Gladys Margot Cahuana
- Andalusian Center for Molecular Biology and Regenerative Medicine, Seville, Spain.,Pablo de Olavide University, Seville, Spain
| | - Ana Belen Hitos
- Andalusian Center for Molecular Biology and Regenerative Medicine, Seville, Spain.,Center for Network Research in Diabetes and Metabolic Diseases (CIBERDEM) Instituto de Salud Carlos III, Madrid, Spain
| | - Irene Diaz
- Andalusian Center for Molecular Biology and Regenerative Medicine, Seville, Spain.,Center for Network Research in Diabetes and Metabolic Diseases (CIBERDEM) Instituto de Salud Carlos III, Madrid, Spain
| | - Abdelkrim Hmadcha
- Andalusian Center for Molecular Biology and Regenerative Medicine, Seville, Spain.,Cell Therapy Network, Madrid (RED-TERCEL), Instituto de Salud Carlos III, Madrid, Spain.,Fundación Progreso y Salud, Seville, Spain
| | - Mario Fraga
- Department of Epigenetics, Oncologic Institute of Principado of Asturias, Oviedo, Spain
| | - Franz Martín
- Andalusian Center for Molecular Biology and Regenerative Medicine, Seville, Spain.,Center for Network Research in Diabetes and Metabolic Diseases (CIBERDEM) Instituto de Salud Carlos III, Madrid, Spain.,Pablo de Olavide University, Seville, Spain
| | - Bernat Soria
- Andalusian Center for Molecular Biology and Regenerative Medicine, Seville, Spain.,Center for Network Research in Diabetes and Metabolic Diseases (CIBERDEM) Instituto de Salud Carlos III, Madrid, Spain.,Cell Therapy Network, Madrid (RED-TERCEL), Instituto de Salud Carlos III, Madrid, Spain.,Fundación Progreso y Salud, Seville, Spain
| | - Juan Rigoberto Tejedo
- Andalusian Center for Molecular Biology and Regenerative Medicine, Seville, Spain.,Center for Network Research in Diabetes and Metabolic Diseases (CIBERDEM) Instituto de Salud Carlos III, Madrid, Spain.,Cell Therapy Network, Madrid (RED-TERCEL), Instituto de Salud Carlos III, Madrid, Spain.,Pablo de Olavide University, Seville, Spain
| | - Francisco Javier Bedoya
- Andalusian Center for Molecular Biology and Regenerative Medicine, Seville, Spain.,Center for Network Research in Diabetes and Metabolic Diseases (CIBERDEM) Instituto de Salud Carlos III, Madrid, Spain.,Cell Therapy Network, Madrid (RED-TERCEL), Instituto de Salud Carlos III, Madrid, Spain.,Pablo de Olavide University, Seville, Spain
| |
Collapse
|
34
|
Significance of dopamine D 1 receptor signalling for steroidogenic differentiation of human induced pluripotent stem cells. Sci Rep 2017; 7:15120. [PMID: 29123220 PMCID: PMC5680317 DOI: 10.1038/s41598-017-15485-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 10/27/2017] [Indexed: 12/18/2022] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) are expected to be both a revolutionary cell source for regenerative medicine and a powerful tool to investigate the molecular mechanisms underlying human cell development in vitro. In the present study, we tried to elucidate the steroidogenic differentiation processes using hiPSC-derived intermediate mesoderm (IM) that is known to be the origin of the human adrenal cortex and gonads. We first performed chemical screening to identify small molecules that induce steroidogenic differentiation of IM cells expressing Odd-skipped related 1 (OSR1), an early IM marker. We identified cabergoline as an inducer of 3β-hydroxysteroid dehydrogenase, an essential enzyme for adrenogonadal steroidogenesis. Although cabergoline is a potent dopamine D2 receptor agonist, additional experiments showed that cabergoline exerted effects as a low-affinity agonist of D1 receptors by increasing intracellular cyclic AMP. Further analysis of OSR1+ cells transfected with steroidogenic factor-1/adrenal 4 binding protein revealed that D1 receptor agonist upregulated expression of various steroidogenic enzymes and increased secretion of steroid hormones synergistically with adrenocorticotropic hormone. These results suggest the importance of dopamine D1 receptor signalling in steroidogenic differentiation, which contributes to effective induction of steroidogenic cells from hiPSCs.
Collapse
|
35
|
Donadel G, Pastore D, Della-Morte D, Capuani B, Lombardo MF, Pacifici F, Bugliani M, Grieco FA, Marchetti P, Lauro D. FGF-2b and h-PL Transform Duct and Non-Endocrine Human Pancreatic Cells into Endocrine Insulin Secreting Cells by Modulating Differentiating Genes. Int J Mol Sci 2017; 18:ijms18112234. [PMID: 29068419 PMCID: PMC5713204 DOI: 10.3390/ijms18112234] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 10/13/2017] [Accepted: 10/16/2017] [Indexed: 12/12/2022] Open
Abstract
Background: Diabetes mellitus (DM) is a multifactorial disease orphan of a cure. Regenerative medicine has been proposed as novel strategy for DM therapy. Human fibroblast growth factor (FGF)-2b controls β-cell clusters via autocrine action, and human placental lactogen (hPL)-A increases functional β-cells. We hypothesized whether FGF-2b/hPL-A treatment induces β-cell differentiation from ductal/non-endocrine precursor(s) by modulating specific genes expression. Methods: Human pancreatic ductal-cells (PANC-1) and non-endocrine pancreatic cells were treated with FGF-2b plus hPL-A at 500 ng/mL. Cytofluorimetry and Immunofluorescence have been performed to detect expression of endocrine, ductal and acinar markers. Bromodeoxyuridine incorporation and annexin-V quantified cells proliferation and apoptosis. Insulin secretion was assessed by RIA kit, and electron microscopy analyzed islet-like clusters. Results: Increase in PANC-1 duct cells de-differentiation into islet-like aggregates was observed after FGF-2b/hPL-A treatment showing ultrastructure typical of islets-aggregates. These clusters, after stimulation with FGF-2b/hPL-A, had significant (p < 0.05) increase in insulin, C-peptide, pancreatic and duodenal homeobox 1 (PDX-1), Nkx2.2, Nkx6.1, somatostatin, glucagon, and glucose transporter 2 (Glut-2), compared with control cells. Markers of PANC-1 (Cytokeratin-19, MUC-1, CA19-9) were decreased (p < 0.05). These aggregates after treatment with FGF-2b/hPL-A significantly reduced levels of apoptosis. Conclusions: FGF-2b and hPL-A are promising candidates for regenerative therapy in DM by inducing de-differentiation of stem cells modulating pivotal endocrine genes.
Collapse
Affiliation(s)
- Giulia Donadel
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy.
| | - Donatella Pastore
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy.
| | - David Della-Morte
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy.
- Department of Human Sciences and Quality of Life Promotion, San Raffaele Roma Open University, 00166 Rome, Italy.
| | - Barbara Capuani
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy.
| | - Marco F Lombardo
- Agenzia regionale per la protezione ambientale (ARPA) Lazio, Sezione di Roma, 00173 Rome, Italy.
| | - Francesca Pacifici
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy.
| | - Marco Bugliani
- Endocrinology and Metabolism of Transplantation, Azienda Ospedaliero-Universitaria (A.O.U.) Pisana, 56126 Pisa, Italy.
| | - Fabio A Grieco
- Department of Medicine, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy.
| | - Piero Marchetti
- Endocrinology and Metabolism of Transplantation, Azienda Ospedaliero-Universitaria (A.O.U.) Pisana, 56126 Pisa, Italy.
| | - Davide Lauro
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy.
| |
Collapse
|
36
|
Pluripotent Stem Cells and Other Innovative Strategies for the Treatment of Ocular Surface Diseases. Stem Cell Rev Rep 2017; 12:171-8. [PMID: 26779895 DOI: 10.1007/s12015-016-9643-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The cornea provides two thirds of the refractive power of the eye and protection against insults such as infection and injury. The outermost tissue of the cornea is renewed by stem cells located in the limbus. Depletion or destruction of these stem cells may lead to blinding limbal stem cell deficiency (LSCD) that concerns millions of patients around the world. Innovative strategies based on adult stem cell therapies have been developed in the recent years but they are still facing numerous unresolved issues, and the long term results can be deceiving. Today there is a clear need to improve these therapies, and/or to develop new approaches for the treatment of LSCD. Here, we review the current cell-based therapies used for the treatment of ocular diseases, and discuss the potential of pluripotent stem cells (embryonic and induced pluripotent stem cells) in corneal repair. As the secretion of paracrine factors is known to have a crucial role in maintaining stem cell homeostasis and in wound repair, we also consider the therapeutic potential of a promising novel pathway, the exosomes. Exosomes are nano-sized vesicles that have the ability to transfer RNAs and proteins to recipient cells, and several studies demonstrated their role in cell protection and wound healing. Exosomes could circumvent the hurdles of stem-cell based approaches, and they could become a strong candidate as an alternative therapy for ocular surface diseases.
Collapse
|
37
|
Espes D, Lau J, Carlsson PO. MECHANISMS IN ENDOCRINOLOGY: Towards the clinical translation of stem cell therapy for type 1 diabetes. Eur J Endocrinol 2017; 177:R159-R168. [PMID: 28487297 DOI: 10.1530/eje-17-0080] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 04/24/2017] [Accepted: 05/08/2017] [Indexed: 01/21/2023]
Abstract
Insulin-producing cells derived from human embryonic stem cells (hESCs) or induced pluripotent stem cells (iPSCs) have for long been a promising, but elusive treatment far from clinical translation into type 1 diabetes therapy. However, the field is now on the verge of moving such insulin-producing cells into clinical trials. Although stem cell therapies provide great opportunities, there are also potential risks such as teratoma formation associated with the treatment. Many considerations are needed on how to proceed with clinical translation, including whether to use hESCs or iPSCs, and whether encapsulation of tissue will be needed. This review aims to give an overview of the current knowledge of stem cell therapy outcomes in animal models of type 1 diabetes and a proposed road map towards the clinical setting with special focus on the potential risks and hurdles which needs to be considered. From a clinical point of view, transplantation of insulin-producing cells derived from stem cells must be performed without immune suppression in order to be an attractive treatment option. Although costly and highly labour intensive, patient-derived iPSCs would be the only solution, if not clinically successful encapsulation or tolerance induction protocols are introduced.
Collapse
Affiliation(s)
- Daniel Espes
- Departments of Medical Cell Biology
- Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Joey Lau
- Departments of Medical Cell Biology
| | - Per-Ola Carlsson
- Departments of Medical Cell Biology
- Medical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
38
|
Reduced MLH3 Expression in the Syndrome of Gan-Shen Yin Deficiency in Patients with Different Diseases. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 2017:4109828. [PMID: 29234393 PMCID: PMC5634572 DOI: 10.1155/2017/4109828] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Revised: 03/27/2017] [Accepted: 04/03/2017] [Indexed: 01/12/2023]
Abstract
Traditional Chinese medicine formulates treatment according to body constitution (BC) differentiation. Different constitutions have specific metabolic characteristics and different susceptibility to certain diseases. This study aimed to assess the characteristic genes of gan-shen Yin deficiency constitution in different diseases. Fifty primary liver cancer (PLC) patients, 94 hypertension (HBP) patients, and 100 diabetes mellitus (DM) patients were enrolled and classified into gan-shen Yin deficiency group and non-gan-shen Yin deficiency group according to the body constitution questionnaire to assess the clinical manifestation of patients. The mRNA expressions of 17 genes in PLC patients with gan-shen Yin deficiency were different from those without gan-shen Yin deficiency. However, considering all patients with PLC, HBP, and DM, only MLH3 was significantly lower in gan-shen Yin deficiency group than that in non-gen-shen Yin deficiency. By ROC analysis, the relationship between MLH3 and gan-shen Yin deficiency constitution was confirmed. Treatment of MLH3 (-/- and -/+) mice with Liuweidihuang wan, classical prescriptions for Yin deficiency, partly ameliorates the body constitution of Yin deficiency in MLH3 (-/+) mice, but not in MLH3 (-/-) mice. MLH3 might be one of material bases of gan-shen Yin deficiency constitution.
Collapse
|
39
|
Harb G, Poh YC, Pagliuca F. Stem Cell-Derived Insulin-Producing β Cells to Treat Diabetes. CURRENT TRANSPLANTATION REPORTS 2017. [DOI: 10.1007/s40472-017-0161-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
40
|
Tsuchiya M, Tsuchiya K, Ohgawara H. Molecular Cloning of the Porcine Insulin cDNA Using a Monolayer Culture of Pancreatic Endocrine Cells. Cell Transplant 2017. [DOI: 10.3727/000000001783986611] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Porcine pancreatic endocrine cells are an attractive candidate for islet cell transplantation in view of the immunological properties and structural similarities of porcine insulin to human insulin. We recently established a method of isolation and a primary monolayer culture of porcine pancreatic endocrine cells. In this study, cloning of the porcine insulin cDNA was performed to clarify the genetic background of the purified isolated cells. A homology-based PCR cloning method was employed to determine the sequence using mRNA extracted from the monolayer-forming cells, and the candidate products were then determined by a homology search on the human insulin cDNA. According to the newly identified sequence, rapid amplification of cDNA ends was applied to the 5′ and 3′ ends, and the entire cDNA sequence was determined. Gene and protein expression was confirmed by Northern blotting, immunohistochemistry, and enzyme assay. To examine the transcriptional level, the cultured cells were incubated in a 20 mM D-glucose medium in the presence or absence of 5 μM forskolin. The porcine insulin cDNA exhibited a high homology to the human cDNA and showed 85% matching with the human amino acid sequence. D-Glucose at 20 mM stimulated the insulin secretion and mRNA expression, and further addition of 5 μM forskolin with the glucose was applied as the strongest stimulus in this culture system.
Collapse
Affiliation(s)
- Mariko Tsuchiya
- Institute of Geriatrics, Aoyama Hospital, Tokyo Women's Medical University, Tokyo, Japan
| | - Ken Tsuchiya
- Department of Medicine IV, Tokyo Women's Medical University, Tokyo, Japan
| | - Hisako Ohgawara
- Medical Research Institute, Tokyo Women's Medical University, Tokyo, Japan
| |
Collapse
|
41
|
Jacobson EF, Tzanakakis ES. Human pluripotent stem cell differentiation to functional pancreatic cells for diabetes therapies: Innovations, challenges and future directions. J Biol Eng 2017; 11:21. [PMID: 28680477 PMCID: PMC5494890 DOI: 10.1186/s13036-017-0066-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 06/02/2017] [Indexed: 12/18/2022] Open
Abstract
Recent advances in the expansion and directed pancreatogenic differentiation of human pluripotent stem cells (hPSCs) have intensified efforts to generate functional pancreatic islet cells, especially insulin-secreting β-cells, for cell therapies against diabetes. However, the consistent generation of glucose-responsive insulin-releasing cells remains challenging. In this article, we first present basic concepts of pancreatic organogenesis, which frequently serves as a basis for engineering differentiation regimens. Next, past and current efforts are critically discussed for the conversion of hPSCs along pancreatic cell lineages, including endocrine β-cells and α-cells, as well as exocrine cells with emphasis placed on the later stages of commitment. Finally, major challenges and future directions are examined, such as the identification of factors for in vivo maturation, large-scale culture and post processing systems, cell loss during differentiation, culture economics, efficiency, and efficacy and exosomes and miRNAs in pancreatic differentiation.
Collapse
Affiliation(s)
- Elena F Jacobson
- Department of Chemical and Biological Engineering, Tufts University, 4 Colby St., Room 276A, Medford, MA 02155 USA
| | - Emmanuel S Tzanakakis
- Department of Chemical and Biological Engineering, Tufts University, 4 Colby St., Room 276A, Medford, MA 02155 USA.,Tufts Clinical and Translational Science Institute, Tufts Medical Center, Boston, MA 02111 USA
| |
Collapse
|
42
|
Omami M, McGarrigle JJ, Reedy M, Isa D, Ghani S, Marchese E, Bochenek MA, Longi M, Xing Y, Joshi I, Wang Y, Oberholzer J. Islet Microencapsulation: Strategies and Clinical Status in Diabetes. Curr Diab Rep 2017; 17:47. [PMID: 28523592 DOI: 10.1007/s11892-017-0877-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE OF REVIEW Type 1 diabetes mellitus (T1DM) is an autoimmune disease that results from the destruction of insulin-producing pancreatic β cells in the islets of Langerhans. Islet cell transplantation has become a successful therapy for specific patients with T1DM with hypoglycemic unawareness. The reversal of T1DM by islet transplantation is now performed at many major medical facilities throughout the world. However, many challenges must still be overcome in order to achieve continuous, long-term successful transplant outcomes. Two major obstacles to this therapy are a lack of islet cells for transplantation and the need for life-long immunosuppressive treatment. Microencapsulation is seen as a technology that can overcome both these limitations of islet cell transplantation. This review depicts the present state of microencapsulated islet transplantation. RECENT FINDINGS Microencapsulation can play a significant role in overcoming the need for immunosuppression and lack of donor islet cells. This review focuses on microencapsulation and the clinical status of the technology in combating T1DM.
Collapse
Affiliation(s)
- Mustafa Omami
- Department of Surgery, University of Illinois at Chicago, 840 S. Wood St. Room 502, Chicago, IL, 60612, USA
| | - James J McGarrigle
- Department of Surgery, University of Illinois at Chicago, 840 S. Wood St. Room 502, Chicago, IL, 60612, USA.
| | - Mick Reedy
- Department of Surgery, University of Illinois at Chicago, 840 S. Wood St. Room 502, Chicago, IL, 60612, USA
| | - Douglas Isa
- Department of Surgery, University of Illinois at Chicago, 840 S. Wood St. Room 502, Chicago, IL, 60612, USA
| | - Sofia Ghani
- Department of Surgery, University of Illinois at Chicago, 840 S. Wood St. Room 502, Chicago, IL, 60612, USA
| | - Enza Marchese
- Department of Surgery, University of Illinois at Chicago, 840 S. Wood St. Room 502, Chicago, IL, 60612, USA
| | - Matthew A Bochenek
- Department of Surgery, University of Illinois at Chicago, 840 S. Wood St. Room 502, Chicago, IL, 60612, USA
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, USA
| | - Maha Longi
- Department of Surgery, University of Illinois at Chicago, 840 S. Wood St. Room 502, Chicago, IL, 60612, USA
| | - Yuan Xing
- Department of Surgery, University of Illinois at Chicago, 840 S. Wood St. Room 502, Chicago, IL, 60612, USA
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, USA
| | - Ira Joshi
- Department of Surgery, University of Illinois at Chicago, 840 S. Wood St. Room 502, Chicago, IL, 60612, USA
| | - Yong Wang
- Department of Surgery, University of Illinois at Chicago, 840 S. Wood St. Room 502, Chicago, IL, 60612, USA
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, USA
| | - José Oberholzer
- Department of Surgery, University of Illinois at Chicago, 840 S. Wood St. Room 502, Chicago, IL, 60612, USA
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
43
|
Zhang T, Li XH, Zhang DB, Liu XY, Zhao F, Lin XW, Wang R, Lang HX, Pang XN. Repression of COUP-TFI Improves Bone Marrow-Derived Mesenchymal Stem Cell Differentiation into Insulin-Producing Cells. MOLECULAR THERAPY. NUCLEIC ACIDS 2017; 8:220-231. [PMID: 28918023 PMCID: PMC5504083 DOI: 10.1016/j.omtn.2017.06.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 05/10/2017] [Accepted: 06/20/2017] [Indexed: 01/09/2023]
Abstract
Identifying molecular mechanisms that regulate insulin expression in bone marrow-derived mesenchymal stem cells (bmMSCs) can provide clues on how to stimulate the differentiation of bmMSCs into insulin-producing cells (IPCs), which can be used as a therapeutic approach against type 1 diabetes (T1D). As repression factors may inhibit differentiation, the efficiency of this process is insufficient for cell transplantation. In this study, we used the mouse insulin 2 (Ins2) promoter sequence and performed a DNA affinity precipitation assay combined with liquid chromatography-mass spectrometry to identify the transcription factor, chicken ovalbumin upstream promoter transcriptional factor I (COUP-TFI). Functionally, bmMSCs were reprogrammed into IPCs via COUP-TFI suppression and MafA overexpression. The differentiated cells expressed higher levels of genes specific for islet endocrine cells, and they released C-peptide and insulin in response to glucose stimulation. Transplantation of IPCs into streptozotocin-induced diabetic mice caused a reduction in hyperglycemia. Mechanistically, COUP-TFI bound to the DR1 (direct repeats with 1 spacer) element in the Ins2 promoter, thereby negatively regulating promoter activity. Taken together, the data provide a novel mechanism by which COUP-TFI acts as a negative regulator in the Ins2 promoter. The differentiation of bmMSCs into IPCs could be improved by knockdown of COUP-TFI, which may provide a novel stem cell-based therapy for T1D.
Collapse
Affiliation(s)
- Tao Zhang
- Department of Stem Cells and Regenerative Medicine, Shenyang Key Laboratory for Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang 110122, People's Republic of China
| | - Xiao-Hang Li
- Department of General Surgery, the First Affiliated Hospital of China Medical University, Shenyang 110001, People's Republic of China
| | - Dian-Bao Zhang
- Department of Stem Cells and Regenerative Medicine, Shenyang Key Laboratory for Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang 110122, People's Republic of China
| | - Xiao-Yu Liu
- Department of Stem Cells and Regenerative Medicine, Shenyang Key Laboratory for Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang 110122, People's Republic of China
| | - Feng Zhao
- Department of Stem Cells and Regenerative Medicine, Shenyang Key Laboratory for Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang 110122, People's Republic of China
| | - Xue-Wen Lin
- Department of Stem Cells and Regenerative Medicine, Shenyang Key Laboratory for Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang 110122, People's Republic of China
| | - Rui Wang
- Department of Stem Cells and Regenerative Medicine, Shenyang Key Laboratory for Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang 110122, People's Republic of China
| | - Hong-Xin Lang
- Department of Stem Cells and Regenerative Medicine, Shenyang Key Laboratory for Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang 110122, People's Republic of China
| | - Xi-Ning Pang
- Department of Stem Cells and Regenerative Medicine, Shenyang Key Laboratory for Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang 110122, People's Republic of China; Science Experiment Center, China Medical University, Shenyang 110122, People's Republic of China.
| |
Collapse
|
44
|
Miki A, Rivas-Carrillo JD, Navarro-Alvarez N, Soto-Gutierrez A, Chen Y, Tanaka K, Narushima M, Tabata Y, Okitsu T, Noguchi H, Matsumoto S, Tanaka N, Kobayashi N. Maintenance of Neovascularization at the Implantation Site of an Artificial Device by bFGF and Endothelial Cell Transplant. Cell Transplant 2017; 15:893-901. [PMID: 17299994 DOI: 10.3727/000000006783981378] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Development of a subcutaneously implantable bioartificial pancreas (BAP) with immunoisolatory function could have a great impact on the treatment of diabetes mellitus. We have developed an implantable BAP device with an ethylene vinyl alcohol (EVAL) membrane. In the present study, we used basic fibroblast growth factors (bFGF), which was incorporated in a carrier for sustained release, in order to induce neovascularization when the device was implanted subcutaneously. To maintain the vasculature thus formed, a cell infusion port was attached to the BAP device, through which the device was filled with human liver vascular endothelial cell line TMNK-1, and the vasculature could be adequately maintained. Mice were divided into the following three groups. In group 1, a bFGF-free BAP device was implanted subcutaneously. In group 2, a sustained-release bFGF-impregnated BAP device was implanted. In group 3, a sustained-release bFGF-impregnated BAP device was implanted, and 3 × 106 TMNK-1 cells were infused into the implanted device every week. Neovascularization induced in the subcutaneous tissue around the implanted BAP device was macroscopically examined and histologically evaluated. In addition, the tissue blood flow was measured using a laser blood flow meter. In mice in group 3, neovascularization was significantly induced and maintained until week 8 postimplantation. It was confirmed by scanning electron microscopy that infused TMNK-1 cells adhered to the inner polyethylene surface of the device. It was demonstrated that the use of bFGF and vascular endothelial TMNK-1 cells induced and maintained adequate vasculature and tissue blood flow surrounding the implantable bag-type BAP device. We believe that the present study will contribute to BAP development for the treatment of diabetes.
Collapse
Affiliation(s)
- Atsushi Miki
- Department of Surgery, Okayama University Graduate School of Medicine and Dentistry, Okayama 700-8558, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Al-Khawaga S, Memon B, Butler AE, Taheri S, Abou-Samra AB, Abdelalim EM. Pathways governing development of stem cell-derived pancreatic β cells: lessons from embryogenesis. Biol Rev Camb Philos Soc 2017. [DOI: 10.1111/brv.12349] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Sara Al-Khawaga
- Diabetes Research Center, Qatar Biomedical Research Institute; Hamad Bin Khalifa University, Qatar Foundation, Education City; Doha Qatar
| | - Bushra Memon
- Diabetes Research Center, Qatar Biomedical Research Institute; Hamad Bin Khalifa University, Qatar Foundation, Education City; Doha Qatar
| | - Alexandra E. Butler
- Larry L. Hillblom Islet Research Center, David Geffen School of Medicine; University of California; Los Angeles CA 90095 U.S.A
| | - Shahrad Taheri
- Department of Medicine; Weill Cornell Medicine in Qatar, Qatar Foundation, Education City, PO BOX 24144; Doha Qatar
- Department of Medicine; Qatar Metabolic Institute, Hamad Medical Corporation; Doha Qatar
| | - Abdul B. Abou-Samra
- Department of Medicine; Weill Cornell Medicine in Qatar, Qatar Foundation, Education City, PO BOX 24144; Doha Qatar
- Department of Medicine; Qatar Metabolic Institute, Hamad Medical Corporation; Doha Qatar
| | - Essam M. Abdelalim
- Diabetes Research Center, Qatar Biomedical Research Institute; Hamad Bin Khalifa University, Qatar Foundation, Education City; Doha Qatar
| |
Collapse
|
46
|
Miyashita H, Suzuki A, Fukao K, Nakauchi H, Taniguchi H. Evidence for Hepatocyte Differentiation from Embryonic Stem Cells In Vitro. Cell Transplant 2017. [DOI: 10.3727/000000002783985675] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
We confirmed hepatocyte differentiation from embryonic stem (ES) cells in vitro. RT-PCR analysis revealed that a broad range of hepatic gene expression was observed in ES cells differentiated through formation of embryoid bodies (EBs) and its attachment culture. Quantitative PCR analysis revealed that hepatic gene expression related to early and late-stage liver development were enhanced through in vitro differentiation of ES cells. The presence of albumin-producing cells in the peripheral region of attached EBs was confirmed by immunocytochemical analysis. Future experiments will reveal the molecules that induce hepatocyte differentiation from ES cells in vitro. This research will provide systems for the investigation of mechanisms in liver development and establish a method of ES cell-based therapy for liver diseases.
Collapse
Affiliation(s)
- Hitoshi Miyashita
- Department of Surgery, Institute of Clinical Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Atsushi Suzuki
- Department of Surgery, Institute of Clinical Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Katashi Fukao
- Department of Surgery, Institute of Clinical Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Hiromitsu Nakauchi
- Department of Immunology, Institute of Basic Medical Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Hideki Taniguchi
- Department of Surgery, Institute of Clinical Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| |
Collapse
|
47
|
Choi D, Oh HJ, Chang UJ, Koo SK, Jiang JX, Hwang SY, Lee JD, Yeoh GC, Shin HS, Lee JS, Oh B. In Vivo Differentiation of Mouse Embryonic Stem Cells into Hepatocytes. Cell Transplant 2017. [DOI: 10.3727/000000002783985792] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Embryonic stem (ES) cells have been regarded as a powerful resource for cell replacement therapy. In recent reports mouse ES cells have been successfully applied in the treatment of spinal cord injury, hereditary myelin disorder of the central nervous system, and diabetes mellitus. Another type of disease that could benefit from the availability of stem cell therapy is liver disease. However, for this potential to be realized, it is necessary to demonstrate the differentiation of ES cells into hepatocytes. To demonstrate the in vivo differentiation potential of mouse ES cells, we injected ES cells into the spleen of immunosuppressed nude mice. Histological analysis of teratomas derived from injected ES cells revealed that some areas contained typical hepatocytes arranged in a sinusoidal structure. The hepatic nature of these cells was further confirmed by showing that transcripts of liver-specific genes were present in the differentiated teratoma using reverse transcriptase-polymerase chain reaction and immunohistochemistry using several liver-specific antibodies including HEP-PAR, phenylalanine hydroxylase, and mouse N-system aminotransferase to identify the respective proteins in the differentiated hepatocytes. This is the first demonstration that mouse ES cells can differentiate in vivo into a mixed population of hepatocytes of varying maturity. This finding extends the potential use of ES cells in the cell replacement therapy by including its possible application for treating liver diseases.
Collapse
Affiliation(s)
- Dongho Choi
- Division of Genetic Disease, Department of Biomedical Science, National Institute of Health, Seoul 122-701, Korea
| | - Hyun-Jeong Oh
- Division of Genetic Disease, Department of Biomedical Science, National Institute of Health, Seoul 122-701, Korea
| | - Uck-Jin Chang
- Division of Genetic Disease, Department of Biomedical Science, National Institute of Health, Seoul 122-701, Korea
| | - Soo Kyung Koo
- Division of Genetic Disease, Department of Biomedical Science, National Institute of Health, Seoul 122-701, Korea
| | - Jean X. Jiang
- Department of Biochemistry, University of Texas Health Science Center, San Antonio, TX 78229-3900
| | - Sue-Yun Hwang
- Research Institute of Immunology, Catholic Institutes of Medical Science, The Catholic University of Korea, Seoul 137-701, Korea
| | - Jung-Dal Lee
- Department of Pathology, SongDo Hospital, Seoul 100-453, Korea
| | - George C. Yeoh
- Department of Biochemistry, University of Western Australia, Nedlands, Western Australia, Australia
| | - Hee-Sup Shin
- Korea Institute of Science and Technology, Seoul 130-650, Korea
| | - Jin-Sung Lee
- Department of Pediatrics, Yonsei University Medial School, Seoul 120-752, Korea
| | - Bermseok Oh
- Division of Genetic Disease, Department of Biomedical Science, National Institute of Health, Seoul 122-701, Korea
| |
Collapse
|
48
|
Gene-Diet Interactions in Type 2 Diabetes: The Chicken and Egg Debate. Int J Mol Sci 2017; 18:ijms18061188. [PMID: 28574454 PMCID: PMC5486011 DOI: 10.3390/ijms18061188] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 05/23/2017] [Accepted: 05/26/2017] [Indexed: 02/07/2023] Open
Abstract
Consistent evidence from both experimental and human studies indicates that Type 2 diabetes mellitus (T2DM) is a complex disease resulting from the interaction of genetic, epigenetic, environmental, and lifestyle factors. Nutrients and dietary patterns are important environmental factors to consider in the prevention, development and treatment of this disease. Nutritional genomics focuses on the interaction between bioactive food components and the genome and includes studies of nutrigenetics, nutrigenomics and epigenetic modifications caused by nutrients. There is evidence supporting the existence of nutrient-gene and T2DM interactions coming from animal studies and family-based intervention studies. Moreover, many case-control, cohort, cross-sectional cohort studies and clinical trials have identified relationships between individual genetic load, diet and T2DM. Some of these studies were on a large scale. In addition, studies with animal models and human observational studies, in different countries over periods of time, support a causative relationship between adverse nutritional conditions during in utero development, persistent epigenetic changes and T2DM. This review provides comprehensive information on the current state of nutrient-gene interactions and their role in T2DM pathogenesis, the relationship between individual genetic load and diet, and the importance of epigenetic factors in influencing gene expression and defining the individual risk of T2DM.
Collapse
|
49
|
Mao GH, Lu P, Wang YN, Tian CG, Huang XH, Feng ZG, Zhang JL, Chang HY. Role of PI3K p110β in the differentiation of human embryonic stem cells into islet-like cells. Biochem Biophys Res Commun 2017; 488:109-115. [PMID: 28479244 DOI: 10.1016/j.bbrc.2017.05.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 05/03/2017] [Indexed: 12/16/2022]
Abstract
To investigate the effects of the PI3K inhibitors on the differentiation of insulin-producing cells derived from human embryonic stem cells. Here, we report that human embryonic stem cells induced by phosphatidylinositol-3-kinase (PI3K) p110β inhibitors could produce more mature islet-like cells. Findings were validated by immunofluorescence analysis, quantitative real-time PCR, insulin secretion in vitro and cell transplantation for the diabetic SCID mice. Immunofluorescence analysis revealed that unihormonal insulin-positive cells were predominant in cultures with rare polyhormonal cells. Real-time PCR data showed that islet-like cells expressed key markers of pancreatic endocrine hormones and mature pancreatic β cells including MAFA. Furthermore, this study showed that the expression of most pancreatic endocrine hormones was similar between groups treated with the LY294002 (nonselective PI3K inhibitor) and TGX-221 (PI3K isoform selective inhibitors of class 1β) derivatives. However, the level of insulin mRNA in TGX-221-treated cells was significantly higher than that in LY294002-treated cells. In addition, islet-like cells displayed glucose-stimulated insulin secretion in vitro. After transplantation, islet-like cells improved glycaemic control and ameliorated the survival outcome in diabetic mice. This study demonstrated an important role for PI3K p110β in regulating the differentiation and maturation of islet-like cells derived from human embryonic stem cells.
Collapse
Affiliation(s)
- Gen-Hong Mao
- Reproductive Medical Centre, The Second Affiliated Hospital of Zhengzhou University, Henan Province, 450014, China.
| | - Ping Lu
- Reproductive Medical Centre, The Second Affiliated Hospital of Zhengzhou University, Henan Province, 450014, China
| | - Ya-Nan Wang
- Reproductive Medical Centre, The Second Affiliated Hospital of Zhengzhou University, Henan Province, 450014, China
| | - Chen-Guang Tian
- Department of Endocrinology and Metabolic Diseases, The Second Affiliated Hospital of Zhengzhou University, Henan Province, 450014, China
| | - Xiao-Hui Huang
- Reproductive Medical Centre, The Second Affiliated Hospital of Zhengzhou University, Henan Province, 450014, China
| | - Zong-Gang Feng
- Reproductive Medical Centre, The Second Affiliated Hospital of Zhengzhou University, Henan Province, 450014, China
| | - Jin-Lan Zhang
- Reproductive Medical Centre, The Second Affiliated Hospital of Zhengzhou University, Henan Province, 450014, China
| | - Hong-Yang Chang
- Reproductive Medical Centre, The Second Affiliated Hospital of Zhengzhou University, Henan Province, 450014, China
| |
Collapse
|
50
|
Chen ACH, Lee YL, Fong SW, Wong CCY, Ng EHY, Yeung WSB. Hyperglycemia impedes definitive endoderm differentiation of human embryonic stem cells by modulating histone methylation patterns. Cell Tissue Res 2017; 368:563-578. [PMID: 28283910 DOI: 10.1007/s00441-017-2583-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 01/27/2017] [Indexed: 12/25/2022]
Abstract
Exposure to maternal diabetes during fetal growth is a risk factor for the development of type II diabetes (T2D) in later life. Discovery of the mechanisms involved in this association should provide valuable background for therapeutic treatments. Early embryogenesis involves epigenetic changes including histone modifications. The bivalent histone methylation marks H3K4me3 and H3K27me3 are important for regulating key developmental genes during early fetal pancreas specification. We hypothesized that maternal hyperglycemia disrupted early pancreas development through changes in histone bivalency. A human embryonic stem cell line (VAL3) was used as the cell model for studying the effects of hyperglycemia upon differentiation into definitive endoderm (DE), an early stage of the pancreatic lineage. Hyperglycemic conditions significantly down-regulated the expression levels of DE markers SOX17, FOXA2, CXCR4 and EOMES during differentiation. This was associated with retention of the repressive histone methylation mark H3K27me3 on their promoters under hyperglycemic conditions. The disruption of histone methylation patterns was observed as early as the mesendoderm stage, with Wnt/β-catenin signaling being suppressed during hyperglycemia. Treatment with Wnt/β-catenin signaling activator CHIR-99021 restored the expression levels and chromatin methylation status of DE markers, even in a hyperglycemic environment. The disruption of DE development was also found in mouse embryos at day 7.5 post coitum from diabetic mothers. Furthermore, disruption of DE differentiation in VAL3 cells led to subsequent impairment in pancreatic progenitor formation. Thus, early exposure to hyperglycemic conditions hinders DE development with a possible relationship to the later impairment of pancreas specification.
Collapse
Affiliation(s)
- A C H Chen
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, People's Republic of China
| | - Y L Lee
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, People's Republic of China.
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong Shenzhen Hospital, The University of Hong Kong, Shenzhen, People's Republic of China.
- Center for Reproduction, Development and Growth, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, People's Republic of China.
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Room 747, 21 Sassoon Road, Hong Kong, SAR, People's Republic of China.
| | - S W Fong
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, People's Republic of China
| | - C C Y Wong
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, People's Republic of China
| | - E H Y Ng
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, People's Republic of China
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong Shenzhen Hospital, The University of Hong Kong, Shenzhen, People's Republic of China
- Center for Reproduction, Development and Growth, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, People's Republic of China
| | - W S B Yeung
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, People's Republic of China
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong Shenzhen Hospital, The University of Hong Kong, Shenzhen, People's Republic of China
- Center for Reproduction, Development and Growth, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, People's Republic of China
| |
Collapse
|