1
|
Yokoo T, Watanabe K, Iida K, Nakachi Y, Suzuki H, Shimano H, Takashima S, Okazaki Y, Yamada N, Toyoshima H. Betagenin ameliorates diabetes by inducing insulin secretion and β-cell proliferation. J Biol Chem 2025:108202. [PMID: 39826690 DOI: 10.1016/j.jbc.2025.108202] [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: 08/15/2024] [Revised: 12/22/2024] [Accepted: 01/02/2025] [Indexed: 01/22/2025] Open
Abstract
Recent success with the use of glucagon-like peptide-1 (GLP-1) receptor analogs and dipeptidyl peptidase-4 (DPP-4) inhibitors for the treatment of patients with diabetes has highlighted the role of the intestine as an endocrine organ. Gut-derived hormones, including GLP-1, glucose-dependent insulinotropic polypeptide (GIP), and ghrelin, have important roles in the control of energy metabolism and food intake, and are associated with the metabolic syndrome. In this study, we isolated and identified a new intestine-derived hormone, betagenin, and showed that it stimulates insulin secretion and β-cell proliferation and suppresses β-cell apoptosis. Adenovirus-mediated expression of betagenin restored the blood glucose concentrations and hemoglobin A1c (HbA1c) levels of mice with streptozotocin (STZ)-induced diabetes to normal and increased their β-cell mass. Transgenic mice overexpressing betagenin exhibited more than three-fold higher β-cell mass than wild-type mass, whereas that of knockout mice was four-fold lower. A synthetic peptide representing the sequence of purified and secreted betagenin enhanced glucose-dependent insulin secretion in human and mouse pancreatic islets and stimulated the proliferation of the pancreatic β-cell line MIN6 through extracellular signal-regulated kinase (ERK) 1/2-dependent signaling. The intravenous administration of this peptide to STZ mice stimulated the proliferation of pancreatic β-cells in vivo, and the intraperitoneal administration of betagenin ameliorated diabetes and restored β-cell mass. These results indicate that betagenin may reduce blood glucose concentration and induce β-cell regeneration in patients with diabetes.
Collapse
Affiliation(s)
- Tomotaka Yokoo
- Division of Experimental Animal, Hidaka Branch, Biomedical Research Center, Saitama Medical University, Saitama, Japan; Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan
| | - Kazuhisa Watanabe
- Department of Endocrinology and Metabolism, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan; Division of Human Genetics, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Kaoruko Iida
- Department of Endocrinology and Metabolism, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan; Department of Food and Nutrition Science, Graduate School of Humanities and Sciences, Ochanomizu University, Tokyo, Japan
| | - Yutaka Nakachi
- Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan; Department of Molecular Brain Science, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hiroaki Suzuki
- Department of Endocrinology and Metabolism, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Hitoshi Shimano
- Department of Endocrinology and Metabolism, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Seiji Takashima
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yasushi Okazaki
- Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan; Diagnostics and Therapeutics of Intractable Diseases, Intractable Disease Research Center, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Nobuhiro Yamada
- Department of Endocrinology and Metabolism, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Hideo Toyoshima
- Division of Experimental Animal, Hidaka Branch, Biomedical Research Center, Saitama Medical University, Saitama, Japan; Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan.
| |
Collapse
|
2
|
Sylvester-Armstrong KR, Reeder CF, Powell A, Becker MW, Hagan DW, Chen J, Mathews CE, Wasserfall CH, Atkinson MA, Egerman R, Phelps EA. Serum from pregnant donors induces human beta cell proliferation. Islets 2024; 16:2334044. [PMID: 38533763 PMCID: PMC10978022 DOI: 10.1080/19382014.2024.2334044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 03/19/2024] [Indexed: 03/28/2024] Open
Abstract
Pancreatic beta cells are among the slowest replicating cells in the human body and have not been observed to increase in number except during the fetal and neonatal period, in cases of obesity, during puberty, as well as during pregnancy. Pregnancy is associated with increased beta cell mass to meet heightened insulin demands. This phenomenon raises the intriguing possibility that factors present in the serum of pregnant individuals may stimulate beta cell proliferation and offer insights into expansion of the beta cell mass for treatment and prevention of diabetes. The primary objective of this study was to test the hypothesis that serum from pregnant donors contains bioactive factors capable of inducing human beta cell proliferation. An immortalized human beta cell line with protracted replication (EndoC-βH1) was cultured in media supplemented with serum from pregnant and non-pregnant female and male donors and assessed for differences in proliferation. This experiment was followed by assessment of proliferation of primary human beta cells. Sera from five out of six pregnant donors induced a significant increase in the proliferation rate of EndoC-βH1 cells. Pooled serum from the cohort of pregnant donors also increased the rate of proliferation in primary human beta cells. This study demonstrates that serum from pregnant donors stimulates human beta cell proliferation. These findings suggest the existence of pregnancy-associated factors that can offer novel avenues for beta cell regeneration and diabetes prevention strategies. Further research is warranted to elucidate the specific factors responsible for this effect.
Collapse
Affiliation(s)
| | - Callie F. Reeder
- Department of Obstetrics & Gynecology, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Andrece Powell
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, USA
| | - Matthew W. Becker
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, USA
| | - D. Walker Hagan
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, USA
| | - Jing Chen
- Department of Pathology, Immunology, and Laboratory Medicine and University of Florida Diabetes Institute, University of Florida, Gainesville, Florida, USA
| | - Clayton E. Mathews
- Department of Pathology, Immunology, and Laboratory Medicine and University of Florida Diabetes Institute, University of Florida, Gainesville, Florida, USA
| | - Clive H. Wasserfall
- Department of Pathology, Immunology, and Laboratory Medicine and University of Florida Diabetes Institute, University of Florida, Gainesville, Florida, USA
| | - Mark A. Atkinson
- Department of Pathology, Immunology, and Laboratory Medicine and University of Florida Diabetes Institute, University of Florida, Gainesville, Florida, USA
| | - Robert Egerman
- Department of Obstetrics & Gynecology, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Edward A. Phelps
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, USA
- Department of Pathology, Immunology, and Laboratory Medicine and University of Florida Diabetes Institute, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
3
|
Liu Y, Yang X, Zhou J, Yang H, Yang R, Zhu P, Zhou R, Wu T, Gao Y, Ye Z, Li X, Liu R, Zhang W, Zhou H, Li Q. OSGEP regulates islet β-cell function by modulating proinsulin translation and maintaining ER stress homeostasis in mice. Nat Commun 2024; 15:10479. [PMID: 39622811 PMCID: PMC11612026 DOI: 10.1038/s41467-024-54905-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 11/25/2024] [Indexed: 12/06/2024] Open
Abstract
Proinsulin translation and folding is crucial for glucose homeostasis. However, islet β-cell control of Proinsulin translation remains incompletely understood. Here, we identify OSGEP, an enzyme responsible for t6A37 modification of tRNANNU that tunes glucose metabolism in β-cells. Global Osgep deletion causes glucose intolerance, while β-cell-specific deletion induces hyperglycemia and glucose intolerance due to impaired insulin activity. Transcriptomics and proteomics reveal activation of the unfolded protein response (UPR) and apoptosis signaling pathways in Osgep-deficient islets, linked to an increase in misfolded Proinsulin from reduced t6A37 modification. Osgep overexpression in pancreas rescues insulin secretion and mitigates diabetes in high-fat diet mice. Osgep enhances translational fidelity and alleviates UPR signaling, highlighting its potential as a therapeutic target for diabetes. Individuals carrying the C allele at rs74512655, which promotes OSGEP transcription, may show reduced susceptibility to T2DM. These findings show OSGEP is essential for islet β-cells and a potential diabetes therapy target.
Collapse
Affiliation(s)
- Yujie Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, 410078, China
- National Clinical Research Center for Geriatric Disorders, Changsha, 410008, China
- Department of Pharmacy, Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Xuechun Yang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, 410078, China
- National Clinical Research Center for Geriatric Disorders, Changsha, 410008, China
| | - Jian Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, 410078, China
- National Clinical Research Center for Geriatric Disorders, Changsha, 410008, China
| | - Haijun Yang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, 410078, China
- National Clinical Research Center for Geriatric Disorders, Changsha, 410008, China
| | - Ruimeng Yang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, 410078, China
- National Clinical Research Center for Geriatric Disorders, Changsha, 410008, China
| | - Peng Zhu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, 410078, China
- National Clinical Research Center for Geriatric Disorders, Changsha, 410008, China
| | - Rong Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, 410078, China
- National Clinical Research Center for Geriatric Disorders, Changsha, 410008, China
| | - Tianyuan Wu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, 410078, China
- National Clinical Research Center for Geriatric Disorders, Changsha, 410008, China
| | - Yongchao Gao
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, 410078, China
- National Clinical Research Center for Geriatric Disorders, Changsha, 410008, China
| | - Zhi Ye
- Department of Anesthesiology, Xiangya Hospital of Central South University, Changsha, 410008, China
| | - Xi Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, 410078, China
- National Clinical Research Center for Geriatric Disorders, Changsha, 410008, China
| | - Rong Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, 410078, China
- National Clinical Research Center for Geriatric Disorders, Changsha, 410008, China
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, 410078, China
- National Clinical Research Center for Geriatric Disorders, Changsha, 410008, China
| | - Honghao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, 410078, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, 410078, China
- National Clinical Research Center for Geriatric Disorders, Changsha, 410008, China
| | - Qing Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, 410078, China.
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, 410078, China.
- National Clinical Research Center for Geriatric Disorders, Changsha, 410008, China.
| |
Collapse
|
4
|
Jin Y, Wang H. Identification of hub genes affecting gestational diabetes mellitus based on GEO database. Biotechnol Genet Eng Rev 2024; 40:4653-4663. [PMID: 37224002 DOI: 10.1080/02648725.2023.2215966] [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: 02/22/2023] [Accepted: 05/14/2023] [Indexed: 05/26/2023]
Abstract
This research aimed to obtain gestational diabetes mellitus (GDM) related hub genes, providing new targets for clinical diagnosis and treatment of GDM. The microarray data of GSE9984 and GSE103552 were obtained from the Gene Expression Omnibus (GEO). The dataset GSE9984 contained placental gene expression profiles of 8 GDM patients and four healthy specimens. The dataset GSE103552 contained 20 specimens from GDM patients and 17 normal specimens. The differentially expressed genes (DEGs) were identified by GEO2R online analysis. DAVID database was applied to conduct functional enrichment analysis of the DEGs. The Search Tool for the Retrieval of Interacting Genes (STRING) database was adopted to acquire protein-protein interaction (PPI) networks. A total of 195 up-regulated and 371 down-regulated DEGs were selected in the GSE9984, and total of 191 up-regulated and 229 down-regulated DEGs were selected in the GSE103552. In the two datasets, 24 common differential genes were obtained and named co-DEGs. The Gene Ontology (GO) annotation analysis indicated the DEGs participated in multi-multicellular organism process, endocrine hormone secretion, long-chain fatty acid biosynthetic process, cell division, unsaturated fatty acid biosynthetic process, cell adhesion and cell recognition. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis suggested that GSE9984 and GSE103552 were related to vitamin digestion and absorption, tryptophan metabolism, steroid hormone biosynthesis, Ras signaling pathway, protein digestion and absorption, PPAR signaling pathway, PI3K-Akt signaling pathway, p53 signaling pathway. PPI was constructed in string database, and six hub genes were selected, including CCNB1, APOA2, AHSG and IGFBP1. Four critical genes were identified to be considered as therapeutic potential biomarkers of GDM, including CCNB1, APOA2, AHSG and IGFBP1.
Collapse
Affiliation(s)
- Yangqiu Jin
- Department of Reproductive Medicine, Liaocheng People's Hospital, Liaocheng, Shandong Province, China
| | - Hui Wang
- Department of Obstetrics and Gynecology,Liaocheng Tird People's Hospital, Liaocheng, Shandong Province, China
| |
Collapse
|
5
|
Luo Y, Yu P, Liu J. The efficiency of stem cell differentiation into functional beta cells for treating insulin-requiring diabetes: Recent advances and current challenges. Endocrine 2024; 86:1-14. [PMID: 38730069 DOI: 10.1007/s12020-024-03855-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 04/29/2024] [Indexed: 05/12/2024]
Abstract
In recent years, the potential of stem cells (SCs) to differentiate into various types of cells, including β-cells, has led to a significant boost in development. The efficiency of this differentiation process and the functionality of the cells post-transplantation are crucial factors for the success of stem cell therapy in diabetes. Herein, this article reviews the current advances and challenges faced by stem cell differentiation into functional β-cells for diabetes treatment. In vitro, researchers have sought to enhance the differentiation efficiency of functional β-cells by mimicking the normal pancreatic development process, using gene manipulation, pharmacological and culture conditions stimulation, three-dimensional (3D) and organoid culture, or sorting for functional β-cells based on mature islet cell markers. Furthermore, in vivo studies have also looked at suitable transplantation sites, the enhancement of the transplantation microenvironment, immune modulation, and vascular function reconstruction to improve the survival rate of functional β-cells, thereby enhancing the treatment of diabetes. Despite these advancements, developing stem cells to produce functional β-cells for efficacious diabetes treatment is a continuous research endeavor requiring significant multidisciplinary collaboration, for the stem-cell-derived beta cells to evolve into an effective cellular therapy.
Collapse
Affiliation(s)
- Yunfei Luo
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Peng Yu
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Jianping Liu
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
| |
Collapse
|
6
|
Yu D, Luo L, Wang H, Shyh-Chang N. Pregnancy-induced metabolic reprogramming and regenerative responses to pro-aging stresses. Trends Endocrinol Metab 2024:S1043-2760(24)00192-9. [PMID: 39122601 DOI: 10.1016/j.tem.2024.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/12/2024] [Accepted: 07/17/2024] [Indexed: 08/12/2024]
Abstract
Pregnancy is associated with physiological adaptations that affect virtually all organs, enabling the mother to support the growing fetus and placenta while withstanding the demands of pregnancy. As a result, mammalian pregnancy is a unique state that exerts paradoxical effects on maternal health. On one hand, the metabolic stress induced by pregnancy can accelerate aging and functional decline in organs. On the other hand, pregnancy activates metabolic programming and tissue regenerative responses that can reverse age-related impairments. In this sense, the oocyte-to-blastocyst transition is not the only physiological reprogramming event in the mammalian body, as pregnancy-induced regeneration could constitute a second physiological reprogramming event. Here, we review findings on how pregnancy dualistically leads to aging and rejuvenation in the maternal body.
Collapse
Affiliation(s)
- Dainan Yu
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Lanfang Luo
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; School of Biological Engineering, Zhuhai Campus of Zunyi Medical University, Guangdong 519000, China
| | - Hongmei Wang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Ng Shyh-Chang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
7
|
Jimenez-Gonzalez M, Stanley S. Optogenetic control of β cell function. Nat Biomed Eng 2024; 8:801-803. [PMID: 38057429 DOI: 10.1038/s41551-023-01125-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/08/2023]
Affiliation(s)
| | - Sarah Stanley
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine, New York, NY, USA.
| |
Collapse
|
8
|
Moon JH, Choe HJ, Lim S. Pancreatic beta-cell mass and function and therapeutic implications of using antidiabetic medications in type 2 diabetes. J Diabetes Investig 2024; 15:669-683. [PMID: 38676410 PMCID: PMC11143426 DOI: 10.1111/jdi.14221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 03/23/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Nowadays, the focus of diabetes treatment has switched from lowering the glucose level to preserving glycemic homeostasis and slowing the disease progression. The main pathophysiology of both type 1 diabetes and long-standing type 2 diabetes is pancreatic β-cell mass loss and dysfunction. According to recent research, human pancreatic β-cells possess the ability to proliferate in response to elevated insulin demands. It has been demonstrated that in insulin-resistant conditions in humans, such as obesity or pregnancy, the β-cell mass increases. This ability could be helpful in developing novel treatment approaches to restore a functional β-cell mass. Treatment strategies aimed at boosting β-cell function and mass may be a useful tool for managing diabetes mellitus and stopping its progression. This review outlines the processes of β-cell failure and detail the many β-cell abnormalities that manifest in people with diabetes mellitus. We also go over standard techniques for determining the mass and function of β-cells. Lastly, we provide the therapeutic implications of utilizing antidiabetic drugs in controlling the mass and function of pancreatic β-cells.
Collapse
Affiliation(s)
- Joon Ho Moon
- Department of Internal MedicineSeoul National University College of MedicineSeongnamSouth Korea
- Department of Internal MedicineSeoul National University Bundang HospitalSeongnamSouth Korea
| | - Hun Jee Choe
- Department of Internal MedicineHallym University Dongtan Sacred Heart HospitalHwaseongSouth Korea
| | - Soo Lim
- Department of Internal MedicineSeoul National University College of MedicineSeongnamSouth Korea
- Department of Internal MedicineSeoul National University Bundang HospitalSeongnamSouth Korea
| |
Collapse
|
9
|
Tanner AR, Kennedy VC, Lynch CS, Winger QA, Anthony RV, Rozance PJ. Increasing maternal glucose concentrations is insufficient to restore placental glucose transfer in chorionic somatomammotropin RNA interference pregnancies. Am J Physiol Endocrinol Metab 2024; 326:E602-E615. [PMID: 38353640 PMCID: PMC11376830 DOI: 10.1152/ajpendo.00331.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 02/01/2024] [Accepted: 02/05/2024] [Indexed: 04/19/2024]
Abstract
We previously demonstrated impaired placental nutrient transfer in chorionic somatomammotropin (CSH) RNA interference (RNAi) pregnancies, with glucose transfer being the most impacted. Thus, we hypothesized that despite experimentally elevating maternal glucose, diminished umbilical glucose uptake would persist in CSH RNAi pregnancies, demonstrating the necessity of CSH for adequate placental glucose transfer. Trophectoderm of sheep blastocysts (9 days of gestational age; dGA) were infected with a lentivirus expressing either nontargeting control (CON RNAi; n = 5) or CSH-specific shRNA (CSH RNAi; n = 7) before transfer into recipient sheep. At 126 dGA, pregnancies were fitted with vascular catheters and underwent steady-state metabolic studies (3H2O transplacental diffusion) at 137 ± 0 dGA, before and during a maternal hyperglycemic clamp. Umbilical glucose and oxygen uptakes, as well as insulin and IGF1 concentrations, were impaired (P ≤ 0.01) in CSH RNAi fetuses and were not rescued by elevated maternal glucose. This is partially due to impaired uterine and umbilical blood flow (P ≤ 0.01). However, uteroplacental oxygen utilization was greater (P ≤ 0.05) during the maternal hyperglycemic clamp, consistent with greater placental oxidation of substrates. The relationship between umbilical glucose uptake and the maternal-fetal glucose gradient was analyzed, and while the slope (CON RNAi, Y = 29.54X +74.15; CSH RNAi, Y = 19.05X + 52.40) was not different, the y-intercepts and elevation were (P = 0.003), indicating reduced maximal glucose transport during maternal hyperglycemia. Together, these data suggested that CSH plays a key role in modulating placental metabolism that ultimately promotes maximal placental glucose transfer.NEW & NOTEWORTHY The current study demonstrated a novel, critical autocrine role for chorionic somatomammotropin in augmenting placental glucose transfer and maintaining placental oxidative metabolism. In pregnancies with CSH deficiency, excess glucose in maternal circulation is insufficient to overcome fetal hypoglycemia due to impaired placental glucose transfer and elevated placental metabolic demands. This suggests that perturbations in glucose transfer in CSH RNAi pregnancies are due to compromised metabolic efficiency along with reduced placental mass.
Collapse
Affiliation(s)
- Amelia R Tanner
- Animal Reproduction and Biotechnology Laboratory, Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States
- Perinatal Research Center, University of Colorado School of Medicine, Aurora, Colorado, United States
| | - Victoria C Kennedy
- Animal Reproduction and Biotechnology Laboratory, Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States
| | - Cameron S Lynch
- Animal Reproduction and Biotechnology Laboratory, Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States
| | - Quinton A Winger
- Animal Reproduction and Biotechnology Laboratory, Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States
| | - Russell V Anthony
- Animal Reproduction and Biotechnology Laboratory, Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States
| | - Paul J Rozance
- Perinatal Research Center, University of Colorado School of Medicine, Aurora, Colorado, United States
| |
Collapse
|
10
|
Cui D, Feng X, Lei S, Zhang H, Hu W, Yang S, Yu X, Su Z. Pancreatic β-cell failure, clinical implications, and therapeutic strategies in type 2 diabetes. Chin Med J (Engl) 2024; 137:791-805. [PMID: 38479993 PMCID: PMC10997226 DOI: 10.1097/cm9.0000000000003034] [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: 10/13/2023] [Indexed: 04/06/2024] Open
Abstract
ABSTRACT Pancreatic β-cell failure due to a reduction in function and mass has been defined as a primary contributor to the progression of type 2 diabetes (T2D). Reserving insulin-producing β-cells and hence restoring insulin production are gaining attention in translational diabetes research, and β-cell replenishment has been the main focus for diabetes treatment. Significant findings in β-cell proliferation, transdifferentiation, pluripotent stem cell differentiation, and associated small molecules have served as promising strategies to regenerate β-cells. In this review, we summarize current knowledge on the mechanisms implicated in β-cell dynamic processes under physiological and diabetic conditions, in which genetic factors, age-related alterations, metabolic stresses, and compromised identity are critical factors contributing to β-cell failure in T2D. The article also focuses on recent advances in therapeutic strategies for diabetes treatment by promoting β-cell proliferation, inducing non-β-cell transdifferentiation, and reprograming stem cell differentiation. Although a significant challenge remains for each of these strategies, the recognition of the mechanisms responsible for β-cell development and mature endocrine cell plasticity and remarkable advances in the generation of exogenous β-cells from stem cells and single-cell studies pave the way for developing potential approaches to cure diabetes.
Collapse
Affiliation(s)
- Daxin Cui
- Molecular Medicine Research Center and Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xingrong Feng
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Siman Lei
- Clinical Translational Innovation Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Hongmei Zhang
- Molecular Medicine Research Center and Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Wanxin Hu
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Shanshan Yang
- Molecular Medicine Research Center and Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xiaoqian Yu
- Molecular Medicine Research Center and Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhiguang Su
- Molecular Medicine Research Center and Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Clinical Translational Innovation Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
11
|
Choi J, Cayabyab F, Perez H, Yoshihara E. Scaling Insulin-Producing Cells by Multiple Strategies. Endocrinol Metab (Seoul) 2024; 39:191-205. [PMID: 38572534 PMCID: PMC11066437 DOI: 10.3803/enm.2023.1910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 01/20/2024] [Accepted: 01/30/2024] [Indexed: 04/05/2024] Open
Abstract
In the quest to combat insulin-dependent diabetes mellitus (IDDM), allogenic pancreatic islet cell therapy sourced from deceased donors represents a significant therapeutic advance. However, the applicability of this approach is hampered by donor scarcity and the demand for sustained immunosuppression. Human induced pluripotent stem cells are a game-changing resource for generating synthetic functional insulin-producing β cells. In addition, novel methodologies allow the direct expansion of pancreatic progenitors and mature β cells, thereby circumventing prolonged differentiation. Nevertheless, achieving practical reproducibility and scalability presents a substantial challenge for this technology. As these innovative approaches become more prominent, it is crucial to thoroughly evaluate existing expansion techniques with an emphasis on their optimization and scalability. This manuscript delineates these cutting-edge advancements, offers a critical analysis of the prevailing strategies, and underscores pivotal challenges, including cost-efficiency and logistical issues. Our insights provide a roadmap, elucidating both the promises and the imperatives in harnessing the potential of these cellular therapies for IDDM.
Collapse
Affiliation(s)
- Jinhyuk Choi
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Fritz Cayabyab
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Harvey Perez
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Eiji Yoshihara
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
- David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
12
|
Kimani CN, Reuter H, Kotzé SH, Venter P, Ramharack P, Muller CJF. Pancreatic beta cell regenerative potential of Zanthoxylum chalybeum Engl. Aqueous stem bark extract. JOURNAL OF ETHNOPHARMACOLOGY 2024; 320:117374. [PMID: 37944876 DOI: 10.1016/j.jep.2023.117374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 10/18/2023] [Accepted: 10/30/2023] [Indexed: 11/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Zanthoxylum chalybeum Engl. is endemic to Africa and has been used traditionally to treat diabetes mellitus. Moreover, its pharmacological efficacy has been confirmed experimentally using in vitro and in vivo models of diabetes. However, the effects of Z. chalybeum extracts and its major constituent compounds on beta cell and islet regeneration are not clear. Further, the mechanisms associated with observed antidiabetic effects at the beta cell level are not fully elucidated. AIM OF THE STUDY We determined the beta cell regenerative efficacy of Z. chalybeum aqueous stem bark extract, identified the chemical compounds in Z. chalybeum aqueous stem bark extracts and explored their putative mechanisms of action. MATERIALS AND METHODS Phytochemical profiling of the Z. chalybeum extract was achieved using ultra high-performance liquid chromatography hyphenated to high-resolution mass spectrometry. Thereafter, molecular interactions of the compounds with beta cell regeneration targets were evaluated via molecular docking. In vitro, effects of the extract on cell viability, proliferation, apoptosis and oxidative stress were investigated in RIN-5F beta cells exposed to palmitate or streptozotocin. In vivo, pancreas tissue sections from streptozotocin-induced diabetic male Wistar rats treated with Z. chalybeum extract were stained for insulin, glucagon, pancreatic duodenal homeobox protein 1 (Pdx-1) and Ki-67. RESULTS Based on ligand target and molecular docking interactions diosmin was identified as a dual specificity tyrosine-phosphorylation-regulated kinase 1A (Dyrk1A) inhibitor. In vitro, Z. chalybeum augmented cell viability and cell proliferation while in palmitate-pre-treated cells, the extract significantly increased cell activity after 72 h. In vivo, although morphometric analysis showed decreased islet and beta cell size and density, observation of increased Pdx-1 and Ki-67 immunoreactivity in extract-treated islets suggests that Z. chalybeum extract has mild beta cell regenerative potential mediated by increased cell proliferation. CONCLUSIONS Overall, the mitogenic effects observed in vitro, were not robust enough to elicit sufficient recovery of functional beta cell mass in our in vivo model, in the context of a sustained diabetic milieu. However, the identification of diosmin as a potential Dyrk1A inhibitor merits further inquiry into the attendant molecular interactions.
Collapse
Affiliation(s)
- Clare Njoki Kimani
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council (SAMRC), Tygerberg, 7505, South Africa; Division of Clinical Pharmacology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, PO Box 241, Cape Town, 8000, South Africa; Department of Non-communicable Diseases, Institute of Primate Research, PO Box 24481, Karen, Nairobi, Kenya.
| | - Helmuth Reuter
- Division of Clinical Pharmacology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, PO Box 241, Cape Town, 8000, South Africa
| | - Sanet Henriët Kotzé
- Division of Clinical Anatomy, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, PO Box 241, Cape Town, 8000, South Africa; Division of Anatomy, Department of Biomedical Sciences, School of Veterinary Medicine, Ross University, PO Box 334, Basseterre, Saint Kitts and Nevis
| | - Pieter Venter
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council (SAMRC), Tygerberg, 7505, South Africa
| | - Pritika Ramharack
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council (SAMRC), Tygerberg, 7505, South Africa; Discipline of Pharmaceutical Sciences, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban, 4001, South Africa
| | - Christo John Frederick Muller
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council (SAMRC), Tygerberg, 7505, South Africa; Centre for Cardio-Metabolic Research in Africa, Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Stellenbosch, 7600, South Africa; Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa, 3886, South Africa
| |
Collapse
|
13
|
Katz LS, Brill G, Wang P, Lambertini L, Zhang P, Haldeman JM, Liu H, Newgard CB, Stewart AF, Garcia-Ocaña A, Scott DK. Transcriptional activation of the Myc gene by glucose in β-cells requires a ChREBP-dependent 3-D chromatin interaction between the Myc and Pvt1 genes. Mol Metab 2024; 79:101848. [PMID: 38042369 PMCID: PMC10714240 DOI: 10.1016/j.molmet.2023.101848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 11/14/2023] [Accepted: 11/28/2023] [Indexed: 12/04/2023] Open
Abstract
OBJECTIVE All forms of diabetes result from insufficient functional β-cell mass. Thus, achieving the therapeutic goal of expanding β-cell mass requires a better mechanistic understanding of how β-cells proliferate. Glucose is a natural β-cell mitogen that mediates its effects in part through the glucose-responsive transcription factor, carbohydrate response element binding protein (ChREBP) and the anabolic transcription factor, MYC. However, mechanistic details by which glucose activates Myc at the transcriptional level are poorly understood. METHODS Here, siRNA was used to test the role of ChREBP in the glucose response of MYC, ChIP and ChIPseq to identify potential regulatory binding sites, chromatin conformation capture to identify DNA/DNA interactions, and an adenovirus was constructed to expresses x-dCas9 and an sgRNA that specifically disrupts the recruitment of ChREBP to a specific targeted ChoRE. RESULTS We found that ChREBP is essential for glucose-mediated transcriptional induction of Myc, and for increases in Myc mRNA and protein abundance. Further, ChIPseq revealed that the carbohydrate response element (ChoRE) nearest to the Myc transcriptional start site (TSS) is immediately upstream of the gene encoding the lncRNA, Pvt1, 60,000 bp downstream of the Myc gene. Chromatin Conformation Capture (3C) confirmed a glucose-dependent interaction between these two sites. Transduction with an adenovirus expressing x-dCas9 and an sgRNA specifically targeting the highly conserved Pvt1 ChoRE, attenuates ChREBP recruitment, decreases Myc-Pvt1 DNA/DNA interaction, and decreases expression of the Pvt1 and Myc genes in response to glucose. Importantly, isolated and dispersed rat islet cells transduced with the ChoRE-disrupting adenovirus also display specific decreases in ChREBP-dependent, glucose-mediated expression of Pvt1 and Myc, as well as decreased glucose-stimulated β-cell proliferation. CONCLUSIONS The mitogenic glucose response of Myc is mediated via glucose-dependent recruitment of ChREBP to the promoter of the Pvt1 gene and subsequent DNA looping with the Myc promoter.
Collapse
Affiliation(s)
- Liora S Katz
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1152, New York, NY 10029, USA
| | - Gabriel Brill
- Pharmacologic Sciences Department, Stony Brook University, Stony Brook, NY, USA(5)
| | - Peng Wang
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1152, New York, NY 10029, USA
| | - Luca Lambertini
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1152, New York, NY 10029, USA
| | - Pili Zhang
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1152, New York, NY 10029, USA
| | | | - Hongtao Liu
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1152, New York, NY 10029, USA
| | | | - Andrew F Stewart
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1152, New York, NY 10029, USA
| | - Adolfo Garcia-Ocaña
- Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope, 1500 E Duarte Rd, Duarte, CA 91010, USA
| | - Donald K Scott
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1152, New York, NY 10029, USA.
| |
Collapse
|
14
|
Choleva L, Wang P, Liu H, Wood O, Lambertini L, Scott DK, Karakose E, Stewart AF. Structure-Function Analysis of p57KIP2 in the Human Pancreatic Beta Cell Reveals a Bipartite Nuclear Localization Signal. Endocrinology 2023; 165:bqad197. [PMID: 38151968 PMCID: PMC11491829 DOI: 10.1210/endocr/bqad197] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 12/12/2023] [Accepted: 12/22/2023] [Indexed: 12/29/2023]
Abstract
Mutations in CDKN1C, encoding p57KIP2, a canonical cell cycle inhibitor, underlie multiple pediatric endocrine syndromes. Despite this central role in disease, little is known about the structure and function of p57KIP2 in the human pancreatic beta cell. Since p57KIP2 is predominantly nuclear in human beta cells, we hypothesized that disease-causing mutations in its nuclear localization sequence (NLS) may correlate with abnormal phenotypes. We prepared RIP1 insulin promoter-driven adenoviruses encoding deletions of multiple disease-associated but unexplored regions of p57KIP2 and performed a comprehensive structure-function analysis of CDKN1C/p57KIP2. Real-time polymerase chain reaction and immunoblot analyses confirmed p57KIP2 overexpression, construct size, and beta cell specificity. By immunocytochemistry, wild-type (WT) p57KIP2 displayed nuclear localization. In contrast, deletion of a putative NLS at amino acids 278-281 failed to access the nucleus. Unexpectedly, we identified a second downstream NLS at amino acids 312-316. Further analysis showed that each individual NLS is required for nuclear localization, but neither alone is sufficient. In summary, p57KIP2 contains a classical bipartite NLS characterized by 2 clusters of positively charged amino acids separated by a proline-rich linker region. Variants in the sequences encoding these 2 NLS sequences account for functional p57KIP2 loss and beta cell expansion seen in human disease.
Collapse
Affiliation(s)
- Lauryn Choleva
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Peng Wang
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Hongtao Liu
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Olivia Wood
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Luca Lambertini
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Donald K Scott
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Esra Karakose
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Andrew F Stewart
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
15
|
Naidoo K, Khathi A. The Potential Role of Gossypetin in the Treatment of Diabetes Mellitus and Its Associated Complications: A Review. Int J Mol Sci 2023; 24:17609. [PMID: 38139436 PMCID: PMC10743819 DOI: 10.3390/ijms242417609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/14/2023] [Accepted: 12/15/2023] [Indexed: 12/24/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a metabolic disorder caused by insulin resistance and dysfunctional beta (β)-cells in the pancreas. Hyperglycaemia is a characteristic of uncontrolled diabetes which eventually leads to fatal organ system damage. In T2DM, free radicals are continuously produced, causing extensive tissue damage and subsequent macro-and microvascular complications. The standard approach to managing T2DM is pharmacological treatment with anti-diabetic medications. However, patients' adherence to treatment is frequently decreased by the side effects and expense of medications, which has a detrimental impact on their health outcomes. Quercetin, a flavonoid, is a one of the most potent anti-oxidants which ameliorates T2DM. Thus, there is an increased demand to investigate quercetin and its derivatives, as it is hypothesised that similar structured compounds may exhibit similar biological activity. Gossypetin is a hexahydroxylated flavonoid found in the calyx of Hibiscus sabdariffa. Gossypetin has a similar chemical structure to quercetin with an extra hydroxyl group. Furthermore, previous literature has elucidated that gossypetin exhibits neuroprotective, hepatoprotective, reproprotective and nephroprotective properties. The mechanisms underlying gossypetin's therapeutic potential have been linked to its anti-oxidant, anti-inflammatory and immunomodulatory properties. Hence, this review highlights the potential role of gossypetin in the treatment of diabetes and its associated complications.
Collapse
Affiliation(s)
| | - Andile Khathi
- Department of Human Physiology, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban 4000, South Africa;
| |
Collapse
|
16
|
Ye L, Lv Y, Wu Q, Chen Y, Zhang X, Su Y. Chronic periodontitis induces the proliferation of pancreatic β-cells to cause hyperinsulinemia in a rat model. J Periodontal Res 2023; 58:1290-1299. [PMID: 37723987 DOI: 10.1111/jre.13185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 08/19/2023] [Accepted: 09/01/2023] [Indexed: 09/20/2023]
Abstract
BACKGROUND AND OBJECTIVE The purpose of this study was to determine if chronic periodontitis (CP) may induce hyperinsulinemia and may have the effect of on pancreatic β-cell proliferation in a rat model. MATERIALS AND METHODS Twelve male Sprague-Dawley rats were divided into two groups: the CP group and the control group (Con group). The following contents were evaluated: pathological changes in periodontal soft and hard tissues; serum lipopolysaccharide (LPS) level, serum fasting insulin (FINS) level, fasting blood glucose (FBG) level, and homeostasis model assessment (HOMA) β (HOMA-β) index; histopathological examination of islets; immunohistochemistry of insulin and p-Smad2 expression in islets; immunofluorescence of changes in the relative number of β-cells and the number of Ki67-positive β-cells. Western blotting was used to analyze p-Smad2/Smad2 levels. Results were analyzed by two independent samples t tests. RESULTS Increased serum LPS level, FINS level, and HOMA-β index were observed in the rats of the CP group; FBG level did not change significantly; histological assessments showed an enlarged islet area, increased insulin content, relatively increased β-cells, increased Ki67-positive β-cells, and decreased p-Smad2 expression in islets in the rats of the CP group. CONCLUSION Our study results link CP-induced hyperinsulinemia with changes in islets, such as islet hyperplasia and compensatory β-cell proliferation, by using a CP rat model.
Collapse
Affiliation(s)
- Leilei Ye
- Department of Periodontology, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Yingtao Lv
- Department of Implantology, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Qianqi Wu
- Stomatology Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, China
| | - Yiyan Chen
- Stomatology Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, China
| | - Xueyang Zhang
- Department of Periodontology, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
- Stomatology Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, China
| | - Yuan Su
- Stomatology Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, China
| |
Collapse
|
17
|
Hirano M, Yamada Y. Reprogramming of pancreatic islet cells for regeneration and rejuvenation. Curr Opin Genet Dev 2023; 82:102099. [PMID: 37611379 DOI: 10.1016/j.gde.2023.102099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 07/13/2023] [Accepted: 07/19/2023] [Indexed: 08/25/2023]
Abstract
The pancreatic β cell, which produces insulin, is a terminally differentiated cell type that divides rarely. Consequently, the regenerative ability of β cells is limited and irreversible diabetes occurs after severe loss of β-cell function. In view of such poor regenerative capacity, considerable research efforts have been made to promote the expansion of functional insulin-producing cells as a regenerative therapy for diabetes. Here, we discuss recent findings regarding the robust expansion of functional mature islet cells both in vivo and ex vivo through MYCL-mediated reprogramming. We also describe the potential prospects for the application of reprogramming technologies to regenerative therapy and rejuvenation of islet cells.
Collapse
Affiliation(s)
- Michitada Hirano
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Yasuhiro Yamada
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan.
| |
Collapse
|
18
|
Rial SA, Shishani R, Cummings BP, Lim GE. Is 14-3-3 the Combination to Unlock New Pathways to Improve Metabolic Homeostasis and β-Cell Function? Diabetes 2023; 72:1045-1054. [PMID: 37471599 PMCID: PMC10382651 DOI: 10.2337/db23-0094] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 05/10/2023] [Indexed: 07/22/2023]
Abstract
Since their discovery nearly five decades ago, molecular scaffolds belonging to the 14-3-3 protein family have been recognized as pleiotropic regulators of diverse cellular and physiological functions. With their ability to bind to proteins harboring specific serine and threonine phosphorylation motifs, 14-3-3 proteins can interact with and influence the function of docking proteins, enzymes, transcription factors, and transporters that have essential roles in metabolism and glucose homeostasis. Here, we will discuss the regulatory functions of 14-3-3 proteins that will be of great interest to the fields of metabolism, pancreatic β-cell biology, and diabetes. We first describe how 14-3-3 proteins play a central role in glucose and lipid homeostasis by modulating key pathways of glucose uptake, glycolysis, oxidative phosphorylation, and adipogenesis. This is followed by a discussion of the contributions of 14-3-3 proteins to calcium-dependent exocytosis and how this relates to insulin secretion from β-cells. As 14-3-3 proteins are major modulators of apoptosis and cell cycle progression, we will explore if 14-3-3 proteins represent a viable target for promoting β-cell regeneration and discuss the feasibility of targeting 14-3-3 proteins to treat metabolic diseases such as diabetes. ARTICLE HIGHLIGHTS 14-3-3 proteins are ubiquitously expressed scaffolds with multiple roles in glucose homeostasis and metabolism. 14-3-3ζ regulates adipogenesis via distinct mechanisms and is required for postnatal adiposity and adipocyte function. 14-3-3ζ controls glucose-stimulated insulin secretion from pancreatic β-cells by regulating mitochondrial function and ATP synthesis as well as facilitating cross talk between β-cells and α-cells.
Collapse
Affiliation(s)
- Sabri A. Rial
- Department of Medicine, University of Montreal, Montreal, Quebec, Canada
- Cardiometabolic Axis, University of Montreal Hospital Research Center (CRCHUM), Montreal, Quebec, Canada
| | - Rahaf Shishani
- Department of Surgery, Center for Alimentary and Metabolic Sciences, School of Medicine, University of California, Davis, Sacramento, CA
| | - Bethany P. Cummings
- Department of Surgery, Center for Alimentary and Metabolic Sciences, School of Medicine, University of California, Davis, Sacramento, CA
| | - Gareth E. Lim
- Department of Medicine, University of Montreal, Montreal, Quebec, Canada
- Cardiometabolic Axis, University of Montreal Hospital Research Center (CRCHUM), Montreal, Quebec, Canada
| |
Collapse
|
19
|
Kimani CN, Reuter H, Kotzé SH, Muller CJF. Regeneration of Pancreatic Beta Cells by Modulation of Molecular Targets Using Plant-Derived Compounds: Pharmacological Mechanisms and Clinical Potential. Curr Issues Mol Biol 2023; 45:6216-6245. [PMID: 37623211 PMCID: PMC10453321 DOI: 10.3390/cimb45080392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/20/2023] [Accepted: 07/22/2023] [Indexed: 08/26/2023] Open
Abstract
Type 2 diabetes (T2D) is characterized by pancreatic beta-cell dysfunction, increased cell death and loss of beta-cell mass despite chronic treatment. Consequently, there has been growing interest in developing beta cell-centered therapies. Beta-cell regeneration is mediated by augmented beta-cell proliferation, transdifferentiation of other islet cell types to functional beta-like cells or the reprograming of beta-cell progenitors into fully differentiated beta cells. This mediation is orchestrated by beta-cell differentiation transcription factors and the regulation of the cell cycle machinery. This review investigates the beta-cell regenerative potential of antidiabetic plant extracts and phytochemicals. Various preclinical studies, including in vitro, in vivo and ex vivo studies, are highlighted. Further, the potential regenerative mechanisms and the intra and extracellular mediators that are of significance are discussed. Also, the potential of phytochemicals to translate into regenerative therapies for T2D patients is highlighted, and some suggestions regarding future perspectives are made.
Collapse
Affiliation(s)
- Clare Njoki Kimani
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council (SAMRC), Cape Town 7505, South Africa;
- Division of Clinical Pharmacology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town 7505, South Africa
| | - Helmuth Reuter
- Division of Clinical Pharmacology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town 7505, South Africa
| | - Sanet Henriët Kotzé
- Division of Clinical Anatomy, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town 7505, South Africa
- Division of Anatomy, Department of Biomedical Sciences, School of Veterinary Medicine, Ross University, Basseterre P.O. Box 334, Saint Kitts and Nevis
| | - Christo John Fredrick Muller
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council (SAMRC), Cape Town 7505, South Africa;
- Centre for Cardio-Metabolic Research in Africa, Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Stellenbosch 7600, South Africa
- Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa 3886, South Africa
| |
Collapse
|
20
|
Su Y, Ye L, Hu C, Zhang Y, Liu J, Shao L. Periodontitis as a promoting factor of T2D: current evidence and mechanisms. Int J Oral Sci 2023; 15:25. [PMID: 37321994 DOI: 10.1038/s41368-023-00227-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 04/12/2023] [Accepted: 04/26/2023] [Indexed: 06/17/2023] Open
Abstract
Periodontitis is an infectious disease caused by an imbalance between the local microbiota and host immune response. Epidemiologically, periodontitis is closely related to the occurrence, development, and poor prognosis of T2D and is recognized as a potential risk factor for T2D. In recent years, increasing attention has been given to the role of the virulence factors produced by disorders of the subgingival microbiota in the pathological mechanism of T2D, including islet β-cell dysfunction and insulin resistance (IR). However, the related mechanisms have not been well summarized. This review highlights periodontitis-derived virulence factors, reviews how these stimuli directly or indirectly regulate islet β-cell dysfunction. The mechanisms by which IR is induced in insulin-targeting tissues (the liver, visceral adipose tissue, and skeletal muscle) are explained, clarifying the influence of periodontitis on the occurrence and development of T2D. In addition, the positive effects of periodontal therapy on T2D are overviewed. Finally, the limitations and prospects of the current research are discussed. In summary, periodontitis is worthy of attention as a promoting factor of T2D. Understanding on the effect of disseminated periodontitis-derived virulence factors on the T2D-related tissues and cells may provide new treatment options for reducing the risk of T2D associated with periodontitis.
Collapse
Affiliation(s)
- Yuan Su
- Stomatology Center, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, China
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Leilei Ye
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Chen Hu
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yanli Zhang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Jia Liu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Longquan Shao
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China.
| |
Collapse
|
21
|
Sugawara H, Imai J, Yamamoto J, Izumi T, Kawana Y, Endo A, Kohata M, Seike J, Kubo H, Komamura H, Munakata Y, Asai Y, Hosaka S, Sawada S, Kodama S, Takahashi K, Kaneko K, Katagiri H. A highly sensitive strategy for monitoring real-time proliferation of targeted cell types in vivo. Nat Commun 2023; 14:3253. [PMID: 37316473 DOI: 10.1038/s41467-023-38897-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 05/22/2023] [Indexed: 06/16/2023] Open
Abstract
Cell proliferation processes play pivotal roles in timely adaptation to many biological situations. Herein, we establish a highly sensitive and simple strategy by which time-series showing the proliferation of a targeted cell type can be quantitatively monitored in vivo in the same individuals. We generate mice expressing a secreted type of luciferase only in cells producing Cre under the control of the Ki67 promoter. Crossing these with tissue-specific Cre-expressing mice allows us to monitor the proliferation time course of pancreatic β-cells, which are few in number and weakly proliferative, by measuring plasma luciferase activity. Physiological time courses, during obesity development, pregnancy and juvenile growth, as well as diurnal variation, of β-cell proliferation, are clearly detected. Moreover, this strategy can be utilized for highly sensitive ex vivo screening for proliferative factors for targeted cells. Thus, these technologies may contribute to advancements in broad areas of biological and medical research.
Collapse
Affiliation(s)
- Hiroto Sugawara
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Junta Imai
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan.
| | - Junpei Yamamoto
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tomohito Izumi
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yohei Kawana
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Akira Endo
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masato Kohata
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Junro Seike
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Haremaru Kubo
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hiroshi Komamura
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yuichiro Munakata
- Division of Metabolism and Diabetes, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Yoichiro Asai
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shinichiro Hosaka
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shojiro Sawada
- Division of Metabolism and Diabetes, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Shinjiro Kodama
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kei Takahashi
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Keizo Kaneko
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hideki Katagiri
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
22
|
Khajavi N, Beck A, Riçku K, Beyerle P, Jacob K, Syamsul SF, Belkacemi A, Reinach PS, Schreier PC, Salah H, Popp T, Novikoff A, Breit A, Chubanov V, Müller TD, Zierler S, Gudermann T. TRPM7 kinase is required for insulin production and compensatory islet responses during obesity. JCI Insight 2023; 8:163397. [PMID: 36574297 PMCID: PMC9977431 DOI: 10.1172/jci.insight.163397] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
Most overweight individuals do not develop diabetes due to compensatory islet responses to restore glucose homeostasis. Therefore, regulatory pathways that promote β cell compensation are potential targets for treatment of diabetes. The transient receptor potential cation channel subfamily M member 7 protein (TRPM7), harboring a cation channel and a serine/threonine kinase, has been implicated in controlling cell growth and proliferation. Here, we report that selective deletion of Trpm7 in β cells disrupted insulin secretion and led to progressive glucose intolerance. We indicate that the diminished insulinotropic response in β cell-specific Trpm7-knockout mice was caused by decreased insulin production because of impaired enzymatic activity of this protein. Accordingly, high-fat-fed mice with a genetic loss of TRPM7 kinase activity displayed a marked glucose intolerance accompanied by hyperglycemia. These detrimental glucoregulatory effects were engendered by reduced compensatory β cell responses because of mitigated protein kinase B (AKT)/ERK signaling. Collectively, our data identify TRPM7 kinase as a potentially novel regulator of insulin synthesis, β cell dynamics, and glucose homeostasis under obesogenic diet.
Collapse
Affiliation(s)
- Noushafarin Khajavi
- Walther Straub Institute of Pharmacology and Toxicology, LMU Munich, Munich, Germany
| | - Andreas Beck
- Institute of Experimental and Clinical Pharmacology and Toxicology, Saarland University, Homburg, Germany
| | - Klea Riçku
- Walther Straub Institute of Pharmacology and Toxicology, LMU Munich, Munich, Germany
| | - Philipp Beyerle
- Walther Straub Institute of Pharmacology and Toxicology, LMU Munich, Munich, Germany
| | - Katharina Jacob
- Walther Straub Institute of Pharmacology and Toxicology, LMU Munich, Munich, Germany
| | - Sabrina F. Syamsul
- Walther Straub Institute of Pharmacology and Toxicology, LMU Munich, Munich, Germany
| | - Anouar Belkacemi
- Institute of Experimental and Clinical Pharmacology and Toxicology, Saarland University, Homburg, Germany
| | - Peter S. Reinach
- Wenzhou Medical University, Ophthalmology Department, Wenzhou, China
| | - Pascale C.F. Schreier
- Walther Straub Institute of Pharmacology and Toxicology, LMU Munich, Munich, Germany
| | - Houssein Salah
- Institute of Experimental and Clinical Pharmacology and Toxicology, Saarland University, Homburg, Germany
| | - Tanja Popp
- Bundeswehr Institute of Radiobiology, Munich, Germany
| | - Aaron Novikoff
- Institute of Diabetes and Obesity, Helmholtz Center Munich, Neuherberg, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Andreas Breit
- Walther Straub Institute of Pharmacology and Toxicology, LMU Munich, Munich, Germany
| | - Vladimir Chubanov
- Walther Straub Institute of Pharmacology and Toxicology, LMU Munich, Munich, Germany
| | - Timo D. Müller
- Institute of Diabetes and Obesity, Helmholtz Center Munich, Neuherberg, Germany.,German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Susanna Zierler
- Walther Straub Institute of Pharmacology and Toxicology, LMU Munich, Munich, Germany.,Institute of Pharmacology, Medical Faculty, Johannes Kepler University Linz, Linz, Austria
| | - Thomas Gudermann
- Walther Straub Institute of Pharmacology and Toxicology, LMU Munich, Munich, Germany.,German Center for Lung Research, Munich, Germany
| |
Collapse
|
23
|
Tremmel DM, Mikat AE, Gupta S, Mitchell SA, Curran AM, Menadue JA, Odorico JS, Sackett SD. Validating expression of beta cell maturation-associated genes in human pancreas development. Front Cell Dev Biol 2023; 11:1103719. [PMID: 36846594 PMCID: PMC9945361 DOI: 10.3389/fcell.2023.1103719] [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: 11/20/2022] [Accepted: 01/16/2023] [Indexed: 02/04/2023] Open
Abstract
The identification of genes associated with human pancreatic beta cell maturation could stimulate a better understanding of normal human islet development and function, be informative for improving stem cell-derived islet (SC-islet) differentiation, and facilitate the sorting of more mature beta cells from a pool of differentiated cells. While several candidate factors to mark beta cell maturation have been identified, much of the data supporting these markers come from animal models or differentiated SC-islets. One such marker is Urocortin-3 (UCN3). In this study, we provide evidence that UCN3 is expressed in human fetal islets well before the acquisition of functional maturation. When SC-islets expressing significant levels of UCN3 were generated, the cells did not exhibit glucose-stimulated insulin secretion, indicating that UCN3 expression is not correlated with functional maturation in these cells. We utilized our tissue bank and SC-islet resources to test an array of other candidate maturation-associated genes, and identified CHGB, G6PC2, FAM159B, GLUT1, IAPP and ENTPD3 as markers with expression patterns that correlate developmentally with the onset of functional maturation in human beta cells. We also find that human beta cell expression of ERO1LB, HDAC9, KLF9, and ZNT8 does not change between fetal and adult stages.
Collapse
Affiliation(s)
- Daniel M. Tremmel
- *Correspondence: Daniel M. Tremmel, ; Sara Dutton Sackett, ; Jon S. Odorico,
| | - Anna E. Mikat
- University of Wisconsin-Madison, Department of Surgery, Transplantation Division, Madison, WI, United States
| | - Sakar Gupta
- University of Wisconsin-Madison, Department of Surgery, Transplantation Division, Madison, WI, United States
| | - Samantha A. Mitchell
- University of Wisconsin-Madison, Department of Surgery, Transplantation Division, Madison, WI, United States
| | - Andrew M. Curran
- University of Wisconsin-Madison, Department of Surgery, Transplantation Division, Madison, WI, United States
| | - Jenna A. Menadue
- University of Wisconsin-Madison, Department of Surgery, Transplantation Division, Madison, WI, United States
| | - Jon S. Odorico
- *Correspondence: Daniel M. Tremmel, ; Sara Dutton Sackett, ; Jon S. Odorico,
| | - Sara Dutton Sackett
- *Correspondence: Daniel M. Tremmel, ; Sara Dutton Sackett, ; Jon S. Odorico,
| |
Collapse
|
24
|
Fernández-Ruiz R, Gasa R. Evaluation of the Effects of CCN4 on Pancreatic Beta Cell Proliferation. Methods Mol Biol 2023; 2582:191-208. [PMID: 36370351 DOI: 10.1007/978-1-0716-2744-0_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Expanding the number of insulin-producing beta cells through reactivation of their replication has been proposed as a therapy to prevent or delay the appearance of diabetes. Using antibody arrays, we identified CCN4/Wisp1 as a circulating factor enriched in preweaning mice, a period in which beta cells exhibit a dramatic increase in number. This finding led us to investigate the involvement of CCN4 in beta cell proliferation. We demonstrated that CCN4 promotes adult beta cell proliferation in vitro in cultured isolated islets, and in vivo in islets transplanted into the anterior chamber of the eye. In this chapter, we present the methodology that was used to study proliferation in both settings.
Collapse
Affiliation(s)
- Rebeca Fernández-Ruiz
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
| | - Rosa Gasa
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain.
| |
Collapse
|
25
|
Yang F, Wang J, Qu K, Wei H, Song Z, Xu H, Wang Z, Chen Y. Responses of INS-1 cells to glucose stimulation patterns. BIOMATERIALS ADVANCES 2023; 144:213199. [PMID: 36424275 DOI: 10.1016/j.bioadv.2022.213199] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 10/30/2022] [Accepted: 11/13/2022] [Indexed: 06/16/2023]
Abstract
Diabetes has become a major public health problem in the world for many years, and it is driving us to probe into its complex mechanism of insulin secretion in pancreatic β cells. The nanoscale resolution characterization of pancreatic β cells in response to glucose led to insights into diverse mechanical and functional processes at the single cell level. Recent advances allowed the direct observations of cytoskeleton dynamics which were quantitatively determined. Here, we firstly performed the glucose stimulation with multiple physiologically relevant glucose patterns. Atomic force microscopy (AFM) produced high spatial resolution mechanical images together with the insulin secretions linking the physical interactions to the biochemical process of INS-1 cells. Altered material properties of the INS-1 cells revealed the regulation of multiple glucose stimulation patterns. Rapidly responded to high glucose (HG), INS-1 cells presented the unique meshing networks of elasticities. The decreases of Young's modulus (YM) and insulin secretion suggested that mechanical changes affected the insulin release. Furthermore, the frequency and gradient of glucose patterns induced nanomechanical and secreting changes of the INS-1 cells and gained the knowledge on the potential controllability of glucose. The relationships between the cellular mechanics and insulin secretion of INS-1 cells could contribute to establish a mechanical cell model for the study of β cells in diabetes. The results also indicated the cell mechanics as promising mechanical biomarkers for β cells, and promoted the understanding of specific mechanical mechanism of glucose regulation, which lighted on the further application of functional glucose regulation in therapy.
Collapse
Affiliation(s)
- Fan Yang
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun 130022, China; Ministry of Education Key Laboratory for Cross-Scale Micro and Nano Manufacturing, Changchun University of Science and Technology, Changchun 130022, China; Jilin Provincial Key Laboratory for Multi-information Nano Detection & Handling of Single Cells, Changchun University of Science and Technology, Changchun 130022, China
| | - Jiajia Wang
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun 130022, China; Ministry of Education Key Laboratory for Cross-Scale Micro and Nano Manufacturing, Changchun University of Science and Technology, Changchun 130022, China; Jilin Provincial Key Laboratory for Multi-information Nano Detection & Handling of Single Cells, Changchun University of Science and Technology, Changchun 130022, China
| | - Kaige Qu
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun 130022, China; Ministry of Education Key Laboratory for Cross-Scale Micro and Nano Manufacturing, Changchun University of Science and Technology, Changchun 130022, China; Jilin Provincial Key Laboratory for Multi-information Nano Detection & Handling of Single Cells, Changchun University of Science and Technology, Changchun 130022, China
| | - Huimiao Wei
- Changli Nano Biotechnology Ltd, Changchun 130022, China
| | - Zhengxun Song
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun 130022, China; Ministry of Education Key Laboratory for Cross-Scale Micro and Nano Manufacturing, Changchun University of Science and Technology, Changchun 130022, China; Jilin Provincial Key Laboratory for Multi-information Nano Detection & Handling of Single Cells, Changchun University of Science and Technology, Changchun 130022, China
| | - Hongmei Xu
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun 130022, China; Ministry of Education Key Laboratory for Cross-Scale Micro and Nano Manufacturing, Changchun University of Science and Technology, Changchun 130022, China; Jilin Provincial Key Laboratory for Multi-information Nano Detection & Handling of Single Cells, Changchun University of Science and Technology, Changchun 130022, China
| | - Zuobin Wang
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun 130022, China; Ministry of Education Key Laboratory for Cross-Scale Micro and Nano Manufacturing, Changchun University of Science and Technology, Changchun 130022, China; Jilin Provincial Key Laboratory for Multi-information Nano Detection & Handling of Single Cells, Changchun University of Science and Technology, Changchun 130022, China; JR3CN & IRAC, University of Bedfordshire, Luton LU1 3JU, UK.
| | - Yujuan Chen
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun 130022, China; Ministry of Education Key Laboratory for Cross-Scale Micro and Nano Manufacturing, Changchun University of Science and Technology, Changchun 130022, China; Jilin Provincial Key Laboratory for Multi-information Nano Detection & Handling of Single Cells, Changchun University of Science and Technology, Changchun 130022, China.
| |
Collapse
|
26
|
Melnik BC, Schmitz G. Milk Exosomal microRNAs: Postnatal Promoters of β Cell Proliferation but Potential Inducers of β Cell De-Differentiation in Adult Life. Int J Mol Sci 2022; 23:ijms231911503. [PMID: 36232796 PMCID: PMC9569743 DOI: 10.3390/ijms231911503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/15/2022] [Accepted: 09/20/2022] [Indexed: 11/16/2022] Open
Abstract
Pancreatic β cell expansion and functional maturation during the birth-to-weaning period is driven by epigenetic programs primarily triggered by growth factors, hormones, and nutrients provided by human milk. As shown recently, exosomes derived from various origins interact with β cells. This review elucidates the potential role of milk-derived exosomes (MEX) and their microRNAs (miRs) on pancreatic β cell programming during the postnatal period of lactation as well as during continuous cow milk exposure of adult humans to bovine MEX. Mechanistic evidence suggests that MEX miRs stimulate mTORC1/c-MYC-dependent postnatal β cell proliferation and glycolysis, but attenuate β cell differentiation, mitochondrial function, and insulin synthesis and secretion. MEX miR content is negatively affected by maternal obesity, gestational diabetes, psychological stress, caesarean delivery, and is completely absent in infant formula. Weaning-related disappearance of MEX miRs may be the critical event switching β cells from proliferation to TGF-β/AMPK-mediated cell differentiation, whereas continued exposure of adult humans to bovine MEX miRs via intake of pasteurized cow milk may reverse β cell differentiation, promoting β cell de-differentiation. Whereas MEX miR signaling supports postnatal β cell proliferation (diabetes prevention), persistent bovine MEX exposure after the lactation period may de-differentiate β cells back to the postnatal phenotype (diabetes induction).
Collapse
Affiliation(s)
- Bodo C. Melnik
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, D-49076 Osnabrück, Germany
- Correspondence: ; Tel.: +49-52-4198-8060
| | - Gerd Schmitz
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital of Regensburg, University of Regensburg, D-93053 Regensburg, Germany
| |
Collapse
|
27
|
Li Y, Li Y, Chen N, Feng L, Gao J, Zeng N, He Z, Gong Q. Icariside II Exerts Anti-Type 2 Diabetic Effect by Targeting PPARα/γ: Involvement of ROS/NF-κB/IRS1 Signaling Pathway. Antioxidants (Basel) 2022; 11:antiox11091705. [PMID: 36139776 PMCID: PMC9495514 DOI: 10.3390/antiox11091705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/26/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a multisystem and complex metabolic disorder which is associated with insulin resistance and impairments of pancreatic β-cells. Previous studies have shown that icariside II (ICS II), one of the main active ingredients of Herba Epimedii, exerts potent anti-inflammatory and anti-oxidative properties. In this study, we investigated whether ICS II exerted anti-T2DM profile and further explored its possible underlying mechanism both in vivo and in vitro. db/db mice were administered ICS II (10, 20, 40 mg·kg−1) for 7 weeks. We found that ICS II dose-dependently attenuated hyperglycemia and dyslipidemia, as well as inhibited hepatic steatosis and islet architecture damage in db/db mice. Moreover, ICS II not only dramatically reduced inflammatory cytokines and oxidative stress, but also up-regulated PPARα/γ protein expressions, phosphorylation of Akt, GSK3β and IR, meanwhile, down-regulated phosphorylation of NF-κB(p65) and IRS1 in db/db mice. In palmitic acid (PA)-treated HepG2 or MIN6 cells, ICS II (5−20 μM) concentration-dependently promoted the cell viability via mediating PPARα/γ/NF-κB signaling pathway. PPARα/γ knockout by CRISPR-Cas9 system partly abolished the protective effects of ICS II on HepG2 or MIN6 cells following PA insults. These findings reveal that ICS II effectively confer anti-T2DM property by targeting PPARα/γ through mediation of ROS/NF-κB/IRS1 signaling pathway.
Collapse
Affiliation(s)
- Yiqi Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, School of Pharmacy, Zunyi Medical University, Zunyi 563000, China
| | - Yeli Li
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, School of Pharmacy, Zunyi Medical University, Zunyi 563000, China
| | - Nana Chen
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, School of Pharmacy, Zunyi Medical University, Zunyi 563000, China
| | - Linying Feng
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, School of Pharmacy, Zunyi Medical University, Zunyi 563000, China
| | - Jianmei Gao
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, School of Pharmacy, Zunyi Medical University, Zunyi 563000, China
| | - Nan Zeng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Zhixu He
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine, Zunyi Medical University, Zunyi 563000, China
| | - Qihai Gong
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, School of Pharmacy, Zunyi Medical University, Zunyi 563000, China
- Correspondence: ; Tel./Fax: +86-851-286-423-03
| |
Collapse
|
28
|
Hospodiuk-Karwowski M, Chi K, Pritchard J, Catchmark JM. Vascularized pancreas-on-a-chip device produced using a printable simulated extracellular matrix. Biomed Mater 2022; 17. [PMID: 36001993 DOI: 10.1088/1748-605x/ac8c74] [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: 04/20/2022] [Accepted: 08/24/2022] [Indexed: 11/12/2022]
Abstract
The extracellular matrix (ECM) influences cellular behavior, function, and fate. The ECM surrounding Langerhans islets has not been investigated in detail to explain its role in the development and maturation of pancreatic β-cells. Herein, a complex combination of the simulated ECM (sECM) has been examined with a comprehensive analysis of cell response and a variety of controls. The most promising results were obtained from group containing fibrin, collagen type I, Matrigel®, hyaluronic acid, methylcellulose, and two compounds of functionalized, ionically crosslinking bacterial cellulose (sECMbc). Even though the cell viability was not significantly impacted, the performance of group of sECMbc showed 2 to 4x higher sprouting number and length, 2 to 4x higher insulin secretion in static conditions, and 2 to 10x higher gene expression of VEGF-A, Endothelin-1, and NOS3 than the control group of fibrin matrix (sECMf). Each material was tested in a hydrogel-based, perfusable, pancreas-on-a-chip device and the best group - sECMbc has been tested with the drug Sunitinib to show the extended possibilities of the device for both diabetes-like screening as well as PDAC chemotherapeutics screening for potential personal medicine approach. It proved its functionality in 7 days dynamic culture and is suitable as a physiological tissue model. Moreover, the device with the pancreatic-like spheroids was 3D bioprintable and perfusable.
Collapse
Affiliation(s)
- Monika Hospodiuk-Karwowski
- Department of Agricultural and Biological Engineering, The Pennsylvania State University, 201 Old Main, University Park, Pennsylvania, 16802-1503, UNITED STATES
| | - Kai Chi
- Department of Agricultural and Biological Engineering, The Pennsylvania State University, 201 Old Main, University Park, Pennsylvania, 16802-1503, UNITED STATES
| | - Justin Pritchard
- Biomedical Engineering Department, The Pennsylvania State University, 201 Old Main, University Park, Pennsylvania, 16802-1503, UNITED STATES
| | - Jeffrey M Catchmark
- Department of Agricultural and Biological Engineering, The Pennsylvania State University, 201 Old Main, University Park, Pennsylvania, 16802-1503, UNITED STATES
| |
Collapse
|
29
|
Wang P, Karakose E, Argmann C, Wang H, Balev M, Brody RI, Rivas HG, Liu X, Wood O, Liu H, Choleva L, Hasson D, Bernstein E, Paulo JA, Scott DK, Lambertini L, DeCaprio JA, Stewart AF. Disrupting the DREAM complex enables proliferation of adult human pancreatic β cells. J Clin Invest 2022; 132:e157086. [PMID: 35700053 PMCID: PMC9337832 DOI: 10.1172/jci157086] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 06/09/2022] [Indexed: 11/17/2022] Open
Abstract
Resistance to regeneration of insulin-producing pancreatic β cells is a fundamental challenge for type 1 and type 2 diabetes. Recently, small molecule inhibitors of the kinase DYRK1A have proven effective in inducing adult human β cells to proliferate, but their detailed mechanism of action is incompletely understood. We interrogated our human insulinoma and β cell transcriptomic databases seeking to understand why β cells in insulinomas proliferate, while normal β cells do not. This search reveals the DREAM complex as a central regulator of quiescence in human β cells. The DREAM complex consists of a module of transcriptionally repressive proteins that assemble in response to DYRK1A kinase activity, thereby inducing and maintaining cellular quiescence. In the absence of DYRK1A, DREAM subunits reassemble into the pro-proliferative MMB complex. Here, we demonstrate that small molecule DYRK1A inhibitors induce human β cells to replicate by converting the repressive DREAM complex to its pro-proliferative MMB conformation.
Collapse
Affiliation(s)
- Peng Wang
- Diabetes Obesity Metabolism Institute
- Department of Medicine, and
| | - Esra Karakose
- Diabetes Obesity Metabolism Institute
- Department of Medicine, and
| | - Carmen Argmann
- Department of Genetics and Genomic Sciences, The Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | | | - Rachel I. Brody
- Department of Pathology, The Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Hembly G. Rivas
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- The Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Xinyue Liu
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Olivia Wood
- Diabetes Obesity Metabolism Institute
- Department of Medicine, and
| | - Hongtao Liu
- Diabetes Obesity Metabolism Institute
- Department of Medicine, and
| | - Lauryn Choleva
- Diabetes Obesity Metabolism Institute
- Department of Pediatrics
| | - Dan Hasson
- The Tisch Cancer Institute
- Department of Oncological Sciences
- Bioinformatics for Next Generation Sequencing (BiNGS) Shared Resource Facility, and
| | - Emily Bernstein
- The Tisch Cancer Institute
- Department of Oncological Sciences
- The Graduate School of Biomedical Sciences, The Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Joao A. Paulo
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Donald K. Scott
- Diabetes Obesity Metabolism Institute
- Department of Medicine, and
| | - Luca Lambertini
- Diabetes Obesity Metabolism Institute
- Department of Medicine, and
| | - James A. DeCaprio
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- The Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | |
Collapse
|
30
|
Katz LS, Brill G, Zhang P, Kumar A, Baumel-Alterzon S, Honig LB, Gómez-Banoy N, Karakose E, Tanase M, Doridot L, Alvarsson A, Davenport B, Wang P, Lambertini L, Stanley SA, Homann D, Stewart AF, Lo JC, Herman MA, Garcia-Ocaña A, Scott DK. Maladaptive positive feedback production of ChREBPβ underlies glucotoxic β-cell failure. Nat Commun 2022; 13:4423. [PMID: 35908073 PMCID: PMC9339008 DOI: 10.1038/s41467-022-32162-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 07/18/2022] [Indexed: 01/05/2023] Open
Abstract
Preservation and expansion of β-cell mass is a therapeutic goal for diabetes. Here we show that the hyperactive isoform of carbohydrate response-element binding protein (ChREBPβ) is a nuclear effector of hyperglycemic stress occurring in β-cells in response to prolonged glucose exposure, high-fat diet, and diabetes. We show that transient positive feedback induction of ChREBPβ is necessary for adaptive β-cell expansion in response to metabolic challenges. Conversely, chronic excessive β-cell-specific overexpression of ChREBPβ results in loss of β-cell identity, apoptosis, loss of β-cell mass, and diabetes. Furthermore, β-cell "glucolipotoxicity" can be prevented by deletion of ChREBPβ. Moreover, ChREBPβ-mediated cell death is mitigated by overexpression of the alternate CHREBP gene product, ChREBPα, or by activation of the antioxidant Nrf2 pathway in rodent and human β-cells. We conclude that ChREBPβ, whether adaptive or maladaptive, is an important determinant of β-cell fate and a potential target for the preservation of β-cell mass in diabetes.
Collapse
Affiliation(s)
- Liora S Katz
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1152, New York, 10029, USA
| | - Gabriel Brill
- Pharmacologic Sciences Department, Stony Brook University, Stony Brook, NY, USA
| | - Pili Zhang
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1152, New York, 10029, USA
| | - Anil Kumar
- Metabolic Phenotyping Core, University of Utah, 15N 2030 E, 585, Radiobiology building, Room 151, Salt Lake City, UT, 84112, USA
| | - Sharon Baumel-Alterzon
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1152, New York, 10029, USA
| | - Lee B Honig
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1152, New York, 10029, USA
| | - Nicolás Gómez-Banoy
- Weill Center for Metabolic Health and Division of Cardiology, Department of Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Esra Karakose
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1152, New York, 10029, USA
| | - Marius Tanase
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1152, New York, 10029, USA
| | - Ludivine Doridot
- Institut Cochin, Université de Paris, INSERM, CNRS, F-75014, Paris, France
| | - Alexandra Alvarsson
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1152, New York, 10029, USA
- Alpenglow Biosciences, Inc., 98103, Seattle, WA, USA
| | - Bennett Davenport
- 12800 East 19th Ave, Anschutz Medical Campus, Room P18-9403, University of Colorado, Aurora, CO, 80045, USA
| | - Peng Wang
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1152, New York, 10029, USA
| | - Luca Lambertini
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1152, New York, 10029, USA
| | - Sarah A Stanley
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1152, New York, 10029, USA
| | - Dirk Homann
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1152, New York, 10029, USA
| | - Andrew F Stewart
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1152, New York, 10029, USA
| | - James C Lo
- Weill Center for Metabolic Health and Division of Cardiology, Department of Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Mark A Herman
- Division of Endocrinology and Metabolism and Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA
- Section of Diabetes, Endocrinology, and Metabolism, Baylor College of Medicine, One Baylor Plaza, MS: 185, R614, 77030, Houston, TX, USA
| | - Adolfo Garcia-Ocaña
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1152, New York, 10029, USA
| | - Donald K Scott
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1152, New York, 10029, USA.
| |
Collapse
|
31
|
Ji Z, Lu M, Xie H, Yuan H, Chen Q. β cell regeneration and novel strategies for treatment of diabetes (Review). Biomed Rep 2022; 17:72. [DOI: 10.3892/br.2022.1555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 03/14/2022] [Indexed: 11/06/2022] Open
Affiliation(s)
- Zengyang Ji
- Department of Endocrinology, Changxing County Hospital of Traditional Chinese Medicine, Huzhou, Zhejiang 313199, P.R. China
| | - Min Lu
- Department of Endocrinology, Changxing County Hospital of Traditional Chinese Medicine, Huzhou, Zhejiang 313199, P.R. China
| | - Huanhuan Xie
- Department of Endocrinology, Changxing County Hospital of Traditional Chinese Medicine, Huzhou, Zhejiang 313199, P.R. China
| | - Honggang Yuan
- Department of Endocrinology, Changxing County Hospital of Traditional Chinese Medicine, Huzhou, Zhejiang 313199, P.R. China
| | - Qing Chen
- Department of Endocrinology, Changxing County Hospital of Traditional Chinese Medicine, Huzhou, Zhejiang 313199, P.R. China
| |
Collapse
|
32
|
Abderrahmani A, Jacovetti C, Regazzi R. Lessons from neonatal β-cell epigenomic for diabetes prevention and treatment. Trends Endocrinol Metab 2022; 33:378-389. [PMID: 35382967 DOI: 10.1016/j.tem.2022.03.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/08/2022] [Accepted: 03/10/2022] [Indexed: 12/30/2022]
Abstract
Pancreatic β-cell expansion and functional maturation during the birth-to-weaning period plays an essential role in the adaptation of plasma insulin levels to metabolic needs. These events are driven by epigenetic programs triggered by growth factors, hormones, and nutrients. These mechanisms operating in the neonatal period can be at least in part reactivated in adult life to increase the functional β-cell mass and face conditions of increased insulin demand such as obesity or pregnancy. In this review, we will highlight the importance of studying these signaling pathways and epigenetic programs to understand the causes of different forms of diabetes and to permit the design of novel therapeutic strategies to prevent and treat this metabolic disorder affecting hundreds of millions of people worldwide.
Collapse
Affiliation(s)
- Amar Abderrahmani
- Universitéde Lille, CNRS, Centrale Lille, Université Polytechnique Hauts-de-France, UMR 8520 - IEMN, F-59000 Lille, France.
| | - Cécile Jacovetti
- Department of Fundamental Neuroscience, University of Lausanne, 1005 Lausanne, Switzerland
| | - Romano Regazzi
- Department of Fundamental Neuroscience, University of Lausanne, 1005 Lausanne, Switzerland; Department of Biomedical Science, University of Lausanne, 1005 Lausanne, Switzerland.
| |
Collapse
|
33
|
Basile G, Qadir MMF, Mauvais-Jarvis F, Vetere A, Shoba V, Modell AE, Pastori RL, Russ HA, Wagner BK, Dominguez-Bendala J. Emerging diabetes therapies: Bringing back the β-cells. Mol Metab 2022; 60:101477. [PMID: 35331962 PMCID: PMC8987999 DOI: 10.1016/j.molmet.2022.101477] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 03/11/2022] [Accepted: 03/14/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Stem cell therapies are finally coming of age as a viable alternative to pancreatic islet transplantation for the treatment of insulin-dependent diabetes. Several clinical trials using human embryonic stem cell (hESC)-derived β-like cells are currently underway, with encouraging preliminary results. Remaining challenges notwithstanding, these strategies are widely expected to reduce our reliance on human isolated islets for transplantation procedures, making cell therapies available to millions of diabetic patients. At the same time, advances in our understanding of pancreatic cell plasticity and the molecular mechanisms behind β-cell replication and regeneration have spawned a multitude of translational efforts aimed at inducing β-cell replenishment in situ through pharmacological means, thus circumventing the need for transplantation. SCOPE OF REVIEW We discuss here the current state of the art in hESC transplantation, as well as the parallel quest to discover agents capable of either preserving the residual mass of β-cells or inducing their proliferation, transdifferentiation or differentiation from progenitor cells. MAJOR CONCLUSIONS Stem cell-based replacement therapies in the mold of islet transplantation are already around the corner, but a permanent cure for type 1 diabetes will likely require the endogenous regeneration of β-cells aided by interventions to restore the immune balance. The promise of current research avenues and a strong pipeline of clinical trials designed to tackle these challenges bode well for the realization of this goal.
Collapse
Affiliation(s)
- G Basile
- Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - M M F Qadir
- Tulane University School of Medicine, New Orleans, LA, USA; Southeast Louisiana Veterans Affairs Medical Center, New Orleans, LA, USA
| | - F Mauvais-Jarvis
- Tulane University School of Medicine, New Orleans, LA, USA; Southeast Louisiana Veterans Affairs Medical Center, New Orleans, LA, USA
| | - A Vetere
- Broad Institute, Cambridge, MA, USA
| | - V Shoba
- Broad Institute, Cambridge, MA, USA
| | | | - R L Pastori
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - H A Russ
- Barbara Davis Center for Diabetes, Colorado University Anschutz Medical Campus, Aurora, CO, USA.
| | | | - J Dominguez-Bendala
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
34
|
Smigoc Schweiger D. Recent Advances in Immune-Based Therapies for Type 1 Diabetes. Horm Res Paediatr 2022; 96:631-645. [PMID: 35533645 DOI: 10.1159/000524866] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 04/18/2022] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Type 1 diabetes (T1D) is a chronic autoimmune disease characterized by progressive destruction of the pancreatic beta cells, leading to a lifelong dependence on insulin. It is associated with an increased morbidity and mortality from diabetes-related complications and a significant treatment burden. However, there has been substantial progress in therapeutic strategies that can affect the course of the disease. SUMMARY This review addresses advances in immunotherapy aimed at preserving residual beta-cell function in individuals with a recent onset of T1D and arresting the disease in pre-symptomatic stages. Recent and ongoing clinical trials have investigated the efficacy and safety of various immunotherapeutic strategies aimed at targeting several mechanisms of autoimmunity, which are thought to be important in disease pathogenesis, and therapies that also address beta-cell health. So far, T-cell-directed therapies that led to a favourable balance between T-effector cell depletion or modulation and preservation or expansion of regulatory T cells have shown the most success. Furthermore, regarding the timing of intervention, teplizumab was the first immunomodulatory agent to demonstrate a significant delay in disease progression in high-risk individuals before clinical onset. KEY MESSAGES As more targeted immune interventions with potentially fewer side effects are closer to the translation into clinical practice, some new challenges may need to be addressed. The use of combination approaches that include immunotherapeutic strategies targeting different aspects of the immune system and interventions that improve beta-cell health may be required, along with the use of individualized patient-tailored approaches, a move towards early intervention, and a focus on patient-reported outcome measures.
Collapse
Affiliation(s)
- Darja Smigoc Schweiger
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Children's Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
35
|
Zhao L, Xiong X, Liu L, Liang Q, Tong R, Feng X, Bai L, Shi J. Recent research and development of DYRK1A inhibitors. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2021.10.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
36
|
Wei T, Wei R, Hong T. Regeneration of β cells from cell phenotype conversion among the pancreatic endocrine cells. Chronic Dis Transl Med 2022; 8:1-4. [PMID: 35620156 PMCID: PMC9128562 DOI: 10.1002/cdt3.15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 01/12/2022] [Indexed: 11/29/2022] Open
Affiliation(s)
- Tianjiao Wei
- Department of Endocrinology and Metabolism Peking University Third Hospital Beijing 100191 China
| | - Rui Wei
- Department of Endocrinology and Metabolism Peking University Third Hospital Beijing 100191 China
| | - Tianpei Hong
- Department of Endocrinology and Metabolism Peking University Third Hospital Beijing 100191 China
| |
Collapse
|
37
|
Wang HL, Wang L, Zhao CY, Lan HY. Role of TGF-Beta Signaling in Beta Cell Proliferation and Function in Diabetes. Biomolecules 2022; 12:373. [PMID: 35327565 PMCID: PMC8945211 DOI: 10.3390/biom12030373] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 02/21/2022] [Accepted: 02/24/2022] [Indexed: 12/27/2022] Open
Abstract
Beta (β) cell dysfunction or loss is the common pathological feature in all types of diabetes mellitus (diabetes). Resolving the underlying mechanism may facilitate the treatment of diabetes by preserving the β cell population and function. It is known that TGF-β signaling plays diverse roles in β cell development, function, proliferation, apoptosis, and dedifferentiation. Inhibition of TGF-β signaling expands β cell lineage in the development. However, deletion of Tgfbr1 has no influence on insulin demand-induced but abolishes inflammation-induced β cell proliferation. Among canonical TGF-β signaling, Smad3 but not Smad2 is the predominant repressor of β cell proliferation in response to systemic insulin demand. Deletion of Smad3 simultaneously improves β cell function, apoptosis, and systemic insulin resistance with the consequence of eliminated overt diabetes in diabetic mouse models, revealing Smad3 as a key mediator and ideal therapeutic target for type-2 diabetes. However, Smad7 shows controversial effects on β cell proliferation and glucose homeostasis in animal studies. On the other hand, overexpression of Tgfb1 prevents β cells from autoimmune destruction without influence on β cell function. All these findings reveal the diverse regulatory roles of TGF-β signaling in β cell biology.
Collapse
Affiliation(s)
- Hong-Lian Wang
- Research Center for Integrative Medicine, The Affiliated Traditional Medicine Hospital of Southwest Medical University, Luzhou 646000, China; (H.-L.W.); (L.W.)
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Li Wang
- Research Center for Integrative Medicine, The Affiliated Traditional Medicine Hospital of Southwest Medical University, Luzhou 646000, China; (H.-L.W.); (L.W.)
| | - Chang-Ying Zhao
- Department of Endocrinology, The Affiliated Traditional Medicine Hospital of Southwest Medical University, Luzhou 646000, China;
| | - Hui-Yao Lan
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China
- Guangdong Academy of Sciences, Guangdong Provincial People’s Hospital Joint Research Laboratory on Immunological and Genetic Kidney Diseases, The Chinese University of Hong Kong, Hong Kong 999077, China
| |
Collapse
|
38
|
Hirano M, So Y, Tsunekawa S, Kabata M, Ohta S, Sagara H, Sankoda N, Taguchi J, Yamada Y, Ukai T, Kato M, Nakamura J, Ozawa M, Yamamoto T, Yamada Y. MYCL-mediated reprogramming expands pancreatic insulin-producing cells. Nat Metab 2022; 4:254-268. [PMID: 35145326 DOI: 10.1038/s42255-022-00530-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 01/11/2022] [Indexed: 11/09/2022]
Abstract
β cells have a limited capacity for regeneration, which predisposes towards diabetes. Here, we show that, of the MYC family members, Mycl plays a key role in proliferation of pancreatic endocrine cells. Genetic ablation of Mycl causes a reduction in the proliferation of pancreatic endocrine cells in neonatal mice. By contrast, the expression of Mycl in adult mice stimulates the proliferation of β and α cells, and the cells persist after withdrawal of Mycl expression. A subset of the expanded α cells give rise to insulin-producing cells after this withdrawal. Transient Mycl expression in vivo is sufficient to normalize the hyperglycaemia of diabetic mice. In vitro expression of Mycl similarly provokes active replication in islet cells, even in those from aged mice. Finally, we show that MYCL stimulates the division of human adult cadaveric islet cells. Our results demonstrate that the induction of Mycl alone expands the functional β-cell population, which may provide a regenerative strategy for β cells.
Collapse
Affiliation(s)
- Michitada Hirano
- Division of Stem Cell Pathology, Center for Experimental Medicine and Systems Biology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Yusei So
- Division of Stem Cell Pathology, Center for Experimental Medicine and Systems Biology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Shin Tsunekawa
- Division of Diabetes, Department of Internal Medicine, Aichi Medical University School of Medicine, Aichi, Japan
| | - Mio Kabata
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Sho Ohta
- Division of Stem Cell Pathology, Center for Experimental Medicine and Systems Biology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Hiroshi Sagara
- Medical Proteomics Laboratory, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Nao Sankoda
- Division of Stem Cell Pathology, Center for Experimental Medicine and Systems Biology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Jumpei Taguchi
- Division of Stem Cell Pathology, Center for Experimental Medicine and Systems Biology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Yosuke Yamada
- Department of Diagnostic Pathology, Kyoto University Hospital, Kyoto, Japan
| | - Tomoyo Ukai
- Division of Stem Cell Pathology, Center for Experimental Medicine and Systems Biology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Makoto Kato
- Division of Diabetes, Department of Internal Medicine, Aichi Medical University School of Medicine, Aichi, Japan
| | - Jiro Nakamura
- Division of Diabetes, Department of Internal Medicine, Aichi Medical University School of Medicine, Aichi, Japan
| | - Manabu Ozawa
- Laboratory of Reproductive Systems Biology, Center for Experimental Medicine and Systems Biology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Takuya Yamamoto
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- AMED-CREST, AMED, Tokyo, Japan
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
- Medical-risk Avoidance Based on iPS Cells Team, RIKEN Center for Advanced Intelligence Project (AIP), Kyoto, Japan
| | - Yasuhiro Yamada
- Division of Stem Cell Pathology, Center for Experimental Medicine and Systems Biology, Institute of Medical Science, University of Tokyo, Tokyo, Japan.
- AMED-CREST, AMED, Tokyo, Japan.
| |
Collapse
|
39
|
Shcheglova E, Blaszczyk K, Borowiak M. Mitogen Synergy: An Emerging Route to Boosting Human Beta Cell Proliferation. Front Cell Dev Biol 2022; 9:734597. [PMID: 35155441 PMCID: PMC8829426 DOI: 10.3389/fcell.2021.734597] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 08/24/2021] [Indexed: 12/11/2022] Open
Abstract
Decreased number and function of beta cells are a key aspect of diabetes mellitus (diabetes), a disease that remains an onerous global health problem. Means of restoring beta cell mass are urgently being sought as a potential cure for diabetes. Several strategies, such as de novo beta cell derivation via pluripotent stem cell differentiation or mature somatic cell transdifferentiation, have yielded promising results. Beta cell expansion is another promising strategy, rendered challenging by the very low proliferative capacity of beta cells. Many effective mitogens have been identified in rodents, but the vast majority do not have similar mitogenic effects in human beta cells. Extensive research has led to the identification of several human beta cell mitogens, but their efficacy and specificity remain insufficient. An approach based on the simultaneous application of several mitogens has recently emerged and can yield human beta cell proliferation rates of up to 8%. Here, we discuss recent advances in restoration of the beta cell population, focusing on mitogen synergy, and the contribution of RNA-sequencing (RNA-seq) to accelerating the elucidation of signaling pathways in proliferating beta cells and the discovery of novel mitogens. Together, these approaches have taken beta cell research up a level, bringing us closer to a cure for diabetes.
Collapse
Affiliation(s)
- Ekaterina Shcheglova
- Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
| | - Katarzyna Blaszczyk
- Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
| | - Malgorzata Borowiak
- Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
- *Correspondence: Malgorzata Borowiak, ;
| |
Collapse
|
40
|
Wang HL, Wei B, He HJ, Huang XR, Sheng JY, Chen XC, Wang L, Tan RZ, Li JC, Liu J, Yang SJ, Ma RCW, Lan HY. Smad3 deficiency improves islet-based therapy for diabetes and diabetic kidney injury by promoting β cell proliferation via the E2F3-dependent mechanism. Am J Cancer Res 2022; 12:379-395. [PMID: 34987651 PMCID: PMC8690916 DOI: 10.7150/thno.67034] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 10/29/2021] [Indexed: 12/15/2022] Open
Abstract
Rationale: Poor β cell proliferation is one of the detrimental factors hindering islet cell replacement therapy for patients with diabetes. Smad3 is an important transcriptional factor of TGF-β signaling and has been shown to promote diabetes by inhibiting β cell proliferation. Therefore, we hypothesize that Smad3-deficient islets may be a novel cell replacement therapy for diabetes. Methods: We examined this hypothesis in streptozocin-induced type-1 diabetic mice and type-2 diabetic db/db mice by transplanting Smad3 knockout (KO) and wild type (WT) islets under the renal capsule, respectively. The effects of Smad3KO versus WT islet replacement therapy on diabetes and diabetic kidney injury were examined. In addition, RNA-seq was applied to identify the downstream target gene underlying Smad3-regulated β cell proliferation in Smad3KO-db/db versus Smad3WT-db/db mouse islets. Results: Compared to Smad3WT islet therapy, treatment with Smad3KO islets produced a much better therapeutic effect on both type-1 and type-2 diabetes by significantly lowering serum levels of blood glucose and HbA1c and protected against diabetic kidney injuries by preventing an increase in serum creatinine and the development of proteinuria, mesangial matrix expansion, and fibrosis. These were associated with a significant increase in grafted β cell proliferation and blood insulin levels, resulting in improved glucose intolerance. Mechanistically, RNA-seq revealed that compared with Smad3WT-db/db mouse islets, deletion of Smad3 from db/db mouse islets markedly upregulated E2F3, a pivotal regulator of cell cycle G1/S entry. Further studies found that Smad3 could bind to the promoter of E2F3, and thus inhibit β cell proliferation via an E2F3-dependent mechanism as silencing E2F3 abrogated the proliferative effect on Smad3KO β cells. Conclusion: Smad3-deficient islet replacement therapy can significantly improve both type-1 and type-2 diabetes and protect against diabetic kidney injury, which is mediated by a novel mechanism of E2F3-dependent β cell proliferation.
Collapse
|
41
|
Title AC, Karsai M, Mir-Coll J, Grining ÖY, Rufer C, Sonntag S, Forschler F, Jawurek S, Klein T, Yesildag B. Evaluation of the Effects of Harmine on β-cell Function and Proliferation in Standardized Human Islets Using 3D High-Content Confocal Imaging and Automated Analysis. Front Endocrinol (Lausanne) 2022; 13:854094. [PMID: 35860702 PMCID: PMC9289187 DOI: 10.3389/fendo.2022.854094] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 05/10/2022] [Indexed: 01/09/2023] Open
Abstract
Restoration of β-cell mass through the induction of proliferation represents an attractive therapeutic approach for the treatment of diabetes. However, intact and dispersed primary islets suffer from rapidly deteriorating viability and function ex vivo, posing a significant challenge for their experimental use in proliferation studies. Here, we describe a novel method for the assessment of compound effects on β-cell proliferation and count using reaggregated primary human islets, or islet microtissues (MTs), which display homogeneous size and tissue architecture as well as robust and stable functionality and viability for 4 weeks in culture. We utilized this platform to evaluate the dose-dependent short- and long-term effects of harmine on β-cell proliferation and function. Following compound treatment and EdU incorporation, islet MTs were stained and confocal-imaged for DAPI (nuclear marker), NKX6.1 (β-cell marker), and EdU (proliferation marker), allowing automated 3D-analysis of number of total cells, β-cells, and proliferating β- and non-β-cells per islet MT. In parallel, insulin secretion, intracellular insulin and ATP contents, and Caspase 3/7 activity were analyzed to obtain a comprehensive overview of islet MT function and viability. We observed that 4-day harmine treatment increased β- and non-β-cell proliferation, NKX6.1 expression, and basal and stimulated insulin secretion in a dose-dependent manner, while fold-stimulation of secretion peaked at intermediate harmine doses. Interestingly, 15-day harmine treatment led to a general reduction in harmine's proliferative effects as well as altered dose-dependent trends. The described methodology provides a unique tool for in vitro high-throughput evaluation of short- and long-term changes in human β-cell proliferation, count and fraction along with a variety of functional parameters, in a representative 3D human islet model.
Collapse
Affiliation(s)
| | - Maria Karsai
- Diabetes Research, InSphero AG, Schlieren, Switzerland
| | - Joan Mir-Coll
- Diabetes Research, InSphero AG, Schlieren, Switzerland
| | | | - Chantal Rufer
- Diabetes Research, InSphero AG, Schlieren, Switzerland
| | | | | | - Sayro Jawurek
- Diabetes Research, InSphero AG, Schlieren, Switzerland
| | - Thomas Klein
- Department of Cardio-Metabolic Diseases, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| | - Burcak Yesildag
- Diabetes Research, InSphero AG, Schlieren, Switzerland
- *Correspondence: Burcak Yesildag,
| |
Collapse
|
42
|
Dinić S, Arambašić Jovanović J, Uskoković A, Mihailović M, Grdović N, Tolić A, Rajić J, Đorđević M, Vidaković M. Oxidative stress-mediated beta cell death and dysfunction as a target for diabetes management. Front Endocrinol (Lausanne) 2022; 13:1006376. [PMID: 36246880 PMCID: PMC9554708 DOI: 10.3389/fendo.2022.1006376] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 09/05/2022] [Indexed: 11/14/2022] Open
Abstract
The biggest drawback of a current diabetes therapy is the treatment of the consequences not the cause of the disease. Regardless of the diabetes type, preservation and recovery of functional pancreatic beta cells stands as the biggest challenge in the treatment of diabetes. Free radicals and oxidative stress are among the major mediators of autoimmune destruction of beta cells in type 1 diabetes (T1D) or beta cell malfunction and death provoked by glucotoxicity and insulin resistance in type 2 diabetes (T2D). Additionally, oxidative stress reduces functionality of beta cells in T2D by stimulating their de-/trans-differentiation through the loss of transcription factors critical for beta cell development, maturity and regeneration. This review summarizes up to date clarified redox-related mechanisms involved in regulating beta cell identity and death, underlining similarities and differences between T1D and T2D. The protective effects of natural antioxidants on the oxidative stress-induced beta cell failure were also discussed. Considering that oxidative stress affects epigenetic regulatory mechanisms involved in the regulation of pancreatic beta cell survival and insulin secretion, this review highlighted huge potential of epigenetic therapy. Special attention was paid on application of the state-of-the-art CRISPR/Cas9 technology, based on targeted epigenome editing with the purpose of changing the differentiation state of different cell types, making them insulin-producing with ability to attenuate diabetes. Clarification of the above-mentioned mechanisms could provide better insight into diabetes etiology and pathogenesis, which would allow development of novel, potentially more efficient therapeutic strategies for the prevention or reversion of beta cell loss.
Collapse
|
43
|
Serra-Navarro B, Fernandez-Ruiz R, García-Alamán A, Pradas-Juni M, Fernandez-Rebollo E, Esteban Y, Mir-Coll J, Mathieu J, Dalle S, Hahn M, Ahlgren U, Weinstein LS, Vidal J, Gomis R, Gasa R. Gsα-dependent signaling is required for postnatal establishment of a functional β-cell mass. Mol Metab 2021; 53:101264. [PMID: 34091063 PMCID: PMC8239471 DOI: 10.1016/j.molmet.2021.101264] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 05/17/2021] [Accepted: 05/30/2021] [Indexed: 12/01/2022] Open
Abstract
OBJECTIVE Early postnatal life is a critical period for the establishment of the functional β-cell mass that will sustain whole-body glucose homeostasis during the lifetime. β cells are formed from progenitors during embryonic development but undergo significant expansion in quantity and attain functional maturity after birth. The signals and pathways involved in these processes are not fully elucidated. Cyclic adenosine monophosphate (cAMP) is an intracellular signaling molecule that is known to regulate insulin secretion, gene expression, proliferation, and survival of adult β cells. The heterotrimeric G protein Gs stimulates the cAMP-dependent pathway by activating adenylyl cyclase. In this study, we sought to explore the role of Gs-dependent signaling in postnatal β-cell development. METHODS To study Gs-dependent signaling, we generated conditional knockout mice in which the α subunit of the Gs protein (Gsα) was ablated from β-cells using the Cre deleter line Ins1Cre. Mice were characterized in terms of glucose homeostasis, including in vivo glucose tolerance, glucose-induced insulin secretion, and insulin sensitivity. β-cell mass was studied using histomorphometric analysis and optical projection tomography. β-cell proliferation was studied by ki67 and phospho-histone H3 immunostatining, and apoptosis was assessed by TUNEL assay. Gene expression was determined in isolated islets and sorted β cells by qPCR. Intracellular cAMP was studied in isolated islets using HTRF-based technology. The activation status of the cAMP and insulin-signaling pathways was determined by immunoblot analysis of the relevant components of these pathways in isolated islets. In vitro proliferation of dissociated islet cells was assessed by BrdU incorporation. RESULTS Elimination of Gsα in β cells led to reduced β-cell mass, deficient insulin secretion, and severe glucose intolerance. These defects were evident by weaning and were associated with decreased proliferation and inadequate expression of key β-cell identity and maturation genes in postnatal β-cells. Additionally, loss of Gsα caused a broad multilevel disruption of the insulin transduction pathway that resulted in the specific abrogation of the islet proliferative response to insulin. CONCLUSION We conclude that Gsα is required for β-cell growth and maturation in the early postnatal stage and propose that this is partly mediated via its crosstalk with insulin signaling. Our findings disclose a tight connection between these two pathways in postnatal β cells, which may have implications for using cAMP-raising agents to promote β-cell regeneration and maturation in diabetes.
Collapse
Affiliation(s)
- Berta Serra-Navarro
- Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149-153, 08036, Barcelona, Spain; University of Barcelona, Barcelona, Spain
| | - Rebeca Fernandez-Ruiz
- Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149-153, 08036, Barcelona, Spain; University of Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Spain
| | - Ainhoa García-Alamán
- Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149-153, 08036, Barcelona, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Spain
| | - Marta Pradas-Juni
- Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149-153, 08036, Barcelona, Spain; University of Barcelona, Barcelona, Spain
| | - Eduardo Fernandez-Rebollo
- Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149-153, 08036, Barcelona, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Spain
| | - Yaiza Esteban
- Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149-153, 08036, Barcelona, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Spain
| | - Joan Mir-Coll
- Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149-153, 08036, Barcelona, Spain; University of Barcelona, Barcelona, Spain
| | - Julia Mathieu
- CHU Montpellier, Laboratory of Cell Therapy for Diabetes (LTCD), Hospital St-Eloi, Montpellier, France
| | - Stephane Dalle
- CHU Montpellier, Laboratory of Cell Therapy for Diabetes (LTCD), Hospital St-Eloi, Montpellier, France
| | - Max Hahn
- Umeå Centre for Molecular Medicine (UCMM), Umeå, Sweden
| | - Ulf Ahlgren
- Umeå Centre for Molecular Medicine (UCMM), Umeå, Sweden
| | - Lee S Weinstein
- Metabolic Diseases Branch, National Institute of Diabetes, Digestive, and Kidney Diseases, NIH, Bethesda, MD, USA
| | - Josep Vidal
- Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149-153, 08036, Barcelona, Spain; University of Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Spain; Department of Endocrinology and Nutrition, Hospital Clinic of Barcelona, Barcelona, Spain
| | - Ramon Gomis
- Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149-153, 08036, Barcelona, Spain; University of Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Spain; Universitat Oberta de Catalunya (UOC), Barcelona, Spain
| | - Rosa Gasa
- Diabetes and Obesity Research Laboratory, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149-153, 08036, Barcelona, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Spain.
| |
Collapse
|
44
|
Pydi SP, Barella LF, Zhu L, Meister J, Rossi M, Wess J. β-Arrestins as Important Regulators of Glucose and Energy Homeostasis. Annu Rev Physiol 2021; 84:17-40. [PMID: 34705480 DOI: 10.1146/annurev-physiol-060721-092948] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
β-Arrestin-1 and -2 (also known as arrestin-2 and -3, respectively) are ubiquitously expressed cytoplasmic proteins that dampen signaling through G protein-coupled receptors. However, β-arrestins can also act as signaling molecules in their own right. To investigate the potential metabolic roles of the two β-arrestins in modulating glucose and energy homeostasis, recent studies analyzed mutant mice that lacked or overexpressed β-arrestin-1 and/or -2 in distinct, metabolically important cell types. Metabolic analysis of these mutant mice clearly demonstrated that both β-arrestins play key roles in regulating the function of most of these cell types, resulting in striking changes in whole-body glucose and/or energy homeostasis. These studies also revealed that β-arrestin-1 and -2, though structurally closely related, clearly differ in their metabolic roles under physiological and pathophysiological conditions. These new findings should guide the development of novel drugs for the treatment of various metabolic disorders, including type 2 diabetes and obesity. Expected final online publication date for the Annual Review of Physiology, Volume 84 is February 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Sai P Pydi
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, US Department of Health and Human Services, Bethesda, Maryland, USA; .,Current affiliation: Department of Biological Sciences and Bioengineering, The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology, Kanpur, India
| | - Luiz F Barella
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, US Department of Health and Human Services, Bethesda, Maryland, USA;
| | - Lu Zhu
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, US Department of Health and Human Services, Bethesda, Maryland, USA;
| | - Jaroslawna Meister
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, US Department of Health and Human Services, Bethesda, Maryland, USA;
| | - Mario Rossi
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, US Department of Health and Human Services, Bethesda, Maryland, USA;
| | - Jürgen Wess
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, US Department of Health and Human Services, Bethesda, Maryland, USA;
| |
Collapse
|
45
|
Tien YW, Chien HJ, Chiang TC, Chung MH, Lee CY, Peng SJ, Chen CC, Chou YH, Hsiao FT, Jeng YM, Tang SC. Local islet remodelling associated with duct lesion-islet complex in adult human pancreas. Diabetologia 2021; 64:2266-2278. [PMID: 34272581 DOI: 10.1007/s00125-021-05504-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 03/19/2021] [Indexed: 12/13/2022]
Abstract
AIMS/HYPOTHESIS Islets are thought to be stably present in the adult human pancreas to maintain glucose homeostasis. However, identification of the pancreatic intraepithelial neoplasia (PanIN)-islet complex in mice and the presence of PanIN lesions in adult humans suggest that similar remodelling of islet structure and environment may occur in the human pancreas. To identify islet remodelling in a clinically related setting, we examine human donor pancreases with 3D histology to detect and characterise the human PanIN-islet complex. METHODS Cadaveric donor pancreases (26-65 years old, n = 10) were fixed and sectioned (350 μm) for tissue labelling, clearing and microscopy to detect local islet remodelling for 3D analysis of the microenvironment. The remodelled microenvironment was subsequently examined via microtome-based histology for clinical assessment. RESULTS In nine pancreases, we identified the unique peri-lobular islet aggregation associated with the PanIN lesion (16 lesion-islet complexes detected; size: 3.18 ± 1.34 mm). Important features of the lesion-islet microenvironment include: (1) formation of intra-islet ducts, (2) acinar atrophy, (3) adipocyte association, (4) inflammation (CD45+), (5) stromal accumulation (α-SMA+), (6) increase in Ki-67 proliferation index but absence of Ki-67+ alpha/beta cells and (7) in-depth and continuous duct-islet cell contacts, forming a cluster. The duct-islet cell cluster and intra-islet ducts suggest likely islet cell neogenesis but not replication. CONCLUSIONS/INTERPRETATION We identify local islet remodelling associated with PanIN-islet complex in the adult human pancreas. The tissue remodelling and the evidence of inflammation and stromal accumulation suggest that the PanIN-islet complex is derived from tissue repair after a local injury.
Collapse
Affiliation(s)
- Yu-Wen Tien
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Hung-Jen Chien
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Tsai-Chen Chiang
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Mei-Hsin Chung
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
- Department of Pathology, National Taiwan University Hospital - Hsinchu Branch, Hsinchu, Taiwan
| | - Chih-Yuan Lee
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Shih-Jung Peng
- Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Chien-Chia Chen
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Ya-Hsien Chou
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
- Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Fu-Ting Hsiao
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Yung-Ming Jeng
- Department of Pathology, National Taiwan University Hospital, Taipei, Taiwan
| | - Shiue-Cheng Tang
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan.
- Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
46
|
Abstract
This review focuses on the human pancreatic islet-including its structure, cell composition, development, function, and dysfunction. After providing a historical timeline of key discoveries about human islets over the past century, we describe new research approaches and technologies that are being used to study human islets and how these are providing insight into human islet physiology and pathophysiology. We also describe changes or adaptations in human islets in response to physiologic challenges such as pregnancy, aging, and insulin resistance and discuss islet changes in human diabetes of many forms. We outline current and future interventions being developed to protect, restore, or replace human islets. The review also highlights unresolved questions about human islets and proposes areas where additional research on human islets is needed.
Collapse
Affiliation(s)
- John T Walker
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Diane C Saunders
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Marcela Brissova
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Alvin C Powers
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- VA Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| |
Collapse
|
47
|
Kaneko S. Novel approaches to pharmacological management of type 2 diabetes in Japan. Expert Opin Pharmacother 2021; 22:2235-2249. [PMID: 34461791 DOI: 10.1080/14656566.2021.1974401] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
INTRODUCTION Newly developed anti-diabetic medications have had multiple activities, beyond a blood glucose-lowering effect. Current drugs for treating type 2 diabetes mellitus (T2DM) are based on the use of gastrointestinal hormones. Representative incretin preparations, such as those with glucagon-like peptide (GLP)-1 or gastric inhibitory polypeptide (GIP) activity, aim to provide new means of controlling blood glucose levels, body weight, and lipid metabolism. AREA COVERED In this manuscript, the pathophysiology of T2DM and the activities and characteristics of novel diabetic drugs are reviewed in the context of the Japanese population. This review also highlights the need for novel medicines to overcome the accompanying challenges. Finally, the author provides the reader with their expert perspectives. EXPERT OPINION The incidence of T2DM has been increasing in the aging of Japanese society. In older people, medical development should focus on safety, easier self-administration, and the relief of caregiver burden in terms of continuous administration. In the young, the focus should be on effectiveness, with a particular emphasis on the protection of organs, increasing the ease of adherence, and safety. Novel medicines will need to push the envelope in these areas.
Collapse
Affiliation(s)
- Shizuka Kaneko
- Department of Diabetes/Endocrinology/Metabolism, Takatsuki Red Cross Hospital, Takatsuki, Osaka, Japan
| |
Collapse
|
48
|
Wang W, Zhang C. Targeting β-cell dedifferentiation and transdifferentiation: opportunities and challenges. Endocr Connect 2021; 10:R213-R228. [PMID: 34289444 PMCID: PMC8428079 DOI: 10.1530/ec-21-0260] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 07/21/2021] [Indexed: 12/02/2022]
Abstract
The most distinctive pathological characteristics of diabetes mellitus induced by various stressors or immune-mediated injuries are reductions of pancreatic islet β-cell populations and activity. Existing treatment strategies cannot slow disease progression; consequently, research to genetically engineer β-cell mimetics through bi-directional plasticity is ongoing. The current consensus implicates β-cell dedifferentiation as the primary etiology of reduced β-cell mass and activity. This review aims to summarize the etiology and proposed mechanisms of β-cell dedifferentiation and to explore the possibility that there might be a time interval from the onset of β-cell dysfunction caused by dedifferentiation to the development of diabetes, which may offer a therapeutic window to reduce β-cell injury and to stabilize functionality. In addition, to investigate β-cell plasticity, we review strategies for β-cell regeneration utilizing genetic programming, small molecules, cytokines, and bioengineering to transdifferentiate other cell types into β-cells; the development of biomimetic acellular constructs to generate fully functional β-cell-mimetics. However, the maturation of regenerated β-cells is currently limited. Further studies are needed to develop simple and efficient reprogramming methods for assembling perfectly functional β-cells. Future investigations are necessary to transform diabetes into a potentially curable disease.
Collapse
Affiliation(s)
- Wenrui Wang
- Department of Endocrinology, The Second Hospital of Jilin University, Changchun, People’s Republic of China
| | - Chuan Zhang
- Department of Endocrinology, The Second Hospital of Jilin University, Changchun, People’s Republic of China
- Correspondence should be addressed to C Zhang:
| |
Collapse
|
49
|
Lupse B, Annamalai K, Ibrahim H, Kaur S, Geravandi S, Sarma B, Pal A, Awal S, Joshi A, Rafizadeh S, Madduri MK, Khazaei M, Liu H, Yuan T, He W, Gorrepati KDD, Azizi Z, Qi Q, Ye K, Oberholzer J, Maedler K, Ardestani A. Inhibition of PHLPP1/2 phosphatases rescues pancreatic β-cells in diabetes. Cell Rep 2021; 36:109490. [PMID: 34348155 PMCID: PMC8421018 DOI: 10.1016/j.celrep.2021.109490] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 06/06/2021] [Accepted: 07/14/2021] [Indexed: 12/16/2022] Open
Abstract
Pancreatic β-cell failure is the key pathogenic element of the complex metabolic deterioration in type 2 diabetes (T2D); its underlying pathomechanism is still elusive. Here, we identify pleckstrin homology domain leucine-rich repeat protein phosphatases 1 and 2 (PHLPP1/2) as phosphatases whose upregulation leads to β-cell failure in diabetes. PHLPP levels are highly elevated in metabolically stressed human and rodent diabetic β-cells. Sustained hyper-activation of mechanistic target of rapamycin complex 1 (mTORC1) is the primary mechanism of the PHLPP upregulation linking chronic metabolic stress to ultimate β-cell death. PHLPPs directly dephosphorylate and regulate activities of β-cell survival-dependent kinases AKT and MST1, constituting a regulatory triangle loop to control β-cell apoptosis. Genetic inhibition of PHLPPs markedly improves β-cell survival and function in experimental models of diabetes in vitro, in vivo, and in primary human T2D islets. Our study presents PHLPPs as targets for functional regenerative therapy of pancreatic β cells in diabetes.
Collapse
Affiliation(s)
- Blaz Lupse
- Centre for Biomolecular Interactions Bremen, University of Bremen, 28359 Bremen, Germany
| | - Karthika Annamalai
- Centre for Biomolecular Interactions Bremen, University of Bremen, 28359 Bremen, Germany
| | - Hazem Ibrahim
- Centre for Biomolecular Interactions Bremen, University of Bremen, 28359 Bremen, Germany
| | - Supreet Kaur
- Centre for Biomolecular Interactions Bremen, University of Bremen, 28359 Bremen, Germany
| | - Shirin Geravandi
- Centre for Biomolecular Interactions Bremen, University of Bremen, 28359 Bremen, Germany
| | - Bhavishya Sarma
- Centre for Biomolecular Interactions Bremen, University of Bremen, 28359 Bremen, Germany
| | - Anasua Pal
- Centre for Biomolecular Interactions Bremen, University of Bremen, 28359 Bremen, Germany
| | - Sushil Awal
- Centre for Biomolecular Interactions Bremen, University of Bremen, 28359 Bremen, Germany
| | - Arundhati Joshi
- Centre for Biomolecular Interactions Bremen, University of Bremen, 28359 Bremen, Germany
| | - Sahar Rafizadeh
- Centre for Biomolecular Interactions Bremen, University of Bremen, 28359 Bremen, Germany
| | - Murali Krishna Madduri
- Centre for Biomolecular Interactions Bremen, University of Bremen, 28359 Bremen, Germany
| | - Mona Khazaei
- Centre for Biomolecular Interactions Bremen, University of Bremen, 28359 Bremen, Germany
| | - Huan Liu
- Centre for Biomolecular Interactions Bremen, University of Bremen, 28359 Bremen, Germany
| | - Ting Yuan
- Centre for Biomolecular Interactions Bremen, University of Bremen, 28359 Bremen, Germany
| | - Wei He
- Centre for Biomolecular Interactions Bremen, University of Bremen, 28359 Bremen, Germany
| | | | - Zahra Azizi
- Centre for Biomolecular Interactions Bremen, University of Bremen, 28359 Bremen, Germany; Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran 1449614535, Iran
| | - Qi Qi
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Keqiang Ye
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jose Oberholzer
- Charles O. Strickler Transplant Center, University of Virginia Medical Center, Charlottesville, VA 22903, USA
| | - Kathrin Maedler
- Centre for Biomolecular Interactions Bremen, University of Bremen, 28359 Bremen, Germany.
| | - Amin Ardestani
- Centre for Biomolecular Interactions Bremen, University of Bremen, 28359 Bremen, Germany; Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran 1449614535, Iran.
| |
Collapse
|
50
|
Sudirman S, Su CY, Tsou D, Lee MC, Kong ZL. Hippocampus kuda protein hydrolysate improves male reproductive dysfunction in diabetic rats. Biomed Pharmacother 2021; 140:111760. [PMID: 34052566 DOI: 10.1016/j.biopha.2021.111760] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 05/05/2021] [Accepted: 05/20/2021] [Indexed: 12/19/2022] Open
Abstract
The global prevalence of diabetes mellitus is rapidly increasing. This disease is associated with many complications including male reproductive dysfunctions and infertility. Seahorse ( Hippocampus kuda) is a marine teleost fish well known for its beneficial effects on the reproductive system in traditional Chinese medicine books. Recently, several studies have been shown that the enzymatic hydrolysate of seahorse has multiple pharmacological activities. This study aimed to investigate the seahorse peptide hydrolysate (SH) ameliorative effects on the diabetic-induced male reproductive dysfunction in rat models. The in vivo studies were carried out with three different doses of SH (4, 8, and 20 mg/kg) and the diabetes condition was induced by administrating with streptozotocin (35 mg/kg) and fed a 40% high-fat diet. Seahorse hydrolysate (20 mg/kg) inhibited lipid peroxidation, increased antioxidant enzyme activity, and restored seminiferous tubules morphology in testis. Moreover, it improved reproductive dysfunction by increasing the level of testosterone, follicle-stimulating hormone, luteinizing hormone, sperm count, and motility. According to these results, we suggested that SH exhibited amelioration effects on the reproductive dysfunction.
Collapse
Affiliation(s)
- Sabri Sudirman
- Fisheries Product Technology, Faculty of Agriculture, Universitas Sriwijaya, Indralaya 30862, Indonesia.
| | - Chieh-Yu Su
- Department of Food Science; National Taiwan Ocean University, Keelung City 20224, Taiwan.
| | - David Tsou
- Department of Food Science; National Taiwan Ocean University, Keelung City 20224, Taiwan.
| | - Meng-Chou Lee
- Department of Aquaculture, National Taiwan Ocean University, Keelung City 20224, Taiwan.
| | - Zwe-Ling Kong
- Department of Food Science; National Taiwan Ocean University, Keelung City 20224, Taiwan.
| |
Collapse
|