1
|
Tonyan ZN, Barbitoff YA, Nasykhova YA, Danilova MM, Kozyulina PY, Mikhailova AA, Bulgakova OL, Vlasova ME, Golovkin NV, Glotov AS. Plasma microRNA Profiling in Type 2 Diabetes Mellitus: A Pilot Study. Int J Mol Sci 2023; 24:17406. [PMID: 38139235 PMCID: PMC10744218 DOI: 10.3390/ijms242417406] [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: 10/23/2023] [Revised: 12/04/2023] [Accepted: 12/10/2023] [Indexed: 12/24/2023] Open
Abstract
Type 2 diabetes mellitus (T2D) is a chronic metabolic disease characterized by insulin resistance and β-cell dysfunction and leading to many micro- and macrovascular complications. In this study we analyzed the circulating miRNA expression profiles in plasma samples from 44 patients with T2D and 22 healthy individuals using next generation sequencing and detected 229 differentially expressed miRNAs. An increased level of miR-5588-5p, miR-125b-2-3p, miR-1284, and a reduced level of miR-496 in T2D patients was verified. We also compared the expression landscapes in the same group of patients depending on body mass index and identified differential expression of miR-144-3p and miR-99a-5p in obese individuals. Identification and functional analysis of putative target genes was performed for miR-5588-5p, miR-125b-2-3p, miR-1284, and miR-496, showing chromatin modifying enzymes and apoptotic genes being among the significantly enriched pathways.
Collapse
Affiliation(s)
- Ziravard N. Tonyan
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, 199034 St. Petersburg, Russia; (Z.N.T.); (Y.A.B.); (Y.A.N.); (M.M.D.); (P.Y.K.); (A.A.M.); (O.L.B.)
| | - Yury A. Barbitoff
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, 199034 St. Petersburg, Russia; (Z.N.T.); (Y.A.B.); (Y.A.N.); (M.M.D.); (P.Y.K.); (A.A.M.); (O.L.B.)
| | - Yulia A. Nasykhova
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, 199034 St. Petersburg, Russia; (Z.N.T.); (Y.A.B.); (Y.A.N.); (M.M.D.); (P.Y.K.); (A.A.M.); (O.L.B.)
| | - Maria M. Danilova
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, 199034 St. Petersburg, Russia; (Z.N.T.); (Y.A.B.); (Y.A.N.); (M.M.D.); (P.Y.K.); (A.A.M.); (O.L.B.)
| | - Polina Y. Kozyulina
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, 199034 St. Petersburg, Russia; (Z.N.T.); (Y.A.B.); (Y.A.N.); (M.M.D.); (P.Y.K.); (A.A.M.); (O.L.B.)
| | - Anastasiia A. Mikhailova
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, 199034 St. Petersburg, Russia; (Z.N.T.); (Y.A.B.); (Y.A.N.); (M.M.D.); (P.Y.K.); (A.A.M.); (O.L.B.)
| | - Olga L. Bulgakova
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, 199034 St. Petersburg, Russia; (Z.N.T.); (Y.A.B.); (Y.A.N.); (M.M.D.); (P.Y.K.); (A.A.M.); (O.L.B.)
| | - Margarita E. Vlasova
- St. Martyr George City Hospital, 194354 St. Petersburg, Russia; (M.E.V.); (N.V.G.)
| | - Nikita V. Golovkin
- St. Martyr George City Hospital, 194354 St. Petersburg, Russia; (M.E.V.); (N.V.G.)
| | - Andrey S. Glotov
- Department of Genomic Medicine, D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, 199034 St. Petersburg, Russia; (Z.N.T.); (Y.A.B.); (Y.A.N.); (M.M.D.); (P.Y.K.); (A.A.M.); (O.L.B.)
| |
Collapse
|
2
|
Colon-Mesa I, Sainz N, Corrales P, Collantes M, Kaldis P, Martinez JA, Medina-Gómez G, Moreno-Aliaga MJ, Escoté X. p27Kip1 Deficiency Impairs Brown Adipose Tissue Function Favouring Fat Accumulation in Mice. Int J Mol Sci 2023; 24:ijms24032664. [PMID: 36768986 PMCID: PMC9916555 DOI: 10.3390/ijms24032664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 01/13/2023] [Accepted: 01/22/2023] [Indexed: 02/01/2023] Open
Abstract
The aim of this work was to investigate the effect of the whole-body deletion of p27 on the activity of brown adipose tissue and the susceptibility to develop obesity and glucose homeostasis disturbances in mice, especially when subjected to a high fat diet. p27 knockout (p27-/-) and wild type (WT) mice were fed a normal chow diet or a high fat diet (HFD) for 10-weeks. Body weight and composition were assessed. Insulin and glucose tolerance tests and indirect calorimetry assays were performed. Histological analysis of interscapular BAT (iBAT) was carried out, and expression of key genes/proteins involved in BAT function were characterized by qPCR and Western blot. iBAT activity was estimated by 18F-fluorodeoxyglucose (18FDG) uptake with microPET. p27-/- mice were more prone to develop obesity and insulin resistance, exhibiting increased size of all fat depots. p27-/- mice displayed a higher respiratory exchange ratio. iBAT presented larger adipocytes in p27-/- HFD mice, accompanied by downregulation of both Glut1 and uncoupling protein 1 (UCP1) in parallel with defective insulin signalling. Moreover, p27-/- HFD mice exhibited impaired response to cold exposure, characterized by a reduced iBAT 18FDG uptake and difficulty to maintain body temperature when exposed to cold compared to WT HFD mice, suggesting reduced thermogenic capacity. These data suggest that p27 could play a role in BAT activation and in the susceptibility to develop obesity and insulin resistance.
Collapse
Affiliation(s)
- Ignacio Colon-Mesa
- Department of Nutrition, Food Science and Physiology and Center for Nutrition Research, School of Pharmacy and Nutrition, University of Navarra, Irunlarrea 1, 31008 Pamplona, Spain
| | - Neira Sainz
- Department of Nutrition, Food Science and Physiology and Center for Nutrition Research, School of Pharmacy and Nutrition, University of Navarra, Irunlarrea 1, 31008 Pamplona, Spain
| | - Patricia Corrales
- Department of Basic Sciences of Health, Area of Biochemistry and Molecular Biology, Universidad Rey Juan Carlos, Alcorcon, 28933 Madrid, Spain
| | - María Collantes
- Nuclear Medicine Department, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Philipp Kaldis
- Department of Clinical Sciences, Lund University, Clinical Research Centre (CRC), P.O. Box 50332, SE-202 13 Malmö, Sweden
- Lund University Diabetes Centre (LUDC), Lund University, SE-202 13 Malmö, Sweden
| | - José Alfredo Martinez
- Department of Nutrition, Food Science and Physiology and Center for Nutrition Research, School of Pharmacy and Nutrition, University of Navarra, Irunlarrea 1, 31008 Pamplona, Spain
| | - Gema Medina-Gómez
- Department of Basic Sciences of Health, Area of Biochemistry and Molecular Biology, Universidad Rey Juan Carlos, Alcorcon, 28933 Madrid, Spain
- LAFEMEX Laboratory, Área de Bioquímica y Biología Molecular, Departamento de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, 28922 Alcorcón, Spain
| | - María Jesús Moreno-Aliaga
- Department of Nutrition, Food Science and Physiology and Center for Nutrition Research, School of Pharmacy and Nutrition, University of Navarra, Irunlarrea 1, 31008 Pamplona, Spain
- IdISNA—Navarra Institute for Health Research, 31008 Pamplona, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-948-425-600
| | - Xavier Escoté
- Department of Nutrition, Food Science and Physiology and Center for Nutrition Research, School of Pharmacy and Nutrition, University of Navarra, Irunlarrea 1, 31008 Pamplona, Spain
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, 43204 Reus, Spain
| |
Collapse
|
3
|
Lee JH, Lee J. Endoplasmic Reticulum (ER) Stress and Its Role in Pancreatic β-Cell Dysfunction and Senescence in Type 2 Diabetes. Int J Mol Sci 2022; 23:ijms23094843. [PMID: 35563231 PMCID: PMC9104816 DOI: 10.3390/ijms23094843] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 04/22/2022] [Accepted: 04/26/2022] [Indexed: 02/07/2023] Open
Abstract
An increased life span and accompanying nutritional affluency have led to a rapid increase in diseases associated with aging, such as obesity and type 2 diabetes, imposing a tremendous economic and health burden on society. Pancreatic β-cells are crucial for controlling glucose homeostasis by properly producing and secreting the glucose-lowering hormone insulin, and the dysfunction of β-cells determines the outcomes for both type 1 and type 2 diabetes. As the native structure of insulin is formed within the endoplasmic reticulum (ER), ER homeostasis should be appropriately maintained to allow for the proper metabolic homeostasis and functioning of β-cells. Recent studies have found that cellular senescence is critically linked with cellular stresses, including ER stress, oxidative stress, and mitochondrial stress. These studies implied that β-cell senescence is caused by ER stress and other cellular stresses and contributes to β-cells’ dysfunction and the impairment of glucose homeostasis. This review documents and discusses the current understanding of cellular senescence, β-cell function, ER stress, its associated signaling mechanism (unfolded protein response), and the effect of ER stress on β-cell senescence and dysfunction.
Collapse
Affiliation(s)
- Ji-Hye Lee
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea;
- New Biology Research Center, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| | - Jaemin Lee
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea;
- New Biology Research Center, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
- Well Aging Research Center, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
- Correspondence:
| |
Collapse
|
4
|
Abou Azar F, Lim GE. Metabolic Contributions of Wnt Signaling: More Than Controlling Flight. Front Cell Dev Biol 2021; 9:709823. [PMID: 34568323 PMCID: PMC8458764 DOI: 10.3389/fcell.2021.709823] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 08/16/2021] [Indexed: 12/12/2022] Open
Abstract
The canonical Wnt signaling pathway is ubiquitous throughout the body and influences a diverse array of physiological processes. Following the initial discovery of the Wnt signaling pathway during wing development in Drosophila melanogaster, it is now widely appreciated that active Wnt signaling in mammals is necessary for the development and growth of various tissues involved in whole-body metabolism, such as brain, liver, pancreas, muscle, and adipose. Moreover, elegant gain- and loss-of-function studies have dissected the tissue-specific roles of various downstream effector molecules in the regulation of energy homeostasis. This review attempts to highlight and summarize the contributions of the Wnt signaling pathway and its downstream effectors on whole-body metabolism and their influence on the development of metabolic diseases, such as diabetes and obesity. A better understanding of the Wnt signaling pathway in these tissues may aid in guiding the development of future therapeutics to treat metabolic diseases.
Collapse
Affiliation(s)
- Frederic Abou Azar
- Department of Medicine, Université de Montréal, Montreal, QC, Canada.,Cardiometabolic Axis, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Gareth E Lim
- Department of Medicine, Université de Montréal, Montreal, QC, Canada.,Cardiometabolic Axis, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| |
Collapse
|
5
|
Aitken TJ, Crabtree JE, Jensen DM, Hess KH, Leininger BR, Tessem JS. Decreased proliferation of aged rat beta cells corresponds with enhanced expression of the cell cycle inhibitor p27 KIP1. Biol Cell 2021; 113:507-521. [PMID: 34523154 DOI: 10.1111/boc.202100035] [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] [Received: 05/18/2021] [Revised: 08/24/2021] [Accepted: 08/26/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Over 400 million people are diabetic. Type 1 and type 2 diabetes are characterized by decreased functional β-cell mass and, consequently, decreased glucose-stimulated insulin secretion. A potential intervention is transplantation of β-cell containing islets from cadaveric donors. A major impediment to greater application of this treatment is the scarcity of transplant-ready β-cells. Therefore, inducing β-cell proliferation ex vivo could be used to expand functional β-cell mass prior to transplantation. Various molecular pathways are sufficient to induce proliferation of young β-cells; however, aged β-cells are refractory to these proliferative signals. Given that the majority of cadaveric donors fit an aged demographic, defining the mechanisms that impede aged β-cell proliferation is imperative. RESULTS We demonstrate that aged rat (5-month-old) β-cells are refractory to mitogenic stimuli that otherwise induce young rat (5-week-old) β-cell proliferation. We hypothesized that this change in proliferative capacity could be due to differences in cyclin-dependent kinase inhibitor expression. We measured levels of p16INK4a , p15INK4b , p18INK4c , p19INK4d , p21CIP1 , p27KIP1 and p57KIP2 by immunofluorescence analysis. Our data demonstrates an age-dependent increase of p27KIP1 in rat β-cells by immunofluorescence and was validated by increased p27KIP1 protein levels by western blot analysis. Interestingly, HDAC1, which modulates the p27KIP1 promoter acetylation state, is downregulated in aged rat islets. These data demonstrate increased p27KIP1 protein levels at 5 months of age, which may be due to decreased HDAC1 mediated repression of p27KIP1 expression. SIGNIFICANCE As the majority of transplant-ready β-cells come from aged donors, it is imperative that we understand why aged β-cells are refractory to mitogenic stimuli. Our findings demonstrate that increased p27KIP1 expression occurs early in β-cell aging, which corresponds with impaired β-cell proliferation. Furthermore, the correlation between HDAC1 and p27 levels suggests that pathways that activate HDAC1 in aged β-cells could be leveraged to decrease p27KIP1 levels and enhance β-cell proliferation.
Collapse
Affiliation(s)
- Talon J Aitken
- Department of Nutrition, Dietetics, and Food Science, Brigham Young University, Provo, Utah, USA.,Medical Education Program, Des Moines University, Des Moines, IA, 50312, USA
| | - Jacqueline E Crabtree
- Department of Nutrition, Dietetics, and Food Science, Brigham Young University, Provo, Utah, USA
| | - Daelin M Jensen
- Department of Nutrition, Dietetics, and Food Science, Brigham Young University, Provo, Utah, USA.,Biomedical Sciences, Ohio State University, Columbus, OH, 43210, USA
| | - Kavan H Hess
- Department of Nutrition, Dietetics, and Food Science, Brigham Young University, Provo, Utah, USA.,Medical Education Program, Idaho College of Osteopathic Medicine, Meridian, ID, 83642, USA
| | - Brennan R Leininger
- Department of Nutrition, Dietetics, and Food Science, Brigham Young University, Provo, Utah, USA.,Dental Education Program, UCLA School of Dentistry, Los Angeles, CA, 90024, USA
| | - Jeffery S Tessem
- Department of Nutrition, Dietetics, and Food Science, Brigham Young University, Provo, Utah, USA
| |
Collapse
|
6
|
Altman MK, Schaub CM, Dadi PK, Dickerson MT, Zaborska KE, Nakhe AY, Graff SM, Galletta TJ, Amarnath G, Thorson AS, Gu G, Jacobson DA. TRPM7 is a crucial regulator of pancreatic endocrine development and high-fat-diet-induced β-cell proliferation. Development 2021; 148:dev194928. [PMID: 34345920 PMCID: PMC8406533 DOI: 10.1242/dev.194928] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 07/20/2021] [Indexed: 12/31/2022]
Abstract
The melastatin subfamily of the transient receptor potential channels (TRPM) are regulators of pancreatic β-cell function. TRPM7 is the most abundant islet TRPM channel; however, the role of TRPM7 in β-cell function has not been determined. Here, we used various spatiotemporal transgenic mouse models to investigate how TRPM7 knockout influences pancreatic endocrine development, proliferation and function. Ablation of TRPM7 within pancreatic progenitors reduced pancreatic size, and α-cell and β-cell mass. This resulted in modestly impaired glucose tolerance. However, TRPM7 ablation following endocrine specification or in adult mice did not impact endocrine expansion or glucose tolerance. As TRPM7 regulates cell proliferation, we assessed how TRPM7 influences β-cell hyperplasia under insulin-resistant conditions. β-Cell proliferation induced by high-fat diet was significantly decreased in TRPM7-deficient β-cells. The endocrine roles of TRPM7 may be influenced by cation flux through the channel, and indeed we found that TRPM7 ablation altered β-cell Mg2+ and reduced the magnitude of elevation in β-cell Mg2+ during proliferation. Together, these findings revealed that TRPM7 controls pancreatic development and β-cell proliferation, which is likely due to regulation of Mg2+ homeostasis.
Collapse
Affiliation(s)
- Molly K. Altman
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 7425B MRB IV, 2213 Garland Ave., Nashville, TN 37232, USA
| | - Charles M. Schaub
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 7425B MRB IV, 2213 Garland Ave., Nashville, TN 37232, USA
| | - Prasanna K. Dadi
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 7425B MRB IV, 2213 Garland Ave., Nashville, TN 37232, USA
| | - Matthew T. Dickerson
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 7425B MRB IV, 2213 Garland Ave., Nashville, TN 37232, USA
| | - Karolina E. Zaborska
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 7425B MRB IV, 2213 Garland Ave., Nashville, TN 37232, USA
| | - Arya Y. Nakhe
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 7425B MRB IV, 2213 Garland Ave., Nashville, TN 37232, USA
| | - Sarah M. Graff
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 7425B MRB IV, 2213 Garland Ave., Nashville, TN 37232, USA
| | - Thomas J. Galletta
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 7425B MRB IV, 2213 Garland Ave., Nashville, TN 37232, USA
| | - Gautami Amarnath
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 7425B MRB IV, 2213 Garland Ave., Nashville, TN 37232, USA
- Molecular Neurophysiology, Institute of Applied Physiology, University of Ulm, 89081 Ulm, Germany
| | - Ariel S. Thorson
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 7425B MRB IV, 2213 Garland Ave., Nashville, TN 37232, USA
| | - Guoqiang Gu
- Vanderbilt Program in Developmental Biology, Vanderbilt Center for Stem Cell Biology, Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - David A. Jacobson
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 7425B MRB IV, 2213 Garland Ave., Nashville, TN 37232, USA
| |
Collapse
|
7
|
Ahmed SM, Elshenawy SE, Sedky S, Elmehrath AO, El-Badri N. Pancreatic β-Cell Senescence: Mechanisms and Association with Diabetes. EUROPEAN MEDICAL JOURNAL 2021. [DOI: 10.33590/emj/20-00128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Senescence occurs as a part of the cellular response to different stressors. With increasing age, continuous exposure to stressors leads to age-induced senescence. Pancreatic β-cell proliferation and glucose homeostasis also decrease with age, which results in a decrease in β cell mass and, eventually, the possible development of diabetes. This process is mediated through impaired cell cycle regulators, along with specific increases in cell cycle inhibitors, telomere shortening, and defective DNA repair mechanisms. Diabetes contributes to β-cell senescence through hyperglycaemia, dyslipidaemia, oxidative stress, and inflammation. β cells isolated from patients with Type 2 diabetes mellitus have been shown to have senescence markers, such as senescence-associated secretory phenotype genes and β-galactosidase. In this paper, the authors discuss the mechanisms of cellular senescence, how senescence is impacted by the diabetic microenvironment, and the possible mechanisms and factors contributing to β-cell senescence.
Collapse
Affiliation(s)
- Sara M Ahmed
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, Giza, Egypt
| | - Shimaa E Elshenawy
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, Giza, Egypt
| | - Sara Sedky
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, Giza, Egypt
| | - Ahmed O Elmehrath
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, Giza, Egypt; Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Nagwa El-Badri
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, Giza, Egypt
| |
Collapse
|
8
|
Hwang YJ, Jung GS, Jeon WB, Lee KM. Arg-Gly-Asp-modified elastin-like polypeptide regulates cell proliferation and cell cycle proteins via the phosphorylation of Erk and Akt in pancreatic β-cell. Heliyon 2020; 6:e04918. [PMID: 32995613 PMCID: PMC7501433 DOI: 10.1016/j.heliyon.2020.e04918] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 08/14/2020] [Accepted: 09/08/2020] [Indexed: 11/24/2022] Open
Abstract
Objective Enhancement of β-cell proliferation plays an important role in maintaining β-cell mass and function, and in improving pancreatic β-cell survival before transplantation. Extracellular matrix (ECM) components increase the adhesion and proliferation of β-cells, and the RGD-modified elastin-like polypeptide (RGD-ELP, REP) has been described as a bioactive matrix. In this study, we investigated whether REP could enhance β-cell adhesion and proliferation and elucidated the signaling pathways involved. Methods We investigated the effect of REP on cell adhesion, proliferation and insulin secretion via assays using Rin-m and rat islets. Crystal violet, CCK-8, and BrdU assay, FACS, western blot, real time q-PCR analyses and insulin ELISA were examined. To explain the associated mechanisms, phosphorylation of Akt and extracellular signal-regulated kinase (Erk) were measured. Results REP more increased the adhesion, proliferation and survival of Rin-m cells compared to elastin-like poly peptide (ELP) without RGD-motif. The enhancement of β-cell proliferation by REP was associated with increased cyclin D1, cyclin D2 and cdk6, and decreased p27 levels. When β-cells were cultured on REP, Erk and the phosphatidylinositol 3-kinase (PI3-kinase) downstream effector, Akt was stimulated. Treatment with the Erk pathway inhibitor and PI3-kinase inhibitor decreased REP-induced β-cell adhesion and proliferation, and regulated REP-induced cell cycle proteins. Additionally, REP increased the mRNA and protein levels of insulin and its transcription factor, PDX-1, and insulin secretion. Conclusions Our results demonstrate that the up-regulation of the PI3K/Akt and Erk signaling pathways and the regulation of cell cycle proteins by REP could serve as effective strategies for improving pancreatic β-cell adhesion and proliferation.
Collapse
Affiliation(s)
- Yeo Jin Hwang
- Division of Electronics & Information System, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
| | - Gwon-Soo Jung
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Republic of Korea
| | - Won Bae Jeon
- Division of Biotechnology, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
| | - Kyeong-Min Lee
- Division of Biotechnology, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
| |
Collapse
|
9
|
Xu W, Cui J, Zhou F, Bai M, Deng R, Wang W. Leonurine protects against dexamethasone-induced cytotoxicity in pancreatic β-cells via PI3K/Akt signaling pathway. Biochem Biophys Res Commun 2020; 529:652-658. [DOI: 10.1016/j.bbrc.2020.05.184] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 05/26/2020] [Indexed: 01/27/2023]
|
10
|
Yahaya T, Salisu T. Genes predisposing to type 1 diabetes mellitus and pathophysiology: a narrative review. MEDICAL JOURNAL OF INDONESIA 2020. [DOI: 10.13181/mji.rev.203732] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The possibility of targeting the causal genes along with the mechanisms of pathogenically complex diseases has led to numerous studies on the genetic etiology of some diseases. In particular, studies have added more genes to the list of type 1 diabetes mellitus (T1DM) suspect genes, necessitating an update for the interest of all stakeholders. Therefore this review articulates T1DM suspect genes and their pathophysiology. Notable electronic databases, including Medline, Scopus, PubMed, and Google-Scholar were searched for relevant information. The search identified over 73 genes suspected in the pathogenesis of T1DM, with human leukocyte antigen, insulin gene, and cytotoxic T lymphocyte-associated antigen 4 accounting for most of the cases. Mutations in these genes, along with environmental factors, may produce a defective immune response in the pancreas, resulting in β-cell autoimmunity, insulin deficiency, and hyperglycemia. The mechanisms leading to these cellular reactions are gene-specific and, if targeted in diabetic individuals, may lead to improved treatment. Medical practitioners are advised to formulate treatment procedures that target these genes in patients with T1DM.
Collapse
|
11
|
Neelankal John A, Jiang FX. An overview of type 2 diabetes and importance of vitamin D3-vitamin D receptor interaction in pancreatic β-cells. J Diabetes Complications 2018; 32:429-443. [PMID: 29422234 DOI: 10.1016/j.jdiacomp.2017.12.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Revised: 12/03/2017] [Accepted: 12/07/2017] [Indexed: 02/07/2023]
Abstract
One significant health issue that plagues contemporary society is that of Type 2 diabetes (T2D). This disease is characterised by higher-than-average blood glucose levels as a result of a combination of insulin resistance and insufficient insulin secretions from the β-cells of pancreatic islets of Langerhans. Previous developmental research into the pancreas has identified how early precursor genes of pancreatic β-cells, such as Cpal, Ngn3, NeuroD, Ptf1a, and cMyc, play an essential role in the differentiation of these cells. Furthermore, β-cell molecular characterization has also revealed the specific role of β-cell-markers, such as Glut2, MafA, Ins1, Ins2, and Pdx1 in insulin expression. The expression of these genes appears to be suppressed in the T2D β-cells, along with the reappearance of the early endocrine marker genes. Glucose transporters transport glucose into β-cells, thereby controlling insulin release during hyperglycaemia. This stimulates glycolysis through rises in intracellular calcium (a process enhanced by vitamin D) (Norman et al., 1980), activating 2 of 4 proteinases. The rise in calcium activates half of pancreatic β-cell proinsulinases, thus releasing free insulin from granules. The synthesis of ATP from glucose by glycolysis, Krebs cycle and oxidative phosphorylation plays a role in insulin release. Some studies have found that the β-cells contain high levels of the vitamin D receptor; however, the role that this plays in maintaining the maturity of the β-cells remains unknown. Further research is required to develop a more in-depth understanding of the role VDR plays in β-cell function and the processes by which the beta cell function is preserved.
Collapse
Affiliation(s)
- Abraham Neelankal John
- Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands, Western Australia, Australia; School of Medicine and Pharmacology, University of Western Australia, Carwley, Western Australia, Australia
| | - Fang-Xu Jiang
- Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands, Western Australia, Australia; School of Medicine and Pharmacology, University of Western Australia, Carwley, Western Australia, Australia.
| |
Collapse
|
12
|
Definition of a Skp2-c-Myc Pathway to Expand Human Beta-cells. Sci Rep 2016; 6:28461. [PMID: 27380896 PMCID: PMC4933882 DOI: 10.1038/srep28461] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 05/25/2016] [Indexed: 12/20/2022] Open
Abstract
Type 2 diabetes (T2D) is characterized by insulin resistance and reduced functional β-cell mass. Developmental differences, failure of adaptive expansion and loss of β-cells via β-cell death or de-differentiation have emerged as the possible causes of this reduced β-cell mass. We hypothesized that the proliferative response to mitogens of human β-cells from T2D donors is reduced, and that this might contribute to the development and progression of T2D. Here, we demonstrate that the proliferative response of human β-cells from T2D donors in response to cdk6 and cyclin D3 is indeed dramatically impaired. We show that this is accompanied by increased nuclear abundance of the cell cycle inhibitor, p27kip1. Increasing nuclear abundance of p27kip1 by adenoviral delivery decreases the proliferative response of β-cells from non-diabetic donors, mimicking T2D β-cells. However, while both p27kip1 gene silencing and downregulation by Skp2 overexpression increased similarly the proliferative response of human β-cells, only Skp2 was capable of inducing a significant human β-cell expansion. Skp2 was also able to double the proliferative response of T2D β-cells. These studies define c-Myc as a central Skp2 target for the induction of cell cycle entry, expansion and regeneration of human T2D β-cells.
Collapse
|
13
|
Wiedemann T, Bielohuby M, Müller TD, Bidlingmaier M, Pellegata NS. Obesity in MENX Rats Is Accompanied by High Circulating Levels of Ghrelin and Improved Insulin Sensitivity. Diabetes 2016; 65:406-20. [PMID: 26512025 DOI: 10.2337/db15-0374] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 10/23/2015] [Indexed: 11/13/2022]
Abstract
Ghrelin, the natural ligand of the growth hormone secretagogue receptor type 1a (GHS-R1a), is mainly secreted from the stomach and regulates food intake and energy homeostasis. p27 regulates cell cycle progression in many cell types. Here, we report that rats affected by the multiple endocrine neoplasia syndrome MENX, caused by a p27 mutation, develop pancreatic islet hyperplasia containing elevated numbers of ghrelin-producing ε-cells. The metabolic phenotype of MENX-affected rats featured high endogenous acylated and unacylated plasma ghrelin levels. Supporting increased ghrelin action, MENX rats show increased food intake, enhanced body fat mass, and elevated plasma levels of triglycerides and cholesterol. Ghrelin effect on food intake was confirmed by treating MENX rats with a GHS-R1a antagonist. At 7.5 months, MENX-affected rats show decreased mRNA levels of hypothalamic GHS-R1a, neuropeptide Y (NPY), and agouti-related protein (AgRP), suggesting that prolonged hyperghrelinemia may lead to decreased ghrelin efficacy. In line with ghrelin's proposed role in glucose metabolism, we find decreased glucose-stimulated insulin secretion in MENX rats, while insulin sensitivity is improved. In summary, we provide a novel nontransgenic rat model with high endogenous ghrelin plasma levels and, interestingly, improved glucose tolerance. This model might aid in identifying new therapeutic approaches for obesity and obesity-related diseases, including type 2 diabetes.
Collapse
Affiliation(s)
- Tobias Wiedemann
- Institute of Pathology, Helmholtz Center Munich, German Research Center for Environmental Health, Technical University Munich, Munich, Germany
| | - Maximilian Bielohuby
- Endocrine Research Unit, Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Ludwigs-Maximilians University, Munich, Germany
| | - Timo D Müller
- Institute for Diabetes and Obesity, Helmholtz Center Munich, German Research Center for Environmental Health, Technical University Munich, Munich, Germany
| | - Martin Bidlingmaier
- Endocrine Research Unit, Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Ludwigs-Maximilians University, Munich, Germany
| | - Natalia S Pellegata
- Institute of Pathology, Helmholtz Center Munich, German Research Center for Environmental Health, Technical University Munich, Munich, Germany
| |
Collapse
|
14
|
Alvarez-Perez JC, Rosa TC, Casinelli GP, Valle SR, Lakshmipathi J, Rosselot C, Rausell-Palamos F, Vasavada RC, García-Ocaña A. Hepatocyte growth factor ameliorates hyperglycemia and corrects β-cell mass in IRS2-deficient mice. Mol Endocrinol 2015; 28:2038-48. [PMID: 25361392 DOI: 10.1210/me.2014-1207] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Insulin resistance, when combined with decreased β-cell mass and relative insufficient insulin secretion, leads to type 2 diabetes. Mice lacking the IRS2 gene (IRS2(-/-) mice) develop diabetes due to uncompensated insulin resistance and β-cell failure. Hepatocyte growth factor (HGF) activates the phosphatidylinositol 3-kinase/Akt signaling pathway in β-cells without recruitment of IRS1 or IRS2 and increases β-cell proliferation, survival, mass, and function when overexpressed in β-cells of transgenic (TG) mice. We therefore hypothesized that HGF may protect against β-cell failure in IRS2 deficiency. For that purpose, we cross-bred TG mice overexpressing HGF in β-cells with IRS2 knockout (KO) mice. Glucose homeostasis analysis revealed significantly reduced hyperglycemia, compensatory hyperinsulinemia, and improved glucose tolerance in TG/KO mice compared with those in KO mice in the context of similar insulin resistance. HGF overexpression also increased glucose-stimulated insulin secretion in IRS2(-/-) islets. To determine whether this glucose homeostasis improvement correlated with alterations in β-cells, we measured β-cell mass, proliferation, and death in these mice. β-Cell proliferation was increased and death was decreased in TG/KO mice compared with those in KO mice. As a result, β-cell mass was significantly increased in TG/KO mice compared with that in KO mice, reaching levels similar to those in wild-type mice. Analysis of the intracellular targets involved in β-cell failure in IRS2 deficiency showed Pdx-1 up-regulation, Akt/FoxO1 phosphorylation, and p27 down-regulation in TG/KO mouse islets. Taken together, these results indicate that HGF can compensate for IRS2 deficiency and subsequent insulin resistance by normalizing β-cell mass and increasing circulating insulin. HGF may be of value as a therapeutic agent against β-cell failure.
Collapse
Affiliation(s)
- Juan C Alvarez-Perez
- Diabetes, Obesity and Metabolism Institute (J.C.A.-P., J.L., C.R., F.R.-P., R.C.V., A.G.-O.), Division of Endocrinology, Diabetes and Bone Diseases, The Mindich Child Health and Development Institute, Icahn School of Medicine at Mt Sinai, New York, New York 10029; and Department of Medicine (T.C.R., G.P.C., S.R.V.), Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Jiménez-Palomares M, López-Acosta JF, Villa-Pérez P, Moreno-Amador JL, Muñoz-Barrera J, Fernández-Luis S, Heras-Pozas B, Perdomo G, Bernal-Mizrachi E, Cózar-Castellano I. Cyclin C stimulates β-cell proliferation in rat and human pancreatic β-cells. Am J Physiol Endocrinol Metab 2015; 308:E450-9. [PMID: 25564474 PMCID: PMC4360017 DOI: 10.1152/ajpendo.00260.2014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Activation of pancreatic β-cell proliferation has been proposed as an approach to replace reduced functional β-cell mass in diabetes. Quiescent fibroblasts exit from G0 (quiescence) to G1 through pRb phosphorylation mediated by cyclin C/cdk3 complexes. Overexpression of cyclin D1, D2, D3, or cyclin E induces pancreatic β-cell proliferation. We hypothesized that cyclin C overexpression would induce β-cell proliferation through G0 exit, thus being a potential therapeutic target to recover functional β-cell mass. We used isolated rat and human islets transduced with adenovirus expressing cyclin C. We measured multiple markers of proliferation: [(3)H]thymidine incorporation, BrdU incorporation and staining, and Ki67 staining. Furthermore, we detected β-cell death by TUNEL, β-cell differentiation by RT-PCR, and β-cell function by glucose-stimulated insulin secretion. Interestingly, we have found that cyclin C increases rat and human β-cell proliferation. This augmented proliferation did not induce β-cell death, dedifferentiation, or dysfunction in rat or human islets. Our results indicate that cyclin C is a potential target for inducing β-cell regeneration.
Collapse
Affiliation(s)
| | | | - Pablo Villa-Pérez
- Instituto de Genética y Biología Molecular, Universidad de Valladolid-CSIC, Valladolid, Spain
| | | | - Jennifer Muñoz-Barrera
- Instituto de Genética y Biología Molecular, Universidad de Valladolid-CSIC, Valladolid, Spain
| | - Sara Fernández-Luis
- Instituto de Genética y Biología Molecular, Universidad de Valladolid-CSIC, Valladolid, Spain
| | - Blanca Heras-Pozas
- Instituto de Genética y Biología Molecular, Universidad de Valladolid-CSIC, Valladolid, Spain
| | - Germán Perdomo
- Departamento de Química Inorgánica, Orgánica y Bioquímica, Universidad de Castilla La Mancha, Spain
| | - Ernesto Bernal-Mizrachi
- Division of Metabolism, Endocrinology and Diabetes, University of Michigan, Ann Arbor, Michigan
| | - Irene Cózar-Castellano
- Instituto de Genética y Biología Molecular, Universidad de Valladolid-CSIC, Valladolid, Spain;
| |
Collapse
|
16
|
Ding Y, Xu Y, Shuai X, Shi X, Chen X, Huang W, Liu Y, Liang X, Zhang Z, Su D. Reg3α Overexpression Protects Pancreatic β Cells from Cytokine-Induced Damage and Improves Islet Transplant Outcome. Mol Med 2015; 20:548-558. [PMID: 25826674 DOI: 10.2119/molmed.2014.00104] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 12/16/2014] [Indexed: 01/12/2023] Open
Abstract
The process of islet transplantation for treating type 1 diabetes has been limited by the high level of graft failure. This may be overcome by locally delivering trophic factors to enhance engraftment. Regenerating islet-derived protein 3α (Reg3α) is a pancreatic secretory protein which functions as an antimicrobial peptide in control of inflammation and cell proliferation. In this study, to investigate whether Reg3α could improve islet engraftment, a marginal mass of syngeneic islets pretransduced with adenoviruses expressing Reg3α or control EGFP were transplanted under the renal capsule of streptozotocin-induced diabetic mice. Mice receiving islets with elevated Reg3α production exhibited significantly lower blood glucose levels (9.057 ± 0.59 mmol/L versus 13.48 ± 0.35 mmol/L, P < 0.05) and improved glucose-stimulated insulin secretion (1.80 ± 0.17 ng/mL versus 1.16 ± 0.16 ng/mL, P < 0.05) compared with the control group. The decline of apoptotic events (0.57% ± 0.15% versus 1.06% ± 0.07%, P < 0.05) and increased β-cell proliferation (0.70% ± 0.10% versus 0.36% ± 0.14%, P < 0.05) were confirmed in islet grafts overexpressing Reg3α by morphometric analysis. Further experiments showed that Reg3α production dramatically protected cultured islets and pancreatic β cells from cytokine-induced apoptosis and the impairment of glucose-stimulated insulin secretion. Moreover, exposure to cytokines led to the activation of MAPKs in pancreatic β cells, which was reversed by Reg3α overexpression in contrast to control group. These results strongly suggest that Reg3α could enhance islet engraftments through its cytoprotective effect and advance the therapeutic efficacy of islet transplantation.
Collapse
Affiliation(s)
- Ying Ding
- Department of Pathology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Yuemei Xu
- Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Xuanyu Shuai
- Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Xuhui Shi
- Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Xiang Chen
- Center of Cellular Therapy, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wenbin Huang
- Department of Pathology, Nanjing First Hospital, Nanjing, China
| | - Yun Liu
- Center of Metabolic Research, Nanjing Medical University, Nanjing, China
| | - Xiubin Liang
- Center of Metabolic Research, Nanjing Medical University, Nanjing, China
| | - Zhihong Zhang
- Department of Pathology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Dongming Su
- Department of Pathology, Nanjing Medical University, Nanjing, China.,Center of Cellular Therapy, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Center of Metabolic Research, Nanjing Medical University, Nanjing, China
| |
Collapse
|
17
|
Rachdi L, Kariyawasam D, Guez F, Aïello V, Arbonés ML, Janel N, Delabar JM, Polak M, Scharfmann R. Dyrk1a haploinsufficiency induces diabetes in mice through decreased pancreatic beta cell mass. Diabetologia 2014; 57:960-9. [PMID: 24477974 DOI: 10.1007/s00125-014-3174-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 01/06/2014] [Indexed: 01/09/2023]
Abstract
AIMS/HYPOTHESIS Growth factors and nutrients are important regulators of pancreatic beta cell mass and function. However, the signalling pathways by which these factors modulate these processes have not yet been fully elucidated. DYRK1A (also named minibrain/MNB) is a member of the dual-specificity tyrosine phosphorylation-regulated kinase (DYRK) family that has been conserved across evolution. A significant amount of data implicates DYRK1A in brain growth and function, as well as in neurodegenerative processes in Alzheimer's disease and Down's syndrome. We investigated here whether DYRK1A would be an attractive candidate for beta cell growth modulation. METHODS To study the role of DYRK1A in beta cell growth, we used Dyrk1a-deficient mice. RESULTS We show that DYRK1A is expressed in pancreatic islets and provide evidence that changes in Dyrk1a gene dosage in mice strongly modulate glycaemia and circulating insulin levels. Specifically, Dyrk1a-haploinsufficient mice show severe glucose intolerance, reduced beta cell mass and decreased beta cell proliferation. CONCLUSIONS/INTERPRETATION Taken together, our data indicate that DYRK1A is a critical kinase for beta cell growth as Dyrk1a-haploinsufficient mice show a diabetic profile.
Collapse
Affiliation(s)
- Latif Rachdi
- INSERM U1016, Institut Cochin, Faculté de Médecine Cochin, Université Paris Descartes, 24 Rue du Faubourg St Jacques, 75014, Paris, France,
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Mussar K, Tucker A, McLennan L, Gearhart A, Jimenez-Caliani AJ, Cirulli V, Crisa L. Macrophage/epithelium cross-talk regulates cell cycle progression and migration in pancreatic progenitors. PLoS One 2014; 9:e89492. [PMID: 24586821 PMCID: PMC3929706 DOI: 10.1371/journal.pone.0089492] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Accepted: 01/21/2014] [Indexed: 01/06/2023] Open
Abstract
Macrophages populate the mesenchymal compartment of all organs during embryogenesis and have been shown to support tissue organogenesis and regeneration by regulating remodeling of the extracellular microenvironment. Whether this mesenchymal component can also dictate select developmental decisions in epithelia is unknown. Here, using the embryonic pancreatic epithelium as model system, we show that macrophages drive the epithelium to execute two developmentally important choices, i.e. the exit from cell cycle and the acquisition of a migratory phenotype. We demonstrate that these developmental decisions are effectively imparted by macrophages activated toward an M2 fetal-like functional state, and involve modulation of the adhesion receptor NCAM and an uncommon "paired-less" isoform of the transcription factor PAX6 in the epithelium. Over-expression of this PAX6 variant in pancreatic epithelia controls both cell motility and cell cycle progression in a gene-dosage dependent fashion. Importantly, induction of these phenotypes in embryonic pancreatic transplants by M2 macrophages in vivo is associated with an increased frequency of endocrine-committed cells emerging from ductal progenitor pools. These results identify M2 macrophages as key effectors capable of coordinating epithelial cell cycle withdrawal and cell migration, two events critical to pancreatic progenitors' delamination and progression toward their differentiated fates.
Collapse
Affiliation(s)
- Kristin Mussar
- Department of Medicine, Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, United States of America
| | - Andrew Tucker
- Department of Medicine, Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, United States of America
| | - Linsey McLennan
- Department of Medicine, Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, United States of America
| | - Addie Gearhart
- Department of Medicine, Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, United States of America
| | - Antonio J. Jimenez-Caliani
- Department of Medicine, Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, United States of America
| | - Vincenzo Cirulli
- Department of Medicine, Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, United States of America
| | - Laura Crisa
- Department of Medicine, Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
19
|
Yang KT, Bayan JA, Zeng N, Aggarwal R, He L, Peng Z, Kassa A, Kim M, Luo Z, Shi Z, Medina V, Boddupally K, Stiles BL. Adult-onset deletion of Pten increases islet mass and beta cell proliferation in mice. Diabetologia 2014; 57:352-61. [PMID: 24162585 PMCID: PMC3918745 DOI: 10.1007/s00125-013-3085-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Accepted: 09/27/2013] [Indexed: 12/31/2022]
Abstract
AIMS/HYPOTHESIS Adult beta cells have a diminished ability to proliferate. Phosphatase and tensin homologue (PTEN) is a lipid phosphatase that antagonises the function of the mitogenic phosphatidylinositol 3-kinase (PI3K) pathway. The objective of this study was to understand the role of PTEN and PI3K signalling in the maintenance of beta cells postnatally. METHODS We developed a Pten (lox/lox); Rosa26 (lacZ); RIP-CreER (+) model that permitted us to induce Pten deletion by treatment with tamoxifen in mature animals. We evaluated islet mass and function as well as beta cell proliferation in 3- and 12-month-old mice treated with tamoxifen (Pten deleted) vs mice treated with vehicle (Pten control). RESULTS Deletion of Pten in juvenile (3-month-old) beta cells significantly induced their proliferation and increased islet mass. The expansion of islet mass occurred concomitantly with the enhanced ability of the Pten-deleted mice to maintain euglycaemia in response to streptozotocin treatment. In older mice (>12 months of age), deletion of Pten similarly increased islet mass and beta cell proliferation. This novel finding suggests that PTEN-regulated mechanisms may override the age-onset diminished ability of beta cells to respond to mitogenic stimulation. We also found that proteins regulating G1/S cell-cycle transition, such as cyclin D1, cyclin D2, p27 and p16, were altered when PTEN was lost, suggesting that they may play a role in PTEN/PI3K-regulated beta cell proliferation in adult tissue. CONCLUSIONS/INTERPRETATION The signals regulated by the PTEN/PI3K pathway are important for postnatal maintenance of beta cells and regulation of their proliferation in adult tissues.
Collapse
Affiliation(s)
- Kai-Ting Yang
- Department of Biochemistry and Molecular Biology, University of Southern California, Los Angeles, CA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Iglesias J, Barg S, Vallois D, Lahiri S, Roger C, Yessoufou A, Pradevand S, McDonald A, Bonal C, Reimann F, Gribble F, Debril MB, Metzger D, Chambon P, Herrera P, Rutter GA, Prentki M, Thorens B, Wahli W. PPARβ/δ affects pancreatic β cell mass and insulin secretion in mice. J Clin Invest 2012; 122:4105-17. [PMID: 23093780 DOI: 10.1172/jci42127] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2012] [Accepted: 07/19/2012] [Indexed: 12/30/2022] Open
Abstract
PPARβ/δ protects against obesity by reducing dyslipidemia and insulin resistance via effects in muscle, adipose tissue, and liver. However, its function in pancreas remains ill defined. To gain insight into its hypothesized role in β cell function, we specifically deleted Pparb/d in the epithelial compartment of the mouse pancreas. Mutant animals presented increased numbers of islets and, more importantly, enhanced insulin secretion, causing hyperinsulinemia. Gene expression profiling of pancreatic β cells indicated a broad repressive function of PPARβ/δ affecting the vesicular and granular compartment as well as the actin cytoskeleton. Analyses of insulin release from isolated PPARβ/δ-deficient islets revealed an accelerated second phase of glucose-stimulated insulin secretion. These effects in PPARβ/δ-deficient islets correlated with increased filamentous actin (F-actin) disassembly and an elevation in protein kinase D activity that altered Golgi organization. Taken together, these results provide evidence for a repressive role for PPARβ/δ in β cell mass and insulin exocytosis, and shed a new light on PPARβ/δ metabolic action.
Collapse
Affiliation(s)
- José Iglesias
- Center for Integrative Genomics, National Research Center Frontiers in Genetics, University of Lausanne, Lausanne, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Blandino-Rosano M, Alejandro EU, Sathyamurthy A, Scheys JO, Gregg B, Chen AY, Rachdi L, Weiss A, Barker DJ, Gould AP, Elghazi L, Bernal-Mizrachi E. Enhanced beta cell proliferation in mice overexpressing a constitutively active form of Akt and one allele of p21Cip. Diabetologia 2012; 55:1380-9. [PMID: 22327314 PMCID: PMC3646796 DOI: 10.1007/s00125-012-2465-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Accepted: 12/19/2011] [Indexed: 12/31/2022]
Abstract
AIMS/HYPOTHESIS The ability of pancreatic beta cells to proliferate is critical both for normal tissue maintenance and in conditions where there is an increased demand for insulin. Protein kinase B(Akt) plays a major role in promoting proliferation in many cell types, including the insulin-producing beta cells. We have previously reported that mice overexpressing a constitutively active form of Akt(caAkt (Tg)) show enhanced beta cell proliferation that is associated with increased protein levels of cyclin D1, cyclin D2 and cyclin-dependent kinase inhibitor 1A (p21(Cip)). In the present study, we sought to assess the mechanisms responsible for augmented p21(Cip) levels in caAkt(Tg) mice and test the role of p21(Cip) in the proliferative responses induced by activation of Akt signalling. METHODS To gain a greater understanding of the relationship between Akt and p21(Cip), we evaluated the mechanisms involved in the modulation of p2(Cip) by Akt and the in vivo role of reduced p21(Cip) in proliferative responses induced by Akt. RESULTS Our experiments showed that Akt signalling regulates p21(Cip) transcription and protein stability. caAkt(Tg) /p21(Cip+/-) mice exhibited fasting and fed hypoglycaemia as well as hyperinsulinaemia when compared with caAkt(Tg) mice. Glucose tolerance tests revealed improved glucose tolerance in caAkt(Tg)/p21(Cip+/-) mice compared with caAkt (Tg). These changes resulted from increased proliferation, survival and beta cell mass in caAkt(Tg)/p21(Cip+/-) compared with caAkt(Tg) mice. CONCLUSIONS/INTERPRETATION Our data indicate that increased p21(Cip) levels in caAkt(Tg) mice act as a compensatory brake, protecting beta cells from unrestrained proliferation. These studies imply that p21(Cip) could play important roles in the adaptive responses of beta cells to proliferate in conditions such as in insulin resistance.
Collapse
Affiliation(s)
- M. Blandino-Rosano
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, Brehm Center for Diabetes Research, University of Michigan Medical Center, Ann Arbor, MI 48109-0678, USA
| | - E. U. Alejandro
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, Brehm Center for Diabetes Research, University of Michigan Medical Center, Ann Arbor, MI 48109-0678, USA
| | - A. Sathyamurthy
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, Brehm Center for Diabetes Research, University of Michigan Medical Center, Ann Arbor, MI 48109-0678, USA
| | - J. O. Scheys
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, Brehm Center for Diabetes Research, University of Michigan Medical Center, Ann Arbor, MI 48109-0678, USA
| | - B. Gregg
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, Brehm Center for Diabetes Research, University of Michigan Medical Center, Ann Arbor, MI 48109-0678, USA
| | - A. Y. Chen
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, Brehm Center for Diabetes Research, University of Michigan Medical Center, Ann Arbor, MI 48109-0678, USA
| | - L. Rachdi
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, Brehm Center for Diabetes Research, University of Michigan Medical Center, Ann Arbor, MI 48109-0678, USA
| | - A. Weiss
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, Brehm Center for Diabetes Research, University of Michigan Medical Center, Ann Arbor, MI 48109-0678, USA
| | - D. J. Barker
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, Brehm Center for Diabetes Research, University of Michigan Medical Center, Ann Arbor, MI 48109-0678, USA
| | - A. P. Gould
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, Brehm Center for Diabetes Research, University of Michigan Medical Center, Ann Arbor, MI 48109-0678, USA
| | - L. Elghazi
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, Brehm Center for Diabetes Research, University of Michigan Medical Center, Ann Arbor, MI 48109-0678, USA
| | - E. Bernal-Mizrachi
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, Brehm Center for Diabetes Research, University of Michigan Medical Center, Ann Arbor, MI 48109-0678, USA
| |
Collapse
|
22
|
Rachdi L, Aïello V, Duvillié B, Scharfmann R. L-leucine alters pancreatic β-cell differentiation and function via the mTor signaling pathway. Diabetes 2012; 61:409-17. [PMID: 22210321 PMCID: PMC3266409 DOI: 10.2337/db11-0765] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Leucine (Leu) is an essential branched-chain amino acid, which activates the mammalian target of rapamycin (mTOR) signaling pathway. The effect of Leu on cell differentiation during embryonic development is unknown. Here, we show that Leu supplementation during pregnancy significantly increased fetal body weight, caused fetal hyperglycemia and hypoinsulinemia, and decreased the relative islet area. We also used rat embryonic pancreatic explant culture for elucidating the mechanism of Leu action on β-cell development. We found that in the presence of Leu, differentiation of pancreatic duodenal homeobox-1-positive progenitor cells into neurogenin3-positive endocrine progenitor cells was inefficient and resulted in decreased β-cell formation. Mechanistically, Leu increases the intracellular levels of hypoxia-inducible factor 1-α, a repressor of endocrine fate in the pancreas, by activating the mTOR complex 1 signaling pathway. Collectively, our findings indicate that Leu supplementation during pregnancy could potentially increase the risk of type 2 diabetes mellitus by inhibiting the differentiation of pancreatic endocrine progenitor cells during a susceptible period of fetal life.
Collapse
Affiliation(s)
- Latif Rachdi
- INSERM U845, Research Center Growth and Signaling, Paris Descartes University, Sorbonne Paris Cité, Necker Hospital, Paris, France.
| | | | | | | |
Collapse
|
23
|
Kim SY, Rane SG. The Cdk4-E2f1 pathway regulates early pancreas development by targeting Pdx1+ progenitors and Ngn3+ endocrine precursors. Development 2011; 138:1903-12. [PMID: 21490060 DOI: 10.1242/dev.061481] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cell division and cell differentiation are intricately regulated processes vital to organ development. Cyclin-dependent kinases (Cdks) are master regulators of the cell cycle that orchestrate the cell division and differentiation programs. Cdk1 is essential to drive cell division and is required for the first embryonic divisions, whereas Cdks 2, 4 and 6 are dispensable for organogenesis but vital for tissue-specific cell development. Here, we illustrate an important role for Cdk4 in regulating early pancreas development. Pancreatic development involves extensive morphogenesis, proliferation and differentiation of the epithelium to give rise to the distinct cell lineages of the adult pancreas. The cell cycle molecules that specify lineage commitment within the early pancreas are unknown. We show that Cdk4 and its downstream transcription factor E2f1 regulate mouse pancreas development prior to and during the secondary transition. Cdk4 deficiency reduces embryonic pancreas size owing to impaired mesenchyme development and fewer Pdx1(+) pancreatic progenitor cells. Expression of activated Cdk4(R24C) kinase leads to increased Nkx2.2(+) and Nkx6.1(+) cells and a rise in the number and proliferation of Ngn3(+) endocrine precursors, resulting in expansion of the β cell lineage. We show that E2f1 binds and activates the Ngn3 promoter to modulate Ngn3 expression levels in the embryonic pancreas in a Cdk4-dependent manner. These results suggest that Cdk4 promotes β cell development by directing E2f1-mediated activation of Ngn3 and increasing the pool of endocrine precursors, and identify Cdk4 as an important regulator of early pancreas development that modulates the proliferation potential of pancreatic progenitors and endocrine precursors.
Collapse
Affiliation(s)
- So Yoon Kim
- Regenerative Biology Section, Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | |
Collapse
|
24
|
Arumugam R, Fleenor D, Lu D, Freemark M. Differential and complementary effects of glucose and prolactin on islet DNA synthesis and gene expression. Endocrinology 2011; 152:856-68. [PMID: 21239441 PMCID: PMC3198965 DOI: 10.1210/en.2010-1258] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The mechanisms by which lactogenic hormones promote β-cell expansion remain poorly understood. Because prolactin (PRL) up-regulates β-cell glucose transporter 2, glucokinase, and pyruvate dehydrogenase activities, we reasoned that glucose availability might mediate or modulate the effects of PRL on β-cell mass. Here, we used male rat islets to show that PRL and glucose have differential but complementary effects on the expression of cell cyclins, cell cycle inhibitors, and various other genes known to regulate β-cell replication, including insulin receptor substrate 2, IGF-II, menin, forkhead box protein M1, tryptophan hydroxylase 1, and the PRL receptor. Differential effects on gene expression are associated with synergistic effects of glucose and PRL on islet DNA synthesis. The effects of PRL on gene expression are mirrored by β-cell overexpression of signal transducer and activator of transcription 5b and are opposed by dexamethasone. An ad-small interfering RNA specific for cyclin D2 attenuates markedly the effects of PRL on islet DNA synthesis. Our studies suggest a new paradigm for the control of β-cell mass and insulin production by hormones and nutrients. PRL up-regulates β-cell glucose uptake and utilization, whereas glucose increases islet PRL receptor expression and potentiates the effects of PRL on cell cycle gene expression and DNA synthesis. These findings suggest novel targets for prevention of neonatal glucose intolerance and gestational diabetes and may provide new insight into the pathogenesis of β-cell hyperplasia in obese subjects with insulin resistance.
Collapse
Affiliation(s)
- Ramamani Arumugam
- Duke University Medical Center, Department of Pediatrics, Durham, North Carolina 27710, USA
| | | | | | | |
Collapse
|
25
|
Köhler CU, Olewinski M, Tannapfel A, Schmidt WE, Fritsch H, Meier JJ. Cell cycle control of β-cell replication in the prenatal and postnatal human pancreas. Am J Physiol Endocrinol Metab 2011; 300:E221-30. [PMID: 20978233 DOI: 10.1152/ajpendo.00496.2010] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
β-Cell regeneration declines with aging, but the molecular mechanisms controlling β-cell replication in humans are not well understood. We compared the expression of selected cell cycle proteins in prenatal and adult tissue and examined the association of these proteins with β-cell replication. Pancreatic tissue from a total of 20 human fetuses and adults was stained for Ki67, cyclin D3, p16 and p27, and insulin. The β-cellular expression of these cell cycle proteins was determined. The frequency of β-cell replication was lower in adult compared with prenatal β-cells (<0.5 vs. 3.4 ± 0.5%, respectively; P < 0.0001). p16 was sporadically expressed in prenatal β-cells (8.0 ± 1.1%) but highly enriched in adult β-cells (63.1 ± 5.2%, P < 0.0001). Likewise, the expression of p27 was much lower in prenatal β-cells (1.7 ± 0.4 vs. 44.1 ± 5.4%, respectively, P < 0.0001), and cyclin D3 expression increased from 24.2 ± 4.1 to 47.25 ± 5.0%, respectively (P < 0.001), with aging. The expression of all three proteins was significantly correlated with each other (P < 0.01 and r > 0.75, respectively). The strong expression of cyclin D3 in adult human β-cells and its correlation to p27 and p16 suggest a positive role in human β-cell cycle regulation. p16 and p27 appear to restrict β-cell replication with aging. The age dependency of cell cycle regulation in human β-cells might explain the reduced β-cell regeneration in adult humans.
Collapse
Affiliation(s)
- Christina U Köhler
- Department of Medicine I, St. Josef-Hospital, Ruhr-University of Bochum, Gudrunstr. 56, 44791 Bochum, Germany
| | | | | | | | | | | |
Collapse
|
26
|
Fajas L, Blanchet E, Annicotte JS. CDK4, pRB and E2F1: connected to insulin. Cell Div 2010; 5:6. [PMID: 20181095 PMCID: PMC2829545 DOI: 10.1186/1747-1028-5-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2010] [Accepted: 02/05/2010] [Indexed: 01/09/2023] Open
Abstract
Pancreatic beta-cells are metabolic sensors involved in the control of glucose homeostasis. This particular cell type controls insulin secretion through a fine-tuned process, which dregulation have important pathological consequences, such as observed during type 2 diabetes. We recently implicated E2F1 in the control of glucose homeostasis. First we showed that E2f1-/- mice have decreased pancreatic size, as the result of impaired postnatal pancreatic growth. We observed in this study that E2F1 was highly expressed in non-proliferating pancreatic beta-cells, suggesting that E2F1, besides the control of beta-cell number could have a role in pancreatic beta-cell function. We demonstrate in our recent study, both in vitro and in vivo that E2F1 directly regulates the expression of Kir6.2, a key component of the KATP channel involved in the regulation of glucose-induced insulin secretion in pancreatic beta-cells. Expression of Kir6.2 is lost in pancreas of E2f1-/- mice, resulting in insulin secretion defects in these mice. Furthermore, we demonstrated by in tissue chromatin immunoprecipitation analysis that regulation of Kir6.2 expression by E2F1 follows the same regulatory pathway that the classical E2F1 target genes, implicating the participation of CDK4 and retinoblastoma protein. Moreover, in this context, E2F1 transcriptional activity is regulated by glucose and insulin through the CDK4-dependent inactivation of the pRB protein. In summary we provide evidence that the CDK4-pRB-E2F1 regulatory pathway is involved in glucose homeostasis. In our recent study we decipher a new function for these factors in the control of insulin secretion and open up new avenues for the treatment of metabolic diseases, in particular type 2 diabetes.
Collapse
Affiliation(s)
- Lluis Fajas
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, F-34298, France.
| | | | | |
Collapse
|
27
|
Fernandez-Marcos PJ, Pantoja C, Gonzalez-Rodriguez A, Martin N, Flores JM, Valverde AM, Hara E, Serrano M. Normal proliferation and tumorigenesis but impaired pancreatic function in mice lacking the cell cycle regulator sei1. PLoS One 2010; 5:e8744. [PMID: 20090907 PMCID: PMC2807453 DOI: 10.1371/journal.pone.0008744] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2009] [Accepted: 12/04/2009] [Indexed: 11/18/2022] Open
Abstract
Sei1 is a positive regulator of proliferation that promotes the assembly of Cdk4-cyclin D complexes and enhances the transcriptional activity of E2f1. The potential oncogenic role of Sei1 is further suggested by its overexpression in various types of human cancers. To study the role of Sei1, we have generated a mouse line deficient for this gene. Sei1-null fibroblasts did not show abnormalities regarding proliferation or susceptibility to neoplastic transformation, nor did we observe defects on Cdk4 complexes or E2f activity. Sei1-null mice were viable, did not present overt pathologies, had a normal lifespan, and had a normal susceptibility to spontaneous and chemically-induced cancer. Pancreatic insulin-producing cells are known to be particularly sensitive to Cdk4-cyclin D and E2f activities, and we have observed that Sei1 is highly expressed in pancreatic islets compared to other tissues. Interestingly, Sei1-null mice present lower number of islets, decreased beta-cell area, impaired insulin secretion, and glucose intolerance. These defects were associated to nuclear accumulation of the cell-cycle inhibitors p21(Cip1) and p27(Kip1) in islet cells. We conclude that Sei1 plays an important role in pancreatic beta-cells, which supports a functional link between Sei1 and the core cell cycle regulators specifically in the context of the pancreas.
Collapse
Affiliation(s)
| | - Cristina Pantoja
- Tumor Suppression Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Agueda Gonzalez-Rodriguez
- Institute of Biomedicine Alberto Sols (CSIC/UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Madrid, Spain
| | - Nicholas Martin
- Cancer Institute of the Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Juana M. Flores
- Department of Animal Surgery and Medicine, Complutense University of Madrid, Madrid, Spain
| | - Angela M. Valverde
- Institute of Biomedicine Alberto Sols (CSIC/UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Madrid, Spain
| | - Eiji Hara
- Cancer Institute of the Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Manuel Serrano
- Tumor Suppression Group, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- * E-mail:
| |
Collapse
|
28
|
Lee JH, Jo J, Hardikar AA, Periwal V, Rane SG. Cdk4 regulates recruitment of quiescent beta-cells and ductal epithelial progenitors to reconstitute beta-cell mass. PLoS One 2010; 5:e8653. [PMID: 20084282 PMCID: PMC2801612 DOI: 10.1371/journal.pone.0008653] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2009] [Accepted: 12/10/2009] [Indexed: 02/06/2023] Open
Abstract
Insulin-producing pancreatic islet β cells (β-cells) are destroyed, severely depleted or functionally impaired in diabetes. Therefore, replacing functional β-cell mass would advance clinical diabetes management. We have previously demonstrated the importance of Cdk4 in regulating β-cell mass. Cdk4-deficient mice display β-cell hypoplasia and develop diabetes, whereas β-cell hyperplasia is observed in mice expressing an active Cdk4R24C kinase. While β-cell replication appears to be the primary mechanism responsible for β-cell mass increase, considerable evidence also supports a contribution from the pancreatic ductal epithelium in generation of new β-cells. Further, while it is believed that majority of β-cells are in a state of ‘dormancy’, it is unclear if and to what extent the quiescent cells can be coaxed to participate in the β-cell regenerative response. Here, we address these queries using a model of partial pancreatectomy (PX) in Cdk4 mutant mice. To investigate the kinetics of the regeneration process precisely, we performed DNA analog-based lineage-tracing studies followed by mathematical modeling. Within a week after PX, we observed considerable proliferation of islet β-cells and ductal epithelial cells. Interestingly, the mathematical model showed that recruitment of quiescent cells into the active cell cycle promotes β-cell mass reconstitution in the Cdk4R24C pancreas. Moreover, within 24–48 hours post-PX, ductal epithelial cells expressing the transcription factor Pdx-1 dramatically increased. We also detected insulin-positive cells in the ductal epithelium along with a significant increase of islet-like cell clusters in the Cdk4R24C pancreas. We conclude that Cdk4 not only promotes β-cell replication, but also facilitates the activation of β-cell progenitors in the ductal epithelium. In addition, we show that Cdk4 controls β-cell mass by recruiting quiescent cells to enter the cell cycle. Comparing the contribution of cell proliferation and islet-like clusters to the total increase in insulin-positive cells suggests a hitherto uncharacterized large non-proliferative contribution.
Collapse
Affiliation(s)
- Ji-Hyeon Lee
- Regenerative Biology Section, Diabetes Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Junghyo Jo
- Laboratory of Biological Modeling, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | | | - Vipul Periwal
- Laboratory of Biological Modeling, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Sushil G. Rane
- Regenerative Biology Section, Diabetes Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
29
|
Tavana O, Puebla-Osorio N, Sang M, Zhu C. Absence of p53-dependent apoptosis combined with nonhomologous end-joining deficiency leads to a severe diabetic phenotype in mice. Diabetes 2010; 59:135-42. [PMID: 19833883 PMCID: PMC2797914 DOI: 10.2337/db09-0792] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
OBJECTIVE Regulation of pancreatic beta-cell mass is essential to preserve sufficient insulin levels for the maintenance of glucose homeostasis. Previously, we reported that DNA double-strand breaks (DSBs) resulting from nonhomologous end-joining (NHEJ) deficiency induce apoptosis and, when combined with p53 deficiency, progressed rapidly into lymphomagenesis in mice. Combination of NHEJ deficiency with a hypomorphic mutation, p53R172P, leads to the abrogation of apoptosis, upregulation of p21, and senescence in precursor lymphocytes. This was sufficient to prevent tumorigenesis. However, these mutant mice succumb to severe diabetes and die at an early age. The aim of this study was to determine the pathogenesis of diabetes in these mutant mice. RESEARCH DESIGN AND METHODS We analyzed the morphology of the pancreatic islets and the function, proliferation rate, and senescence of beta-cells. We also profiled DNA damage and p53 and p21 expression in the pancreas. RESULTS NHEJ-p53R172P mutant mice succumb to diabetes at 3-5 months of age. These mice show a progressive decrease in pancreatic islet mass that is independent of apoptosis and innate immunity. We observed an accumulation of DNA damage, accompanied with increased levels of p53 and p21, a significant decrease in beta-cell proliferation, and cellular senescence in the mutant pancreatic islets. CONCLUSIONS Combined DSBs with an absence of p53-dependent apoptosis activate p53-dependent senescence, which leads to a diminished beta-cell self-replication, massive depletion of the pancreatic islets, and severe diabetes. This is a model that connects impaired DNA repair and accumulative DNA damage, a common phenotype in aging individuals, to the onset of diabetes.
Collapse
Affiliation(s)
- Omid Tavana
- Department of Immunology, University of Texas, M.D. Anderson Cancer Center, Houston, Texas
- University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas
| | - Nahum Puebla-Osorio
- Department of Immunology, University of Texas, M.D. Anderson Cancer Center, Houston, Texas
| | - Mei Sang
- Department of Immunology, University of Texas, M.D. Anderson Cancer Center, Houston, Texas
| | - Chengming Zhu
- Department of Immunology, University of Texas, M.D. Anderson Cancer Center, Houston, Texas
- University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas
- Corresponding author: Chengming Zhu,
| |
Collapse
|
30
|
Annicotte JS, Blanchet E, Chavey C, Iankova I, Costes S, Assou S, Teyssier J, Dalle S, Sardet C, Fajas L. The CDK4-pRB-E2F1 pathway controls insulin secretion. Nat Cell Biol 2009; 11:1017-23. [PMID: 19597485 DOI: 10.1038/ncb1915] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2008] [Accepted: 04/28/2009] [Indexed: 12/20/2022]
Abstract
CDK4-pRB-E2F1 cell-cycle regulators are robustly expressed in non-proliferating beta cells, suggesting that besides the control of beta-cell number the CDK4-pRB-E2F1 pathway has a role in beta-cell function. We show here that E2F1 directly regulates expression of Kir6.2, which is a key component of the K(ATP) channel involved in the regulation of glucose-induced insulin secretion. We demonstrate, through chromatin immunoprecipitation analysis from tissues, that Kir6.2 expression is regulated at the promoter level by the CDK4-pRB-E2F1 pathway. Consistently, inhibition of CDK4, or genetic inactivation of E2F1, results in decreased expression of Kir6.2, impaired insulin secretion and glucose intolerance in mice. Furthermore we show that rescue of Kir6.2 expression restores insulin secretion in E2f1(-/-) beta cells. Finally, we demonstrate that CDK4 is activated by glucose through the insulin pathway, ultimately resulting in E2F1 activation and, consequently, increased expression of Kir6.2. In summary we provide evidence that the CDK4-pRB-E2F1 regulatory pathway is involved in glucose homeostasis, defining a new link between cell proliferation and metabolism.
Collapse
|
31
|
Sachdeva MM, Stoffers DA. Minireview: Meeting the demand for insulin: molecular mechanisms of adaptive postnatal beta-cell mass expansion. Mol Endocrinol 2009; 23:747-58. [PMID: 19196831 DOI: 10.1210/me.2008-0400] [Citation(s) in RCA: 121] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Type 2 diabetes results from pancreatic ss-cell failure in the setting of insulin resistance. This model of disease progression has received recent support from the results of genome-wide association studies that identify genes potentially regulating ss-cell growth and function as type 2 diabetes susceptibility loci. Normal ss-cell compensation for an increased insulin demand includes both enhanced insulin-secretory capacity and an expansion of morphological ss-cell mass, due largely to changes in the balance between ss-cell proliferation and apoptosis. Recent years have brought significant progress in the understanding of both extrinsic signals stimulating ss-cell growth as well as mediators intrinsic to the ss-cell that regulate the compensatory response. Here, we review the current knowledge of mechanisms underlying adaptive expansion of ss-cell mass, focusing on lessons learned from experimental models of physiologically occurring insulin-resistant states including diet-induced obesity and pregnancy, and highlighting the potential importance of interorgan cross talk. The identification of critical mediators of islet compensation may direct the development of future therapeutic strategies to enhance the response of ss-cells to insulin resistance.
Collapse
Affiliation(s)
- Mira M Sachdeva
- Department of Medicine, Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania School of Medicine, Philadelphia, 19104, USA
| | | |
Collapse
|
32
|
Abstract
Type 2 diabetes mellitus is a complex disease characterized by beta-cell failure in the setting of insulin resistance. In early stages of the disease, pancreatic beta-cells adapt to insulin resistance by increasing mass and function. As nutrient excess persists, hyperglycemia and elevated free fatty acids negatively impact beta-cell function. This happens by numerous mechanisms, including the generation of reactive oxygen species, alterations in metabolic pathways, increases in intracellular calcium and the activation of endoplasmic reticulum stress. These processes adversely affect beta-cells by impairing insulin secretion, decreasing insulin gene expression and ultimately causing apoptosis. In this review, we will first discuss the regulation of beta-cell mass during normal conditions. Then, we will discuss the mechanisms of beta-cell failure, including glucotoxicity, lipotoxicity and endoplasmic reticulum stress. Further research into mechanisms will reveal the key modulators of beta-cell failure and thus identify possible novel therapeutic targets. Type 2 diabetes mellitus is a multifactorial disease that has greatly risen in prevalence in part due to the obesity and inactivity that characterize the modern Western lifestyle. Pancreatic beta-cells possess the potential to greatly expand their function and mass in both physiologic and pathologic states of nutrient excess and increased insulin demand. beta-cell response to nutrient excess occurs by several mechanisms, including hypertrophy and proliferation of existing beta-cells, increased insulin production and secretion, and formation of new beta-cells from progenitor cells [1, 2]. Failure of pancreatic beta-cells to adequately expand in settings of increased insulin demand results in hyperglycemia and diabetes. In this review, we will first discuss the factors involved in beta-cell growth and then discuss the mechanisms by which beta-cell expansion fails and leads to beta-cell failure and diabetes (Fig. 1).
Collapse
Affiliation(s)
- KJ Chang-Chen
- Washington University School of Medicine, Division of Endocrinology, Metabolism & Lipid Research, St. Louis, Missouri, USA
| | - R Mullur
- Division of Medical Education, Department of Internal Medicine, Washington University School of Medicine
| | - E Bernal-Mizrachi
- To whom correspondence should be addressed: Ernesto Bernal-Mizrachi, Washington University School of Medicine, 660 S. Euclid Avenue, Campus Box 8127, St. Louis, MO 63110, USA. Phone: (314) 362-7693 Fax: (314) 747-2692
| |
Collapse
|
33
|
Agudo J, Ayuso E, Jimenez V, Salavert A, Casellas A, Tafuro S, Haurigot V, Ruberte J, Segovia JC, Bueren J, Bosch F. IGF-I mediates regeneration of endocrine pancreas by increasing beta cell replication through cell cycle protein modulation in mice. Diabetologia 2008; 51:1862-72. [PMID: 18663428 DOI: 10.1007/s00125-008-1087-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2008] [Accepted: 06/09/2008] [Indexed: 12/30/2022]
Abstract
AIMS/HYPOTHESIS Recovery from diabetes requires restoration of beta cell mass. Igf1 expression in beta cells of transgenic mice regenerates the endocrine pancreas during type 1 diabetes. However, the IGF-I-mediated mechanism(s) restoring beta cell mass are not fully understood. Here, we examined the contribution of pre-existing beta cell proliferation and transdifferentiation of progenitor cells from bone marrow in IGF-I-induced islet regeneration. METHODS Streptozotocin (STZ)-treated Igf1-expressing transgenic mice transplanted with green fluorescent protein (GFP)-expressing bone marrow cells were used. Bone marrow cell transdifferentiation and beta cell replication were measured by GFP/insulin and by the antigen identified by monoclonal antibody Ki67/insulin immunostaining of pancreatic sections respectively. Key cell cycle proteins were measured by western blot, quantitative RT-PCR and immunohistochemistry. RESULTS Despite elevated IGF-I production, recruitment and differentiation of bone marrow cells to beta cells was not increased either in healthy or STZ-treated transgenic mice. In contrast, after STZ treatment, IGF-I overproduction decreased beta cell apoptosis and increased beta cell replication by modulating key cell cycle proteins. Decreased nuclear levels of cyclin-dependent kinase inhibitor 1B (p27) and increased nuclear localisation of cyclin-dependent kinase (CDK)-4 were consistent with increased beta cell proliferation. However, islet expression of cyclin D1 increased only after STZ treatment. In contrast, higher levels of cyclin-dependent kinase inhibitor 1A (p21) were detected in islets from non-STZ-treated transgenic mice. CONCLUSIONS/INTERPRETATION These findings indicate that IGF-I modulates cell cycle proteins and increases replication of pre-existing beta cells after damage. Therefore, our study suggests that local production of IGF-I may be a safe approach to regenerate endocrine pancreas to reverse diabetes.
Collapse
Affiliation(s)
- J Agudo
- Center of Animal Biotechnology and Gene Therapy, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Elghazi L, Rachdi L, Weiss AJ, Cras-Méneur C, Bernal-Mizrachi E. Regulation of beta-cell mass and function by the Akt/protein kinase B signalling pathway. Diabetes Obes Metab 2007; 9 Suppl 2:147-57. [PMID: 17919189 DOI: 10.1111/j.1463-1326.2007.00783.x] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The insulin receptor substrate-2/phosphoinositide 3-kinase (PI3K) pathway plays a critical role in the regulation of beta-cell mass and function, demonstrated both in vitro and in vivo. The serine threonine kinase Akt is one of the promising downstream molecules of this pathway that has been identified as a potential target to regulate function and induce proliferation and survival of beta cells. Here we summarize some of the molecular mechanisms, downstream signalling pathways and critical components involved in the regulation of beta-cell mass and function by Akt.
Collapse
Affiliation(s)
- L Elghazi
- Department of Internal Medicine, Division of Endocrinology, Washington University School of Medicine, Metabolism & Lipid Research, St Louis, MO 63110, USA
| | | | | | | | | |
Collapse
|
35
|
β-cell regeneration: Neogenesis, replication or both? J Mol Med (Berl) 2007; 86:247-58. [DOI: 10.1007/s00109-007-0259-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2007] [Revised: 07/30/2007] [Accepted: 08/21/2007] [Indexed: 12/11/2022]
|
36
|
Gupta RK, Gao N, Gorski RK, White P, Hardy OT, Rafiq K, Brestelli JE, Chen G, Stoeckert CJ, Kaestner KH. Expansion of adult beta-cell mass in response to increased metabolic demand is dependent on HNF-4alpha. Genes Dev 2007; 21:756-69. [PMID: 17403778 PMCID: PMC1838528 DOI: 10.1101/gad.1535507] [Citation(s) in RCA: 134] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The failure to expand functional pancreatic beta-cell mass in response to increased metabolic demand is a hallmark of type 2 diabetes. Lineage tracing studies indicate that replication of existing beta-cells is the principle mechanism for beta-cell expansion in adult mice. Here we demonstrate that the proliferative response of beta-cells is dependent on the orphan nuclear receptor hepatocyte nuclear factor-4alpha (HNF-4alpha), the gene that is mutated in Maturity-Onset Diabetes of the Young 1 (MODY1). Computational analysis of microarray expression profiles from isolated islets of mice lacking HNF-4alpha in pancreatic beta-cells reveals that HNF-4alpha regulates selected genes in the beta-cell, many of which are involved in proliferation. Using a physiological model of beta-cell expansion, we show that HNF-4alpha is required for beta-cell replication and the activation of the Ras/ERK signaling cascade in islets. This phenotype correlates with the down-regulation of suppression of tumorigenicity 5 (ST5) in HNF-4alpha mutants, which we identify as a novel regulator of ERK phosphorylation in beta-cells and a direct transcriptional target of HNF-4alpha in vivo. Together, these results indicate that HNF-4alpha is essential for the physiological expansion of adult beta-cell mass in response to increased metabolic demand.
Collapse
Affiliation(s)
- Rana K. Gupta
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Nan Gao
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Regina K. Gorski
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
- Center for Bioinformatics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Peter White
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Olga T. Hardy
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Kiran Rafiq
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - John E. Brestelli
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Guang Chen
- Center for Bioinformatics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Christian J. Stoeckert
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
- Center for Bioinformatics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Klaus H. Kaestner
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
- Corresponding author.E-MAIL ; FAX (215) 573-5892
| |
Collapse
|