1
|
Zhang Z, Piro AL, Allalou A, Alexeeff SE, Dai FF, Gunderson EP, Wheeler MB. Prolactin and Maternal Metabolism in Women With a Recent GDM Pregnancy and Links to Future T2D: The SWIFT Study. J Clin Endocrinol Metab 2022; 107:2652-2665. [PMID: 35666146 PMCID: PMC9387721 DOI: 10.1210/clinem/dgac346] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Indexed: 11/19/2022]
Abstract
CONTEXT Prolactin is a multifaceted hormone known to regulate lactation. In women with gestational diabetes mellitus (GDM) history, intensive lactation has been associated with lower relative risk of future type 2 diabetes (T2D). However, the role of prolactin in T2D development and maternal metabolism in women with a recent GDM pregnancy has not been ascertained. OBJECTIVE We examined the relationships among prolactin, future T2D risk, and key clinical and metabolic parameters. METHODS We utilized a prospective GDM research cohort (the SWIFT study) and followed T2D onset by performing 2-hour 75-g research oral glucose tolerance test (OGTT) at study baseline (6-9 weeks postpartum) and again annually for 2 years, and also by retrieving clinical diagnoses of T2D from 2 years through 10 years of follow up from electronic medical records. Targeted metabolomics and lipidomics were applied on fasting plasma samples collected at study baseline from 2-hour 75-g research OGTTs in a nested case-control study (100 future incident T2D cases vs 100 no T2D controls). RESULTS Decreasing prolactin quartiles were associated with increased future T2D risk (adjusted odds ratio 2.48; 95% CI, 0.81-7.58; P = 0.05). In women who maintained normoglycemia during the 10-year follow-up period, higher prolactin at baseline was associated with higher insulin sensitivity (P = 0.038) and HDL-cholesterol (P = 0.01), but lower BMI (P = 0.001) and leptin (P = 0.002). Remarkably, among women who developed future T2D, prolactin was not correlated with a favorable metabolic status (all P > 0.05). Metabolomics and lipidomics showed that lower circulating prolactin strongly correlated with a T2D-high risk lipid profile, with elevated circulating neutral lipids and lower concentrations of specific phospholipids/sphingolipids. CONCLUSION In women with recent GDM pregnancy, low circulating prolactin is associated with specific clinical and metabolic parameters and lipid metabolites linked to a high risk of developing T2D.
Collapse
Affiliation(s)
- Ziyi Zhang
- Department of Physiology, Faculty of Medicine, University of Toronto, Ontario M5S 1A8, Canada
- Department of Endocrinology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, Zhejiang 310016, China
| | - Anthony L Piro
- Department of Physiology, Faculty of Medicine, University of Toronto, Ontario M5S 1A8, Canada
| | - Amina Allalou
- Department of Physiology, Faculty of Medicine, University of Toronto, Ontario M5S 1A8, Canada
| | - Stacey E Alexeeff
- Division of Research, Kaiser Permanente Northern California, Oakland, CA 94612, USA
| | - Feihan F Dai
- Correspondence: Feihan F. Dai, PhD, Department of Physiology, Faculty of Medicine, University of Toronto, 1 King’s College Circle, M5S 1A8 Ontario, Canada.
| | - Erica P Gunderson
- Correspondence: Erica P. Gunderson, PhD, MS, MPH, Division of Research, Kaiser Permanente Northern California, 1 King’s College Circle, M5S 1A8 Oakland, CA, USA.
| | - Michael B Wheeler
- Correspondence: Michael B. Wheeler, PhD, Department of Physiology, Faculty of Medicine, University of Toronto, 1 King’s College Circle, M5S 1A8 Ontario, Canada.
| |
Collapse
|
2
|
Abderrahmani A, Jacovetti C, Regazzi R. Lessons from neonatal β-cell epigenomic for diabetes prevention and treatment. Trends Endocrinol Metab 2022; 33:378-389. [PMID: 35382967 DOI: 10.1016/j.tem.2022.03.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/08/2022] [Accepted: 03/10/2022] [Indexed: 12/30/2022]
Abstract
Pancreatic β-cell expansion and functional maturation during the birth-to-weaning period plays an essential role in the adaptation of plasma insulin levels to metabolic needs. These events are driven by epigenetic programs triggered by growth factors, hormones, and nutrients. These mechanisms operating in the neonatal period can be at least in part reactivated in adult life to increase the functional β-cell mass and face conditions of increased insulin demand such as obesity or pregnancy. In this review, we will highlight the importance of studying these signaling pathways and epigenetic programs to understand the causes of different forms of diabetes and to permit the design of novel therapeutic strategies to prevent and treat this metabolic disorder affecting hundreds of millions of people worldwide.
Collapse
Affiliation(s)
- Amar Abderrahmani
- Universitéde Lille, CNRS, Centrale Lille, Université Polytechnique Hauts-de-France, UMR 8520 - IEMN, F-59000 Lille, France.
| | - Cécile Jacovetti
- Department of Fundamental Neuroscience, University of Lausanne, 1005 Lausanne, Switzerland
| | - Romano Regazzi
- Department of Fundamental Neuroscience, University of Lausanne, 1005 Lausanne, Switzerland; Department of Biomedical Science, University of Lausanne, 1005 Lausanne, Switzerland.
| |
Collapse
|
3
|
Shcheglova E, Blaszczyk K, Borowiak M. Mitogen Synergy: An Emerging Route to Boosting Human Beta Cell Proliferation. Front Cell Dev Biol 2022; 9:734597. [PMID: 35155441 PMCID: PMC8829426 DOI: 10.3389/fcell.2021.734597] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 08/24/2021] [Indexed: 12/11/2022] Open
Abstract
Decreased number and function of beta cells are a key aspect of diabetes mellitus (diabetes), a disease that remains an onerous global health problem. Means of restoring beta cell mass are urgently being sought as a potential cure for diabetes. Several strategies, such as de novo beta cell derivation via pluripotent stem cell differentiation or mature somatic cell transdifferentiation, have yielded promising results. Beta cell expansion is another promising strategy, rendered challenging by the very low proliferative capacity of beta cells. Many effective mitogens have been identified in rodents, but the vast majority do not have similar mitogenic effects in human beta cells. Extensive research has led to the identification of several human beta cell mitogens, but their efficacy and specificity remain insufficient. An approach based on the simultaneous application of several mitogens has recently emerged and can yield human beta cell proliferation rates of up to 8%. Here, we discuss recent advances in restoration of the beta cell population, focusing on mitogen synergy, and the contribution of RNA-sequencing (RNA-seq) to accelerating the elucidation of signaling pathways in proliferating beta cells and the discovery of novel mitogens. Together, these approaches have taken beta cell research up a level, bringing us closer to a cure for diabetes.
Collapse
Affiliation(s)
- Ekaterina Shcheglova
- Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
| | - Katarzyna Blaszczyk
- Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
| | - Malgorzata Borowiak
- Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
- *Correspondence: Malgorzata Borowiak, ;
| |
Collapse
|
4
|
Wu HY, Zhang XC, Jia BB, Cao Y, Yan K, Li JY, Tao L, Jie ZG, Liu QW. Exosomes derived from human umbilical cord mesenchymal stem cells alleviate acetaminophen-induced acute liver failure through activating ERK and IGF-1R/PI3K/AKT signaling pathway. J Pharmacol Sci 2021; 147:143-155. [PMID: 34294366 DOI: 10.1016/j.jphs.2021.06.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 06/11/2021] [Accepted: 06/15/2021] [Indexed: 12/16/2022] Open
Abstract
This study aimed to investigate the therapeutic potential of human umbilical cord mesenchymal stem cells derived exosomes (hUCMSC-Exo) in acute liver failure (ALF) in mice as well as its underlying mechanism. We found that a single tail vein administration of hucMSC-Exo effectively enhanced the survival rate, inhibited apoptosis in hepatocytes, and improved liver function in APAP-induced mouse model of ALF. Furthermore, the deletion of glutathione (GSH) and superoxide dismutase (SOD), generation of malondialdehyde (MDA), and the over production of cytochrome P450 E1 (CYP2E1) and 4-hydroxynonenal (4-HNE) caused by APAP were also inhibited by hucMSC-Exo, indicating that hucMSC-Exo inhibited APAP-induced apoptosis of hepatocytes by reducing oxidative stress. Moreover, hucMSC-Exo significantly down-regulated the levels of inflammatory cytokines IL-6, IL-1β, and TNF-α in APAP-treated livers. Western blot showed that hucMSC-Exo significantly promoted the activation of ERK1/2 and IGF-1R/PI3K/AKT signaling pathways in APAP-injured LO2 cells, resulting in the inhibition of apoptosis of LO2 cells. Importantly, PI3K inhibitor LY294002 and ERK1/2 inhibitor PD98059 could reverse the function of hucMSC-Exo on APAP-injured LO2 cells in some extent. Our results suggest that hucMSC-Exo offer antioxidant hepatoprotection against APAP in vitro and in vivo by inhibitiing oxidative stress-induced apoptosis via upregulation of ERK1/2 and PI3K/AKT signaling pathways.
Collapse
Affiliation(s)
- Han-You Wu
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, 330031, PR China
| | - Xiang-Cheng Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, PR China
| | - Bing-Bing Jia
- Zhejiang Provincial Key Lab of Geriatrics, Department of Geriatrics, Zhejiang Hospital, Hangzhou, 310013, PR China
| | - Ye Cao
- Information Engineering School of NanChang University, Nanchang, 330031, PR China
| | - Kai Yan
- Department of Pediatrics, The First Affiliated Hospital of NanChang University, NanChang, 330006, PR China
| | - Jing-Yuan Li
- School of Chemistry, Biology and Material Science, East China University of Technology, Nanchang, 330013, China
| | - Li Tao
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, PR China
| | - Zhi-Gang Jie
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, PR China
| | - Quan-Wen Liu
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang, 330031, PR China.
| |
Collapse
|
5
|
Docherty FM, Sussel L. Islet Regeneration: Endogenous and Exogenous Approaches. Int J Mol Sci 2021; 22:ijms22073306. [PMID: 33804882 PMCID: PMC8037662 DOI: 10.3390/ijms22073306] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/17/2021] [Accepted: 03/17/2021] [Indexed: 02/07/2023] Open
Abstract
Both type 1 and type 2 diabetes are characterized by a progressive loss of beta cell mass that contributes to impaired glucose homeostasis. Although an optimal treatment option would be to simply replace the lost cells, it is now well established that unlike many other organs, the adult pancreas has limited regenerative potential. For this reason, significant research efforts are focusing on methods to induce beta cell proliferation (replication of existing beta cells), promote beta cell formation from alternative endogenous cell sources (neogenesis), and/or generate beta cells from pluripotent stem cells. In this article, we will review (i) endogenous mechanisms of beta cell regeneration during steady state, stress and disease; (ii) efforts to stimulate endogenous regeneration and transdifferentiation; and (iii) exogenous methods of beta cell generation and transplantation.
Collapse
|
6
|
Karakose E, Wang H, Inabnet W, Thakker RV, Libutti S, Fernandez-Ranvier G, Suh H, Stevenson M, Kinoshita Y, Donovan M, Antipin Y, Li Y, Liu X, Jin F, Wang P, Uzilov A, Argmann C, Schadt EE, Stewart AF, Scott DK, Lambertini L. Aberrant methylation underlies insulin gene expression in human insulinoma. Nat Commun 2020; 11:5210. [PMID: 33060578 PMCID: PMC7566641 DOI: 10.1038/s41467-020-18839-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 09/16/2020] [Indexed: 12/23/2022] Open
Abstract
Human insulinomas are rare, benign, slowly proliferating, insulin-producing beta cell tumors that provide a molecular "recipe" or "roadmap" for pathways that control human beta cell regeneration. An earlier study revealed abnormal methylation in the imprinted p15.5-p15.4 region of chromosome 11, known to be abnormally methylated in another disorder of expanded beta cell mass and function: the focal variant of congenital hyperinsulinism. Here, we compare deep DNA methylome sequencing on 19 human insulinomas, and five sets of normal beta cells. We find a remarkably consistent, abnormal methylation pattern in insulinomas. The findings suggest that abnormal insulin (INS) promoter methylation and altered transcription factor expression create alternative drivers of INS expression, replacing canonical PDX1-driven beta cell specification with a pathological, looping, distal enhancer-based form of transcriptional regulation. Finally, NFaT transcription factors, rather than the canonical PDX1 enhancer complex, are predicted to drive INS transactivation.
Collapse
Affiliation(s)
- Esra Karakose
- From the Diabetes Obesity and Metabolism Institute, The Department of Surgery, The Department of Pathology, The Department of Genetics and Genomics Sciences and The Institute for Genomics and Multiscale Biology, The Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | | | - William Inabnet
- From the Diabetes Obesity and Metabolism Institute, The Department of Surgery, The Department of Pathology, The Department of Genetics and Genomics Sciences and The Institute for Genomics and Multiscale Biology, The Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Rajesh V Thakker
- The Academic Endocrine Unit, University of Oxford, OX3 7LJ, Oxford, UK
| | - Steven Libutti
- The Cancer Institute of New Jersey, New Brunswick, NJ, 08901, USA
| | - Gustavo Fernandez-Ranvier
- From the Diabetes Obesity and Metabolism Institute, The Department of Surgery, The Department of Pathology, The Department of Genetics and Genomics Sciences and The Institute for Genomics and Multiscale Biology, The Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Hyunsuk Suh
- From the Diabetes Obesity and Metabolism Institute, The Department of Surgery, The Department of Pathology, The Department of Genetics and Genomics Sciences and The Institute for Genomics and Multiscale Biology, The Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Mark Stevenson
- The Academic Endocrine Unit, University of Oxford, OX3 7LJ, Oxford, UK
| | - Yayoi Kinoshita
- From the Diabetes Obesity and Metabolism Institute, The Department of Surgery, The Department of Pathology, The Department of Genetics and Genomics Sciences and The Institute for Genomics and Multiscale Biology, The Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Michael Donovan
- From the Diabetes Obesity and Metabolism Institute, The Department of Surgery, The Department of Pathology, The Department of Genetics and Genomics Sciences and The Institute for Genomics and Multiscale Biology, The Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Yevgeniy Antipin
- From the Diabetes Obesity and Metabolism Institute, The Department of Surgery, The Department of Pathology, The Department of Genetics and Genomics Sciences and The Institute for Genomics and Multiscale Biology, The Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Sema4, Stamford, CT, 06902, USA
| | - Yan Li
- The Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Xiaoxiao Liu
- The Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Fulai Jin
- The Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Peng Wang
- From the Diabetes Obesity and Metabolism Institute, The Department of Surgery, The Department of Pathology, The Department of Genetics and Genomics Sciences and The Institute for Genomics and Multiscale Biology, The Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Andrew Uzilov
- From the Diabetes Obesity and Metabolism Institute, The Department of Surgery, The Department of Pathology, The Department of Genetics and Genomics Sciences and The Institute for Genomics and Multiscale Biology, The Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Sema4, Stamford, CT, 06902, USA
| | - Carmen Argmann
- From the Diabetes Obesity and Metabolism Institute, The Department of Surgery, The Department of Pathology, The Department of Genetics and Genomics Sciences and The Institute for Genomics and Multiscale Biology, The Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Eric E Schadt
- From the Diabetes Obesity and Metabolism Institute, The Department of Surgery, The Department of Pathology, The Department of Genetics and Genomics Sciences and The Institute for Genomics and Multiscale Biology, The Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Sema4, Stamford, CT, 06902, USA
| | - Andrew F Stewart
- From the Diabetes Obesity and Metabolism Institute, The Department of Surgery, The Department of Pathology, The Department of Genetics and Genomics Sciences and The Institute for Genomics and Multiscale Biology, The Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| | - Donald K Scott
- From the Diabetes Obesity and Metabolism Institute, The Department of Surgery, The Department of Pathology, The Department of Genetics and Genomics Sciences and The Institute for Genomics and Multiscale Biology, The Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Luca Lambertini
- From the Diabetes Obesity and Metabolism Institute, The Department of Surgery, The Department of Pathology, The Department of Genetics and Genomics Sciences and The Institute for Genomics and Multiscale Biology, The Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| |
Collapse
|
7
|
Hu H, Shao D, Wang L, He F, Huang X, Lu Y, Xiang X, Zhu S, Zhang P, Li J, Chen J. Phospho‑regulation of Cdc14A by polo‑like kinase 1 is involved in β‑cell function and cell cycle regulation. Mol Med Rep 2019; 20:4277-4284. [PMID: 31545409 DOI: 10.3892/mmr.2019.10653] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 03/29/2019] [Indexed: 11/06/2022] Open
Abstract
The objective of the present study was to investigate the effects of polo‑like kinase 1 (PLK1) and the phosphorylation of human cell division cycle protein 14A (Cdc14A) by PLK1 on β‑cell function and cell cycle regulation. Mouse β‑TC3 cells were incubated with small interfering RNA (siRNA) to knock down the expression of PLK1. Cell cycle analysis was performed using flow cytometry, and cell proliferation and apoptosis was determined. Insulin secretion was evaluated by a radioimmunoassay under both low and high glucose conditions. Mouse β‑TC3 cells were transfected with a wild type or a non‑phosphorylatable Cdc14A mutant (Cdc14AS351A/363A; Cdc14AAA) to investigate whether the phosphorylation of Cdc14A is involved in cellular regulation of PLK1 under high glucose conditions. It was found that PLK1 siRNA significantly promoted cellular apoptosis, inhibited cell proliferation, decreased insulin secretion and reduced Cdc14A expression under both low and high glucose conditions. Cdc14A overexpression promoted β‑TC3 cell proliferation and insulin secretion, while Cdc14AAA overexpression inhibited cell proliferation and insulin secretion under high glucose conditions. PLK1 siRNA partially reversed the proliferation‑promoting effects of Cdc14A and further intensified the inhibition of proliferation by Cdc14AAA under high glucose conditions. Similarly, Cdc14A overexpression partially reversed the insulin‑inhibiting effects of PLK1 siRNA, while Cdc14AAA overexpression showed a synergistic inhibitory effect on insulin secretion with PLK1 siRNA under high glucose conditions. In conclusion, PLK1 promoted cell proliferation and insulin secretion while inhibiting cellular apoptosis in β‑TC3 cell lines under both low and high glucose conditions. In addition, the phospho‑regulation of Cdc14A by PLK1 may be involved in β‑TC3 cell cycle regulation and insulin secretion under high glucose conditions.
Collapse
Affiliation(s)
- Haiying Hu
- Department of Clinical Nutrition, The Second Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Dandan Shao
- Department of Clinical Nutrition, The Second Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Leilei Wang
- Department of Clinical Nutrition, The Second Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Fang He
- Department of Clinical Nutrition, The Second Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Xiaoxu Huang
- Department of Clinical Nutrition, The Second Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Yanyu Lu
- Department of Clinical Nutrition, The Second Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Xiaona Xiang
- Department of Clinical Nutrition, The Second Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Susu Zhu
- Department of Clinical Nutrition, The Second Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Pianhong Zhang
- Department of Clinical Nutrition, The Second Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Jianru Li
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Jingsen Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| |
Collapse
|
8
|
Abstract
PURPOSE OF REVIEW Pancreatic β-cells play a critical role in whole-body glucose homeostasis by regulating the release of insulin in response to minute by minute alterations in metabolic demand. As such, β-cells are staunchly resilient but there are circumstances where they can become functionally compromised or physically lost due to pathophysiological changes which culminate in overt hyperglycemia and diabetes. RECENT FINDINGS In humans, β-cell mass appears to be largely defined in the postnatal period and this early replicative and generative phase is followed by a refractory state which persists throughout life. Despite this, efforts to identify physiological and pharmacological factors which might re-initiate β-cell replication (or cause the replenishment of β-cells by neogenesis or transdifferentiation) are beginning to bear fruit. Controlled manipulation of β-cell mass in humans still represents a holy grail for therapeutic intervention in diabetes, but progress is being made which may lead to ultimate success.
Collapse
Affiliation(s)
- Giorgio Basile
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA 02215, USA
| | - Rohit N. Kulkarni
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA 02215, USA
| | - Noel G. Morgan
- Institute of Biomedical & Clinical Science, University of Exeter Medical School, Exeter EX2 5DW, UK
| |
Collapse
|
9
|
Abstract
The principal role of prolactin in mammals is the regulation of lactation. Prolactin is a hormone that is mainly synthesized and secreted by lactotroph cells in the anterior pituitary gland. Prolactin signalling occurs via a unique transmembrane prolactin receptor (PRL-R). The structure of the PRL-R has now been elucidated and is similar to that of many biologically fundamental receptors of the class 1 haematopoietic cytokine receptor family such as the growth hormone receptor. The PRL-R is expressed in a wide array of tissues, and a growing number of biological processes continue to be attributed to prolactin. In this Review, we focus on the newly discovered roles of prolactin in human health and disease, particularly its involvement in metabolic homeostasis including body weight control, adipose tissue, skin and hair follicles, pancreas, bone, the adrenal response to stress, the control of lactotroph cell homeostasis and maternal behaviour. New data concerning the pathological states of hypoprolactinaemia and hyperprolactinaemia will also be presented and discussed.
Collapse
Affiliation(s)
- Valérie Bernard
- Inserm U1185, Faculté de Médecine Paris Sud, Université Paris-Saclay, Le Kremlin Bicêtre, France
- Hôpital Saint Antoine, Service d'Endocrinologie et des Maladies de la Reproduction, Paris, France
| | - Jacques Young
- Inserm U1185, Faculté de Médecine Paris Sud, Université Paris-Saclay, Le Kremlin Bicêtre, France
- Hôpital Bicêtre, Service d'Endocrinologie et des Maladies de la Reproduction, Paris, France
| | - Nadine Binart
- Inserm U1185, Faculté de Médecine Paris Sud, Université Paris-Saclay, Le Kremlin Bicêtre, France.
| |
Collapse
|
10
|
Trimal K, Shah T, Joshi K, Mulla G. Association of EGF A61G polymorphism and EGF expression with type 2 diabetes mellitus in Indian population. GENE REPORTS 2019. [DOI: 10.1016/j.genrep.2019.100384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
11
|
Dirice E, De Jesus DF, Kahraman S, Basile G, Ng RW, El Ouaamari A, Teo AKK, Bhatt S, Hu J, Kulkarni RN. Human duct cells contribute to β cell compensation in insulin resistance. JCI Insight 2019; 4:99576. [PMID: 30996131 PMCID: PMC6538348 DOI: 10.1172/jci.insight.99576] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 03/05/2019] [Indexed: 12/19/2022] Open
Abstract
The identification of new sources of β cells is an important endeavor with therapeutic implications for diabetes. Insulin resistance, in physiological states such as pregnancy or in pathological states such as type 2 diabetes (T2D), is characterized by a compensatory increase in β cell mass. To explore the existence of a dynamic β cell reserve, we superimposed pregnancy on the liver-specific insulin receptor-KO (LIRKO) model of insulin resistance that already exhibits β cell hyperplasia and used lineage tracing to track the source of new β cells. Although both control and LIRKO mice displayed increased β cell mass in response to the relative insulin resistance of pregnancy, the further increase in mass in the latter supported a dynamic source that could be traced to pancreatic ducts. Two observations support the translational significance of these findings. First, NOD/SCID-γ LIRKO mice that became pregnant following cotransplantation of human islets and human ducts under the kidney capsule showed enhanced β cell proliferation and an increase in ductal cells positive for transcription factors expressed during β cell development. Second, we identified duct cells positive for immature β cell markers in pancreas sections from pregnant humans and in individuals with T2D. Taken together, during increased insulin demand, ductal cells contribute to the compensatory β cell pool by differentiation/neogenesis.
Collapse
Affiliation(s)
- Ercument Dirice
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts. USA
| | - Dario F. De Jesus
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts. USA
- Graduate Program in Areas of Basic and Applied Biology, Abel Salazar Biomedical Sciences Institute, University of Porto, Porto, Portugal
| | - Sevim Kahraman
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts. USA
| | - Giorgio Basile
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts. USA
| | - Raymond W.S. Ng
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts. USA
| | - Abdelfattah El Ouaamari
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts. USA
| | - Adrian Kee Keong Teo
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts. USA
| | - Shweta Bhatt
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts. USA
| | - Jiang Hu
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts. USA
| | - Rohit N. Kulkarni
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts. USA
- Harvard Stem Cell Institute, Boston, Massachusetts, USA
| |
Collapse
|
12
|
Danilova T, Belevich I, Li H, Palm E, Jokitalo E, Otonkoski T, Lindahl M. MANF Is Required for the Postnatal Expansion and Maintenance of Pancreatic β-Cell Mass in Mice. Diabetes 2019; 68:66-80. [PMID: 30305368 DOI: 10.2337/db17-1149] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 09/30/2018] [Indexed: 11/13/2022]
Abstract
Global lack of mesencephalic astrocyte-derived neurotropic factor (MANF) leads to progressive postnatal loss of β-cell mass and insulin-dependent diabetes in mice. Similar to Manf-/- mice, embryonic ablation of MANF specifically from the pancreas results in diabetes. In this study, we assessed the importance of MANF for the postnatal expansion of pancreatic β-cell mass and for adult β-cell maintenance in mice. Detailed analysis of Pdx-1Cre+/- ::Manffl/fl mice revealed mosaic MANF expression in postnatal pancreata and a significant correlation between the number of MANF-positive β-cells and β-cell mass in individual mice. In vitro, recombinant MANF induced β-cell proliferation in islets from aged mice and protected from hyperglycemia-induced endoplasmic reticulum (ER) stress. Consequently, excision of MANF from β-cells of adult MIP-1CreERT::Manffl/fl mice resulted in reduced β-cell mass and diabetes caused largely by β-cell ER stress and apoptosis, possibly accompanied by β-cell dedifferentiation and reduced rates of β-cell proliferation. Thus, MANF expression in adult mouse β-cells is needed for their maintenance in vivo. We also revealed a mechanistic link between ER stress and inflammatory signaling pathways leading to β-cell death in the absence of MANF. Hence, MANF might be a potential target for regenerative therapy in diabetes.
Collapse
Affiliation(s)
- Tatiana Danilova
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Ilya Belevich
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Huini Li
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Erik Palm
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Eija Jokitalo
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Timo Otonkoski
- Research Programs Unit, Molecular Neurology, Biomedicum Stem Cell Center, University of Helsinki, Helsinki, Finland
- Children's Hospital, Helsinki University Central Hospital, Helsinki, Finland
| | - Maria Lindahl
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| |
Collapse
|
13
|
Khatami F, Tavangar SM. Multiple Endocrine Neoplasia Syndromes from Genetic and Epigenetic Perspectives. Biomark Insights 2018; 13:1177271918785129. [PMID: 30013307 PMCID: PMC6043927 DOI: 10.1177/1177271918785129] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 05/24/2018] [Indexed: 12/20/2022] Open
Abstract
Multiple endocrine neoplasia (MEN) syndromes are infrequent inherited disorders in which more than one endocrine glands develop noncancerous (benign) or cancerous (malignant) tumors or grow excessively without forming tumors. There are 3 famous and well-known forms of MEN syndromes (MEN 1, MEN 2A, and MEN 2B) and a newly documented one (MEN4). These syndromes are infrequent and occurred in all ages and both men and women. Usually, germ line mutations that can be resulted in neoplastic transformation of anterior pituitary, parathyroid glands, and pancreatic islets in addition to gastrointestinal tract can be an indicator for MEN1. The medullary thyroid cancer (MTC) in association with pheochromocytoma and/or multiple lesions of parathyroid glands with hyperparathyroidism can be pointer of MEN2 which can be subgrouped into the MEN 2A, MEN 2B, and familial MTC syndromes. There are no distinct biochemical markers that allow identification of familial versus nonfamilial forms of the tumors, but familial MTC usually happens at a younger age than sporadic MTC. The MEN1 gene (menin protein) is in charge of MEN 1 disease, CDNK1B for MEN 4, and RET proto-oncogene for MEN 2. The focus over the molecular targets can bring some hope for both diagnosis and management of MEN syndromes. In the current review, we look at this disease and responsible genes and their cell signaling pathway involved.
Collapse
Affiliation(s)
- Fatemeh Khatami
- Chronic Diseases Research Center, Endocrinology and Metabolism Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Mohammad Tavangar
- Chronic Diseases Research Center, Endocrinology and Metabolism Research Institute, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pathology, Doctor Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
14
|
Rattananinsruang P, Dechsukhum C, Leeanansaksiri W. Establishment of Insulin-Producing Cells From Human Embryonic Stem Cells Underhypoxic Condition for Cell Based Therapy. Front Cell Dev Biol 2018; 6:49. [PMID: 29868580 PMCID: PMC5962719 DOI: 10.3389/fcell.2018.00049] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 04/16/2018] [Indexed: 12/27/2022] Open
Abstract
Diabetes mellitus (DM) is a group of diseases characterized by abnormally high levels of glucose in the blood stream. In developing a potential therapy for diabetic patients, pancreatic cells transplantation has drawn great attention. However, the hinder of cell transplantation for diabetes treatment is insufficient sources of insulin-producing cells. Therefore, new cell based therapy need to be developed. In this regard, human embryonic stem cells (hESCs) may serve as good candidates for this based on their capability of differentiation into various cell types. In this study, we designed a new differentiation protocol that can generate hESC-derived insulin-producing cells (hES-DIPCs) in a hypoxic condition. We also emphasized on the induction of definitive endoderm during embryoid bodies (EBs) formation. After induction of hESCs differentiation into insulin-producing cells (IPCs), the cells obtained from the cultures exhibited pancreas-related genes such as Pdx1, Ngn3, Nkx6.1, GLUT2, and insulin. These cells also showed positive for DTZ-stained cellular clusters and contained ability of insulin secretion in a glucose-dependent manner. After achievement to generated functional hES-DIPCs in vitro, some of the hES-DIPCs were then encapsulated named encapsulated hES-DIPCs. The data showed that the encapsulated cells could possess the function of insulin secretion in a time-dependent manner. The hES-DIPCs and encapsulated hES-DIPCs were then separately transplanted into STZ-induced diabetic mice. The findings showed the significant blood glucose levels regulation capacity and declination of IL-1β concentration in all transplanted mice. These results indicated that both hES-DIPCs and encapsulated hES-DIPCs contained the ability to sustain hyperglycemia condition as well as decrease inflammatory cytokine level in vivo. The findings of this study may apply for generation of a large number of hES-DIPCs in vitro. In addition, the implication of this work is therapeutic value in type I diabetes treatment in the future. The application for type II diabetes treatment remain to be investigated.
Collapse
Affiliation(s)
- Piyaporn Rattananinsruang
- School of Preclinic, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima, Thailand
| | - Chavaboon Dechsukhum
- School of Pathology, Institute of Medicine, Suranaree University of Technology, Nakhon Ratchasima, Thailand
| | - Wilairat Leeanansaksiri
- School of Preclinic, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima, Thailand
| |
Collapse
|
15
|
Abstract
PURPOSE OF REVIEW Inadequate insulin-producing pancreatic β-cell mass is a key feature of both type 1 and type 2 diabetes. Efforts to regenerate β-cell mass from pancreatic precursors may thus ameliorate absolute or relative insulin deficiency, thereby improving glucose homeostasis. A clear understanding of the processes that govern the generation of new β-cells in the mature pancreas will be fundamental to success in this effort. This review discusses the current state of knowledge regarding β-cell regeneration and emphasizes recent studies of significance. RECENT FINDINGS Recent reports demonstrate regenerative potential in the adult human pancreas. Further, they build on the strong existing evidence that proliferation of preexisting β-cells is the predominant source of new β-cells in adulthood by dissecting the cell cycle machinery components and critical signaling pathways required for β-cell proliferation. Finally, β-cell trophic peptides have demonstrated preclinical potential as pharmacologic regenerative agents and may form the basis for clinical interventions in the future. SUMMARY Efforts to augment β-cell regeneration by enhancing β-cell viability and proliferation may lead to novel therapeutic approaches for type 1 and type 2 diabetes. An intimate understanding of the molecular mechanisms underlying the regulation of β-cell proliferation and survival will be fundamental to the optimization of endogenous β-cell regeneration.
Collapse
|
16
|
Epshtein A, Rachi E, Sakhneny L, Mizrachi S, Baer D, Landsman L. Neonatal pancreatic pericytes support β-cell proliferation. Mol Metab 2017; 6:1330-1338. [PMID: 29031732 PMCID: PMC5641631 DOI: 10.1016/j.molmet.2017.07.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 07/13/2017] [Accepted: 07/14/2017] [Indexed: 12/26/2022] Open
Abstract
OBJECTIVE The maintenance and expansion of β-cell mass rely on their proliferation, which reaches its peak in the neonatal stage. β-cell proliferation was found to rely on cells of the islet microenvironment. We hypothesized that pericytes, which are components of the islet vasculature, support neonatal β-cell proliferation. METHODS To test our hypothesis, we combined in vivo and in vitro approaches. Briefly, we used a Diphtheria toxin-based transgenic mouse system to specifically deplete neonatal pancreatic pericytes in vivo. We further cultured neonatal pericytes isolated from the neonatal pancreas and combined the use of a β-cell line and primary cultured mouse β-cells. RESULTS Our findings indicate that neonatal pancreatic pericytes are required and sufficient for β-cell proliferation. We observed impaired proliferation of neonatal β-cells upon in vivo depletion of pancreatic pericytes. Furthermore, exposure to pericyte-conditioned medium stimulated proliferation in cultured β-cells. CONCLUSIONS This study introduces pancreatic pericytes as regulators of neonatal β-cell proliferation. In addition to advancing current understanding of the physiological β-cell replication process, these findings could facilitate the development of protocols aimed at expending these cells as a potential cure for diabetes.
Collapse
Affiliation(s)
- Alona Epshtein
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Eleonor Rachi
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Lina Sakhneny
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shani Mizrachi
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Daria Baer
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Limor Landsman
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
17
|
Assefa Z, Akbib S, Lavens A, Stangé G, Ling Z, Hellemans KH, Pipeleers D. Direct effect of glucocorticoids on glucose-activated adult rat β-cells increases their cell number and their functional mass for transplantation. Am J Physiol Endocrinol Metab 2016; 311:E698-E705. [PMID: 27555297 DOI: 10.1152/ajpendo.00070.2016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 08/17/2016] [Indexed: 01/02/2023]
Abstract
Compounds that increase β-cell number can serve as β-cell replacement therapies in diabetes. In vitro studies have identified several agents that can activate DNA synthesis in primary β-cells but only in small percentages of cells and without demonstration of increases in cell number. We used whole well multiparameter imaging to first screen a library of 1,280 compounds for their ability to recruit adult rat β-cells into DNA synthesis and then assessed influences of stimulatory agents on the number of living cells. The four compounds with highest β-cell recruitment were glucocorticoid (GC) receptor ligands. The GC effect occurred in glucose-activated β-cells and was associated with increased glucose utilization and oxidation. Hydrocortisone and methylprednisolone almost doubled the number of β-cells in 2 wk. The expanded cell population provided an increased functional β-cell mass for transplantation in diabetic animals. These effects are age dependent; they did not occur in neonatal rat β-cells, where GC exposure suppressed basal replication and was cytotoxic. We concluded that GCs can induce the replication of adult rat β-cells through a direct action, with intercellular differences in responsiveness that have been related to differences in glucose activation and in age. These influences can explain variability in GC-induced activation of DNA synthesis in rat and human β-cells. Our study also demonstrated that β-cells can be expanded in vitro to increase the size of metabolically adequate grafts.
Collapse
Affiliation(s)
- Zerihun Assefa
- Diabetes Research Center, Brussels Free University-VUB, University Hospital Brussels, and Center for Beta Cell Therapy, Brussels, Belgium
| | - Sarah Akbib
- Diabetes Research Center, Brussels Free University-VUB, University Hospital Brussels, and Center for Beta Cell Therapy, Brussels, Belgium
| | - Astrid Lavens
- Diabetes Research Center, Brussels Free University-VUB, University Hospital Brussels, and Center for Beta Cell Therapy, Brussels, Belgium
| | - Geert Stangé
- Diabetes Research Center, Brussels Free University-VUB, University Hospital Brussels, and Center for Beta Cell Therapy, Brussels, Belgium
| | - Zhidong Ling
- Diabetes Research Center, Brussels Free University-VUB, University Hospital Brussels, and Center for Beta Cell Therapy, Brussels, Belgium
| | - Karine H Hellemans
- Diabetes Research Center, Brussels Free University-VUB, University Hospital Brussels, and Center for Beta Cell Therapy, Brussels, Belgium
| | - Daniel Pipeleers
- Diabetes Research Center, Brussels Free University-VUB, University Hospital Brussels, and Center for Beta Cell Therapy, Brussels, Belgium
| |
Collapse
|
18
|
Pepaj M, Bredahl MK, Gjerlaugsen N, Thorsby PM. Proteomic analysis of the INS-1E secretome identify novel vitamin D-regulated proteins. Diabetes Metab Res Rev 2016; 32:514-21. [PMID: 26788927 DOI: 10.1002/dmrr.2777] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 11/17/2015] [Accepted: 01/12/2016] [Indexed: 11/07/2022]
Abstract
BACKGROUND Experimental evidence indicates that vitamin D may have a beneficial role in pancreatic β-cell function. METHODS In the present study, stable isotope labelling by amino acids in cell culture (SILAC) in combination with liquid chromatography-tandem mass spectrometry was used to quantitatively assess the impact of the active vitamin D metabolite, 1,25-(OH)2 D3 , on global protein expression in INS-1E cell secretome. RESULTS Twenty-one proteins were found up-regulated (≥1.5 fold changes) and three down-regulated (≤0.67) after treatment of INS-1E cells with 1,25-(OH)2 D3 . Up-regulation of proteins implicated in β-cell growth and proliferation, such as IGF2, IGFBP7 and gelsolin, suggest that 1,25-(OH)2 D3 has a positive effect on β-cell growth and proliferation. Moreover, modulations of several proteins implicated in prohormone processing and insulin exocytosis (IGF2, IGFBP7, Scg5, ProSAAS, Fabp5, Ptprn2 and gelsolin) appear to support the hypothesis that 1,25-(OH)2 D3 plays positive modulatory role in insulin processing and secretion. CONCLUSIONS Together, we reveal a number of novel vitamin D-regulated proteins that may contribute to a better understanding of the reported beneficial effects of vitamin D on pancreatic β-cells. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Milaim Pepaj
- Hormone Laboratory, Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - May K Bredahl
- Hormone Laboratory, Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - Nina Gjerlaugsen
- Hormone Laboratory, Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - Per M Thorsby
- Hormone Laboratory, Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
19
|
Doiron B, Hu W, DeFronzo RA. Beta Cell Formation in vivo Through Cellular Networking, Integration and Processing (CNIP) in Wild Type Adult Mice. Curr Pharm Biotechnol 2016; 17:376-88. [PMID: 26696016 PMCID: PMC5421132 DOI: 10.2174/1389201017666151223124031] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Revised: 11/18/2015] [Accepted: 11/26/2015] [Indexed: 11/22/2022]
Abstract
Insulin replacement therapy is essential in type 1 diabetic individuals and is required in ~40-50% of type 2 diabetics during their lifetime. Prior attempts at beta cell regeneration have relied upon pancreatic injury to induce beta cell proliferation, dedifferentiation and activation of the embryonic pathway, or stem cell replacement. We report an alternative method to transform adult non-stem (somatic) cells into pancreatic beta cells. The Cellular Networking, Integration and Processing (CNIP) approach targets cellular mechanisms involved in pancreatic function in the organ’s adult state and utilizes a synergistic mechanism that integrates three important levels of cellular regulation to induce beta cell formation: (i) glucose metabolism, (ii) membrane receptor function, and (iii) gene transcription. The aim of the present study was to induce pancreatic beta cell formation in vivo in adult animals without stem cells and without dedifferentiating cells to recapitulate the embryonic pathway as previously published (1-3). Our results employing CNIP demonstrate that: (i) insulin secreting cells can be generated in adult pancreatic tissue in vivo and circumvent the problem of generating endocrine (glucagon and somatostatin) cells that exert deleterious effects on glucose homeostasis, and (ii) long-term normalization of glucose tolerance and insulin secretion can be achieved in a wild type diabetic mouse model. The CNIP cocktail has the potential to be used as a preventative or therapeutic treatment or cure for both type 1 and type 2 diabetes.
Collapse
Affiliation(s)
- Bruno Doiron
- Diabetes Division University of Texas Health Science Center 7703 Floyd Curl Drive San Antonio, Texas, 78231.
| | | | | |
Collapse
|
20
|
Speckmann T, Sabatini PV, Nian C, Smith RG, Lynn FC. Npas4 Transcription Factor Expression Is Regulated by Calcium Signaling Pathways and Prevents Tacrolimus-induced Cytotoxicity in Pancreatic Beta Cells. J Biol Chem 2015; 291:2682-95. [PMID: 26663079 DOI: 10.1074/jbc.m115.704098] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Indexed: 12/16/2022] Open
Abstract
Cytosolic calcium influx activates signaling pathways known to support pancreatic beta cell function and survival by modulating gene expression. Impaired calcium signaling leads to decreased beta cell mass and diabetes. To appreciate the causes of these cytotoxic perturbations, a more detailed understanding of the relevant signaling pathways and their respective gene targets is required. In this study, we examined the calcium-induced expression of the cytoprotective beta cell transcription factor Npas4. Pharmacological inhibition implicated the calcineurin, Akt/protein kinase B, and Ca(2+)/calmodulin-dependent protein kinase signaling pathways in the regulation of Npas4 transcription and translation. Both Npas4 mRNA and protein had high turnover rates, and, at the protein level, degradation was mediated via the ubiquitin-proteasome pathway. Finally, beta cell cytotoxicity of the calcineurin inhibitor and immunosuppressant tacrolimus (FK-506) was prevented by Npas4 overexpression. These results delineate the pathways regulating Npas4 expression and stability and demonstrate its importance in clinical settings such as islet transplantation.
Collapse
Affiliation(s)
- Thilo Speckmann
- From the Diabetes Research Program, Child and Family Research Institute, Vancouver, British Columbia V5Z 4H4, Canada and the Department of Surgery and Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia V5Z 1M9, Canada
| | - Paul V Sabatini
- From the Diabetes Research Program, Child and Family Research Institute, Vancouver, British Columbia V5Z 4H4, Canada and the Department of Surgery and Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia V5Z 1M9, Canada
| | - Cuilan Nian
- From the Diabetes Research Program, Child and Family Research Institute, Vancouver, British Columbia V5Z 4H4, Canada and the Department of Surgery and Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia V5Z 1M9, Canada
| | - Riley G Smith
- From the Diabetes Research Program, Child and Family Research Institute, Vancouver, British Columbia V5Z 4H4, Canada and
| | - Francis C Lynn
- From the Diabetes Research Program, Child and Family Research Institute, Vancouver, British Columbia V5Z 4H4, Canada and the Department of Surgery and Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia V5Z 1M9, Canada
| |
Collapse
|
21
|
Song I, Patel O, Himpe E, Muller CJF, Bouwens L. Beta Cell Mass Restoration in Alloxan-Diabetic Mice Treated with EGF and Gastrin. PLoS One 2015; 10:e0140148. [PMID: 26452142 PMCID: PMC4599944 DOI: 10.1371/journal.pone.0140148] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 09/21/2015] [Indexed: 01/19/2023] Open
Abstract
One week of treatment with EGF and gastrin (EGF/G) was shown to restore normoglycemia and to induce islet regeneration in mice treated with the diabetogenic agent alloxan. The mechanisms underlying this regeneration are not fully understood. We performed genetic lineage tracing experiments to evaluate the contribution of beta cell neogenesis in this model. One day after alloxan administration, mice received EGF/G treatment for one week. The treatment could not prevent the initial alloxan-induced beta cell mass destruction, however it did reverse glycemia to control levels within one day, suggesting improved peripheral glucose uptake. In vitro experiments with C2C12 cell line showed that EGF could stimulate glucose uptake with an efficacy comparable to that of insulin. Subsequently, EGF/G treatment stimulated a 3-fold increase in beta cell mass, which was partially driven by neogenesis and beta cell proliferation as assessed by beta cell lineage tracing and BrdU-labeling experiments, respectively. Acinar cell lineage tracing failed to show an important contribution of acinar cells to the newly formed beta cells. No appearance of transitional cells co-expressing insulin and glucagon, a hallmark for alpha-to-beta cell conversion, was found, suggesting that alpha cells did not significantly contribute to the regeneration. An important fraction of the beta cells significantly lost insulin positivity after alloxan administration, which was restored to normal after one week of EGF/G treatment. Alloxan-only mice showed more pronounced beta cell neogenesis and proliferation, even though beta cell mass remained significantly depleted, suggesting ongoing beta cell death in that group. After one week, macrophage infiltration was significantly reduced in EGF/G-treated group compared to the alloxan-only group. Our results suggest that EGF/G-induced beta cell regeneration in alloxan-diabetic mice is driven by beta cell neogenesis, proliferation and recovery of insulin. The glucose-lowering effect of the treatment might play an important role in the regeneration process.
Collapse
Affiliation(s)
- Imane Song
- Cell Differentiation Lab, Vrije Universiteit Brussel (Brussels Free University), Brussels, Belgium
- * E-mail:
| | - Oelfah Patel
- Diabetes Discovery Platform, South African Medical Research Council, Tygerberg, South Africa
| | - Eddy Himpe
- Cell Differentiation Lab, Vrije Universiteit Brussel (Brussels Free University), Brussels, Belgium
| | - Christo J. F. Muller
- Diabetes Discovery Platform, South African Medical Research Council, Tygerberg, South Africa
| | - Luc Bouwens
- Cell Differentiation Lab, Vrije Universiteit Brussel (Brussels Free University), Brussels, Belgium
| |
Collapse
|
22
|
Tiwari S, Roel C, Wills R, Casinelli G, Tanwir M, Takane KK, Fiaschi-Taesch NM. Early and Late G1/S Cyclins and Cdks Act Complementarily to Enhance Authentic Human β-Cell Proliferation and Expansion. Diabetes 2015; 64:3485-98. [PMID: 26159177 PMCID: PMC4876788 DOI: 10.2337/db14-1885] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 06/20/2015] [Indexed: 12/20/2022]
Abstract
β-Cell regeneration is a key goal of diabetes research. Progression through the cell cycle is associated with retinoblastoma protein (pRb) inactivation via sequential phosphorylation by the "early" cyclins and cyclin-dependent kinases (cdks) (d-cyclins cdk4/6) and the "late" cyclins and cdks (cyclin A/E and cdk1/2). In β-cells, activation of either early or late G1/S cyclins and/or cdks is an efficient approach to induce cycle entry, but it is unknown whether the combined expression of early and late cyclins and cdks might have synergistic or additive effects. Thus, we explored whether a combination of both early and late cyclins and cdks might more effectively drive human β-cell cell cycle entry than either group alone. We also sought to determine whether authentic replication with the expansion of adult human β-cells could be demonstrated. Late cyclins and cdks do not traffic in response to the induction of replication by early cyclins and cdks in human β-cells but are capable of nuclear translocation when overexpressed. Early plus late cyclins and cdks, acting via pRb phosphorylation on distinct residues, complementarily induce greater proliferation in human β-cells than either group alone. Importantly, the combination of early and late cyclins and cdks clearly increased human β-cell numbers in vitro. These findings provide additional insight into human β-cell expansion. They also provide a novel tool for assessing β-cell expansion in vitro.
Collapse
Affiliation(s)
- Shiwani Tiwari
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Chris Roel
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Rachel Wills
- Division of Endocrinology and Metabolism, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Gabriella Casinelli
- Division of Endocrinology and Metabolism, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Mansoor Tanwir
- Division of Endocrinology and Metabolism, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Karen K Takane
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Nathalie M Fiaschi-Taesch
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY Division of Endocrinology and Metabolism, University of Pittsburgh School of Medicine, Pittsburgh, PA
| |
Collapse
|
23
|
Leibiger B, Moede T, Paschen M, Yunn NO, Lim JH, Ryu SH, Pereira T, Berggren PO, Leibiger IB. PI3K-C2α Knockdown Results in Rerouting of Insulin Signaling and Pancreatic Beta Cell Proliferation. Cell Rep 2015; 13:15-22. [PMID: 26387957 DOI: 10.1016/j.celrep.2015.08.058] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 07/20/2015] [Accepted: 08/20/2015] [Indexed: 11/26/2022] Open
Abstract
Insulin resistance is a syndrome that affects multiple insulin target tissues, each having different biological functions regulated by insulin. A remaining question is to mechanistically explain how an insulin target cell/tissue can be insulin resistant in one biological function and insulin sensitive in another at the same time. Here, we provide evidence that in pancreatic β cells, knockdown of PI3K-C2α expression results in rerouting of the insulin signal from insulin receptor (IR)-B/PI3K-C2α/PKB-mediated metabolic signaling to IR-B/Shc/ERK-mediated mitogenic signaling, which allows the β cell to switch from a highly glucose-responsive, differentiated state to a proliferative state. Our data suggest the existence of IR-cascade-selective insulin resistance, which allows rerouting of the insulin signal within the same target cell. Hence, factors involved in the rerouting of the insulin signal represent tentative therapeutic targets in the treatment of insulin resistance.
Collapse
Affiliation(s)
- Barbara Leibiger
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Tilo Moede
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Meike Paschen
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Na-Oh Yunn
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| | - Jong Hoon Lim
- Aptamer Initiative Program, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| | - Sung Ho Ryu
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea; Aptamer Initiative Program, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea; Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| | - Teresa Pereira
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Per-Olof Berggren
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, 171 76 Stockholm, Sweden; Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 637 553, Singapore.
| | - Ingo B Leibiger
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, 171 76 Stockholm, Sweden.
| |
Collapse
|
24
|
Kondegowda NG, Fenutria R, Pollack IR, Orthofer M, Garcia-Ocaña A, Penninger JM, Vasavada RC. Osteoprotegerin and Denosumab Stimulate Human Beta Cell Proliferation through Inhibition of the Receptor Activator of NF-κB Ligand Pathway. Cell Metab 2015; 22:77-85. [PMID: 26094891 PMCID: PMC4597781 DOI: 10.1016/j.cmet.2015.05.021] [Citation(s) in RCA: 125] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 03/18/2015] [Accepted: 05/22/2015] [Indexed: 01/06/2023]
Abstract
Diabetes results from a reduction of pancreatic β-cells. Stimulating replication could normalize β-cell mass. However, adult human β-cells are recalcitrant to proliferation. We identified osteoprotegerin, a bone-related decoy receptor, as a β-cell mitogen. Osteoprotegerin was induced by and required for lactogen-mediated rodent β-cell replication. Osteoprotegerin enhanced β-cell proliferation in young, aged, and diabetic mice. This resulted in increased β-cell mass in young mice and significantly delayed hyperglycemia in diabetic mice. Osteoprotegerin stimulated replication of adult human β-cells, without causing dedifferentiation. Mechanistically, osteoprotegerin induced human and rodent β-cell replication by modulating CREB and GSK3 pathways, through binding Receptor Activator of NF-κB (RANK) Ligand (RANKL), a brake in β-cell proliferation. Denosumab, an FDA-approved osteoporosis drug, and RANKL-specific antibody induced human β-cell proliferation in vitro, and in vivo, in humanized mice. Thus, osteoprotegerin and Denosumab prevent RANKL/RANK interaction to stimulate β-cell replication, highlighting the potential for repurposing an osteoporosis drug to treat diabetes.
Collapse
Affiliation(s)
- Nagesha Guthalu Kondegowda
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Division of Endocrinology and Bone Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Rafael Fenutria
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Division of Endocrinology and Bone Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ilana R Pollack
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Division of Endocrinology and Bone Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Michael Orthofer
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Dr. Bohrgasse 3,1030 Vienna, Austria
| | - Adolfo Garcia-Ocaña
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Division of Endocrinology and Bone Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Josef M Penninger
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Dr. Bohrgasse 3,1030 Vienna, Austria
| | - Rupangi C Vasavada
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Division of Endocrinology and Bone Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
25
|
Scheinman EJ, Damouni R, Caspi A, Shen-Orr Z, Tiosano D, LeRoith D. The beneficial effect of growth hormone treatment on islet mass in streptozotocin-treated mice. Diabetes Metab Res Rev 2015; 31:492-9. [PMID: 25529355 DOI: 10.1002/dmrr.2631] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Accepted: 12/09/2014] [Indexed: 12/22/2022]
Abstract
BACKGROUND Type 1 diabetes is an autoimmune disease, characterized by a loss of pancreatic β-cell mass and function, which results in dramatic reductions in insulin secretion and circulating insulin levels. Patients with type 1 diabetes are traditionally treated with insulin injections and insulin pumps ex vivo or undergo transplantation. Growth hormone (GH) has been shown to be involved in β-cell function and survival in culture. METHODS Twelve-week-old female C57BL/6 mice were treated with streptozotocin and monitored for their weight and blood glucose levels. Fourteen days post-initial injection, these mice were separated into two groups at random. One group was treated with GH while the other treated with vehicle for up to 3 weeks. These mice were compared with mice not treated with streptozotocin. RESULTS Under our experimental conditions, we observed that mice treated with GH had larger islets and higher serum insulin levels than streptozotocin-treated mice treated with saline (0.288 vs. 0.073 ng/mL, p < 0.01). CONCLUSIONS Our data demonstrate that GH may rescue islets and therefore may possess therapeutic potential in the treatment of type 1 diabetes, although consideration should be made regarding GH's effect on insulin resistance.
Collapse
Affiliation(s)
- Eyal J Scheinman
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Rawan Damouni
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Avishay Caspi
- Diabetes and Metabolism Clinical Research Center of Excellence, Clinical Research Institute at Rambam (CRIR), Rambam Health Care Campus, Haifa, Israel
| | - Zila Shen-Orr
- Diabetes and Metabolism Clinical Research Center of Excellence, Clinical Research Institute at Rambam (CRIR), Rambam Health Care Campus, Haifa, Israel
| | - Dov Tiosano
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
- Pediatric Endocrinology Unit, Meyer Children's Hospital, Rambam Health Care Campus, Haifa, Israel
| | - Derek LeRoith
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
- Diabetes and Metabolism Clinical Research Center of Excellence, Clinical Research Institute at Rambam (CRIR), Rambam Health Care Campus, Haifa, Israel
| |
Collapse
|
26
|
Maffei A, Segal AM, Alvarez-Perez JC, Garcia-Ocaña A, Harris PE. Anti-incretin, Anti-proliferative Action of Dopamine on β-Cells. Mol Endocrinol 2015; 29:542-57. [PMID: 25751312 DOI: 10.1210/me.2014-1273] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Human islet β-cells exploit an autocrine dopamine (DA)-mediated inhibitory circuit to regulate insulin secretion. β-Cells also express the DA active transporter and the large neutral amino acid transporter heterodimer enabling them to import circulating DA or its biosynthetic precursor, L-3,4-dihydroxyphenylalanine (L-DOPA). The capacity to import DA or L-DOPA from the extracellular space possibly indicates that DA may be an endocrine signal as well. In humans, a mixed meal stimulus is accompanied by contemporary serum excursions of incretins, DA and L-DOPA, suggesting that DA may act as an anti-incretin as postulated by the foregut hypothesis proposed to explain the early effects of bariatric surgery on type 2 diabetes. In this report, we take a translational step backwards and characterize the kinetics of plasma DA and incretin production after a mixed meal challenge in a rat model and study the integration of incretin and DA signaling at the biochemical level in a rodent β-cell line and islets. We found that there are similar excursions of incretins and DA in rats, as those reported in humans, after a mixed meal challenge and that DA counters incretin enhanced glucose-stimulated insulin secretion and intracellular signaling at multiple points from dampening calcium fluxes to inhibiting proliferation as well as apoptosis. Our data suggest that DA is an important regulator of insulin secretion and may represent 1 axis of a gut level circuit of glucose and β-cell mass homeostasis.
Collapse
Affiliation(s)
- Antonella Maffei
- Division of Endocrinology (A.M., P.H.), Department of Medicine, and Department of Surgery (A.M.S.), Columbia University Medical College, New York, New York 10032; Institute of Genetics and Biophysics (A.M.), Adriano Buzzati-Traverso, Consiglio Nazionale delle Ricerche, 80131 Naples, Italy; and Division of Endocrinology, Diabetes and Bone Diseases (J.C.A.-P., A.G.-O.), Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai and The Mindich Child Health and Development Institute, New York, New York 10029
| | | | | | | | | |
Collapse
|
27
|
Lee KL, Middleditch MJ, Williams GM, Brimble MA, Cooper GJS. Using mass spectrometry to detect, differentiate, and semiquantitate closely related peptide hormones in complex milieu: measurement of IGF-II and vesiculin. Endocrinology 2015; 156:1194-9. [PMID: 25545381 DOI: 10.1210/en.2014-1593] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The search for an islet β-cell growth factor has been a key objective in recent diabetes research, because the ability to regenerate and/or protect the functioning β-cell population in patients could result in a great advancement for diabetes treatment. IGF-I and IGF-II are known to play crucial roles in fetal growth and prenatal development, and there is growing evidence that IGF-II increases β-cell proliferation and survival in vitro and in vivo. A search for the source of IGF-II-like immunoreactivity in isolated β-cell secretory granules from the murine cell line βTC6-F7 revealed a novel 2-chain IGF-II-derived peptide, which we named vesiculin and which has been shown to be a full insulin agonist. Here, we present a liquid chromatography-tandem mass spectrometry method that enables selective detection and semiquantitation of the highly related IGF-II and vesiculin molecules. We have used this method to measure these 2 peptides in conditioned media from 2 β-cell lines, produced under increasing glucose concentrations. This technique detected both IGF-II and vesiculin in media conditioned by MIN6 and βTC6-F7 cells at levels in the range of 0 to 6 μM (total insulin, 80-450 μM) and revealed a glucose-stimulated increase in insulin, IGF-II, and vesiculin. IGF-II was detected in adult human and neonatal mouse serum in high levels, but vesiculin was not present. The methodology we present herein has utility for detecting and differentiating active peptides that are highly related and of low abundance.
Collapse
Affiliation(s)
- Kate L Lee
- School of Biological Sciences (K.L.L., M.J.M., G.J.S.C.), School of Chemical Sciences (G.M.W., M.A.B.), and Maurice Wilkins Centre for Molecular BioDiscovery (M.J.M., M.A.B., G.J.S.C.), The University of Auckland, Auckland 1010, New Zealand; and Centre for Advanced Discovery and Experimental Therapeutics (G.J.S.C.), Manchester Biomedical Research Centre, Central Manchester University Hospitals NHS Foundation Trust, and the School of Biomedicine, the Medical School, University of Manchester, Manchester M13 9WL, United Kingdom
| | | | | | | | | |
Collapse
|
28
|
Jamaluddin JL, Huri HZ, Vethakkan SR, Mustafa N. Pancreatic gene variants potentially associated with dipeptidyl peptidase-4 inhibitor treatment response in Type 2 diabetes. Pharmacogenomics 2015; 15:235-49. [PMID: 24444412 DOI: 10.2217/pgs.13.234] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
In the adult pancreas, the expression of the genes PAX4, KCNQ1, TCF7L2, KCNJ11, ABCC8, MTNR1B and WFS1 are mainly restricted to β cells to maintain glucose homeostasis. We have identified these genes as the main regulators of incretin-mediated actions, and therefore they may potentially influence the response of DPP-4 inhibitors. This review represents the first detailed exploration of pancreatic β-cell genes and their variant mechanisms, which could potentially affect the response of DPP-4 inhibitors in Type 2 diabetes. We have focused on the signaling pathways of these genes to understand their roles in gastrointestinal incretin-mediated effects; and finally, we sought to associate gene mechanisms with their Type 2 diabetes risk variants to predict the responses of DPP-4 inhibitors for this disease.
Collapse
Affiliation(s)
- Jazlina Liza Jamaluddin
- Department of Pharmacy, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | | | | | | |
Collapse
|
29
|
Bruun C, Christensen GL, Jacobsen MLB, Kanstrup MB, Jensen PR, Fjordvang H, Mandrup-Poulsen T, Billestrup N. Inhibition of beta cell growth and function by bone morphogenetic proteins. Diabetologia 2014; 57:2546-54. [PMID: 25260823 DOI: 10.1007/s00125-014-3384-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 09/02/2014] [Indexed: 11/25/2022]
Abstract
AIMS/HYPOTHESIS Impairment of beta cell mass and function is evident in both type 1 and type 2 diabetes. In healthy physiological conditions pancreatic beta cells adapt to the body's increasing insulin requirements by proliferation and improved function. We hypothesised that during the development of diabetes, there is an increase in the expression of inhibitory factors that prevent the beta cells from adapting to the increased need for insulin. We evaluated the effects of bone morphogenetic protein (BMP) 2 and -4 on beta cells. METHODS The effects of BMP2 and -4 on beta cell proliferation, apoptosis, gene expression and insulin release were studied in isolated islets of Langerhans from rats, mice and humans. The expression of BMPs was analysed by immunocytochemistry and real-time PCR. The role of endogenous BMP was investigated using a soluble and neutralising form of the BMP receptor 1A. RESULTS BMP2 and -4 were found to inhibit basal as well as growth factor-stimulated proliferation of primary beta cells from rats and mice. Bmp2 and Bmp4 mRNA and protein were expressed in islets and regulated by inflammatory cytokines. Neutralisation of endogenous BMP activity resulted in enhanced proliferation of rodent beta cells. The expression of Id mRNAs was induced by BMP4 in rat and human islets. Finally, glucose-induced insulin secretion was significantly impaired in rodent and human islets pre-treated with BMP4, and inhibition of BMP activity resulted in enhanced insulin release. CONCLUSIONS/INTERPRETATION These data show that BMP2 and -4 exert inhibitory actions on beta cells in vitro and suggest that BMPs exert regulatory roles of beta cell growth and function.
Collapse
|
30
|
Nalla A, Ringholm L, Søstrup B, Højrup P, Thim L, Levery SB, Vakhrushev SY, Billestrup N, Mathiesen ER, Damm P, Nielsen JH. Implications for the offspring of circulating factors involved in beta cell adaptation in pregnancy. Acta Obstet Gynecol Scand 2014; 93:1181-9. [DOI: 10.1111/aogs.12505] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Accepted: 09/10/2014] [Indexed: 01/01/2023]
Affiliation(s)
- Amarnadh Nalla
- Department of Biomedical Sciences; Faculty of Health and Medical Sciences; University of Copenhagen; Copenhagen Denmark
- The Danish Diabetes Academy; Odense University Hospital; Odense Denmark
- Centre for Fetal Programming; Copenhagen Denmark
| | - Lene Ringholm
- Centre for Pregnant Women with Diabetes; Rigshospitalet; Faculty of Health Sciences; University of Copenhagen; Copenhagen Denmark
- Department of Endocrinology; Rigshospitalet; Faculty of Health Sciences; University of Copenhagen; Copenhagen Denmark
| | - Brigitte Søstrup
- Department of Biomedical Sciences; Faculty of Health and Medical Sciences; University of Copenhagen; Copenhagen Denmark
- Centre for Fetal Programming; Copenhagen Denmark
| | - Peter Højrup
- Department of Biochemistry and Molecular Biology; University of Southern Denmark; Odense Denmark
| | - Lars Thim
- Protein Engineering; Novo Nordisk A/S; Bagsvaerd Denmark
| | - Steven B. Levery
- Department of Cellular and Molecular Medicine; Faculty of Health and Medical Sciences; University of Copenhagen; Copenhagen Denmark
| | - Sergey Y. Vakhrushev
- Department of Cellular and Molecular Medicine; Faculty of Health and Medical Sciences; University of Copenhagen; Copenhagen Denmark
| | - Nils Billestrup
- Department of Biomedical Sciences; Faculty of Health and Medical Sciences; University of Copenhagen; Copenhagen Denmark
| | - Elisabeth R. Mathiesen
- Centre for Pregnant Women with Diabetes; Rigshospitalet; Faculty of Health Sciences; University of Copenhagen; Copenhagen Denmark
- Department of Endocrinology; Rigshospitalet; Faculty of Health Sciences; University of Copenhagen; Copenhagen Denmark
| | - Peter Damm
- Centre for Pregnant Women with Diabetes; Rigshospitalet; Faculty of Health Sciences; University of Copenhagen; Copenhagen Denmark
- Department of Obstetrics; Rigshospitalet; Faculty of Health Sciences; University of Copenhagen; Copenhagen Denmark
| | - Jens H. Nielsen
- Department of Biomedical Sciences; Faculty of Health and Medical Sciences; University of Copenhagen; Copenhagen Denmark
- Centre for Fetal Programming; Copenhagen Denmark
| |
Collapse
|
31
|
Rodríguez-Rodero S, Delgado-Álvarez E, Fernández AF, Fernández-Morera JL, Menéndez-Torre E, Fraga MF. Epigenetic alterations in endocrine-related cancer. Endocr Relat Cancer 2014; 21:R319-30. [PMID: 24898948 DOI: 10.1530/erc-13-0070] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Aberrant epigenetics is a hallmark of cancer, and endocrine-related tumors are no exception. Recent research has been identifying an ever-growing number of epigenetic alterations in both genomic DNA methylation and histone post-translational modification in tumors of the endocrine system. Novel microarray and ultra-deep sequencing technologies have allowed the identification of genome-wide epigenetic patterns in some tumor types such as adrenocortical, parathyroid, and breast carcinomas. However, in other cancer types, such as the multiple endocrine neoplasia syndromes and thyroid cancer, tumor information is limited to candidate genes alone. Future research should fill this gap and deepen our understanding of the functional role of these alterations in cancer, as well as defining their possible clinical uses.
Collapse
Affiliation(s)
- Sandra Rodríguez-Rodero
- Endocrinology and Nutrition ServiceHospital Universitario Central de Asturias, Av. Julian Clavería s/n, 33006 Oviedo, SpainCancer Epigenetics LaboratoryInstituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de Oviedo, 33006 Oviedo, SpainDepartment of Immunology and OncologyNational Center for Biotechnology, CNB-CSIC, Cantoblanco, Madrid E-28049, SpainEndocrinology and Nutrition ServiceHospital Universitario Central de Asturias, Av. Julian Clavería s/n, 33006 Oviedo, SpainCancer Epigenetics LaboratoryInstituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de Oviedo, 33006 Oviedo, SpainDepartment of Immunology and OncologyNational Center for Biotechnology, CNB-CSIC, Cantoblanco, Madrid E-28049, Spain
| | - Elías Delgado-Álvarez
- Endocrinology and Nutrition ServiceHospital Universitario Central de Asturias, Av. Julian Clavería s/n, 33006 Oviedo, SpainCancer Epigenetics LaboratoryInstituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de Oviedo, 33006 Oviedo, SpainDepartment of Immunology and OncologyNational Center for Biotechnology, CNB-CSIC, Cantoblanco, Madrid E-28049, Spain
| | - Agustín F Fernández
- Endocrinology and Nutrition ServiceHospital Universitario Central de Asturias, Av. Julian Clavería s/n, 33006 Oviedo, SpainCancer Epigenetics LaboratoryInstituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de Oviedo, 33006 Oviedo, SpainDepartment of Immunology and OncologyNational Center for Biotechnology, CNB-CSIC, Cantoblanco, Madrid E-28049, Spain
| | - Juan L Fernández-Morera
- Endocrinology and Nutrition ServiceHospital Universitario Central de Asturias, Av. Julian Clavería s/n, 33006 Oviedo, SpainCancer Epigenetics LaboratoryInstituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de Oviedo, 33006 Oviedo, SpainDepartment of Immunology and OncologyNational Center for Biotechnology, CNB-CSIC, Cantoblanco, Madrid E-28049, Spain
| | - Edelmiro Menéndez-Torre
- Endocrinology and Nutrition ServiceHospital Universitario Central de Asturias, Av. Julian Clavería s/n, 33006 Oviedo, SpainCancer Epigenetics LaboratoryInstituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de Oviedo, 33006 Oviedo, SpainDepartment of Immunology and OncologyNational Center for Biotechnology, CNB-CSIC, Cantoblanco, Madrid E-28049, Spain
| | - Mario F Fraga
- Endocrinology and Nutrition ServiceHospital Universitario Central de Asturias, Av. Julian Clavería s/n, 33006 Oviedo, SpainCancer Epigenetics LaboratoryInstituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de Oviedo, 33006 Oviedo, SpainDepartment of Immunology and OncologyNational Center for Biotechnology, CNB-CSIC, Cantoblanco, Madrid E-28049, SpainEndocrinology and Nutrition ServiceHospital Universitario Central de Asturias, Av. Julian Clavería s/n, 33006 Oviedo, SpainCancer Epigenetics LaboratoryInstituto Universitario de Oncología del Principado de Asturias (IUOPA), Universidad de Oviedo, 33006 Oviedo, SpainDepartment of Immunology and OncologyNational Center for Biotechnology, CNB-CSIC, Cantoblanco, Madrid E-28049, Spain
| |
Collapse
|
32
|
Abstract
PURPOSE OF REVIEW Betatrophin is a newly described hormone, which potently stimulates beta cell replication in mice. This discovery has engendered great hope that it could prove clinically important in the treatment of type 1 and type 2 diabetes. RECENT FINDINGS Betatrophin, a 198-amino acid protein secreted by liver and adipose tissue, stimulates growth of pancreatic beta cell mass in insulin-resistant mice. Betatrophin has previously been named RIFL, lipasin, and ANGPLT8, and its salutory effects on lipid metabolism have been described in mouse and human studies. Serum betatrophin levels in humans correlate with improved adipose tissue lipid storage and lower serum triglyceride levels in the fed state, but do not correlate with insulin resistance or carbohydrate tolerance in humans. Betatrophin has not yet been shown to have an effect on beta cell replication in human pancreatic islets. SUMMARY Many endocrine and paracrine factors, of which betatrophin is the newest described, increase beta cell mass in murine models. None of these factors, including betatrophin, have displayed the same activity in clinical studies. This may reflect a profound species difference in beta cell regeneration pathways in mice and humans.
Collapse
|
33
|
Mozar A, Kondegowda NG, Pollack I, Fenutria R, Vasavada RC. The Role of PTHrP in Pancreatic Beta-Cells and Implications for Diabetes Pathophysiology and Treatment. Clin Rev Bone Miner Metab 2014. [DOI: 10.1007/s12018-014-9168-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
34
|
Lindahl M, Danilova T, Palm E, Lindholm P, Võikar V, Hakonen E, Ustinov J, Andressoo JO, Harvey BK, Otonkoski T, Rossi J, Saarma M. MANF is indispensable for the proliferation and survival of pancreatic β cells. Cell Rep 2014; 7:366-375. [PMID: 24726366 DOI: 10.1016/j.celrep.2014.03.023] [Citation(s) in RCA: 152] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 02/12/2014] [Accepted: 03/07/2014] [Indexed: 12/30/2022] Open
Abstract
All forms of diabetes mellitus (DM) are characterized by the loss of functional pancreatic β cell mass, leading to insufficient insulin secretion. Thus, identification of novel approaches to protect and restore β cells is essential for the development of DM therapies. Mesencephalic astrocyte-derived neurotrophic factor (MANF) is an endoplasmic reticulum (ER)-stress-inducible protein, but its physiological role in mammals has remained obscure. We generated MANF-deficient mice that strikingly develop severe diabetes due to progressive postnatal reduction of β cell mass, caused by decreased proliferation and increased apoptosis. Additionally, we show that lack of MANF in vivo in mouse leads to chronic unfolded protein response (UPR) activation in pancreatic islets. Importantly, MANF protein enhanced β cell proliferation in vitro and overexpression of MANF in the pancreas of diabetic mice enhanced β cell regeneration. We demonstrate that MANF specifically promotes β cell proliferation and survival, thereby constituting a therapeutic candidate for β cell protection and regeneration.
Collapse
Affiliation(s)
- Maria Lindahl
- Institute of Biotechnology, University of Helsinki, Viikinkaari 9, 00014 Helsinki, Finland.
| | - Tatiana Danilova
- Institute of Biotechnology, University of Helsinki, Viikinkaari 9, 00014 Helsinki, Finland
| | - Erik Palm
- Institute of Biotechnology, University of Helsinki, Viikinkaari 9, 00014 Helsinki, Finland
| | - Päivi Lindholm
- Institute of Biotechnology, University of Helsinki, Viikinkaari 9, 00014 Helsinki, Finland
| | - Vootele Võikar
- Neuroscience Center, University of Helsinki, Viikinkaari 4, 00014 Helsinki, Finland
| | - Elina Hakonen
- Research Program for Molecular Neurology and Biomedicum Stem Cell Center, University of Helsinki, Haartmaninkatu 8, 00014 Helsinki, Finland
| | - Jarkko Ustinov
- Research Program for Molecular Neurology and Biomedicum Stem Cell Center, University of Helsinki, Haartmaninkatu 8, 00014 Helsinki, Finland
| | - Jaan-Olle Andressoo
- Institute of Biotechnology, University of Helsinki, Viikinkaari 9, 00014 Helsinki, Finland
| | - Brandon K Harvey
- Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, 251 Bayview Boulevard, Baltimore, MD 21224, USA
| | - Timo Otonkoski
- Research Program for Molecular Neurology and Biomedicum Stem Cell Center, University of Helsinki, Haartmaninkatu 8, 00014 Helsinki, Finland; Children's Hospital, Helsinki University Central Hospital, Haartmaninkatu 8, 00014 Helsinki, Finland
| | - Jari Rossi
- Institute of Biomedicine, Anatomy, University of Helsinki, Haartmaninkatu 8, 00014 Helsinki, Finland
| | - Mart Saarma
- Institute of Biotechnology, University of Helsinki, Viikinkaari 9, 00014 Helsinki, Finland
| |
Collapse
|
35
|
Migliorini A, Bader E, Lickert H. Islet cell plasticity and regeneration. Mol Metab 2014; 3:268-74. [PMID: 24749056 PMCID: PMC3986629 DOI: 10.1016/j.molmet.2014.01.010] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 01/13/2014] [Accepted: 01/15/2014] [Indexed: 01/09/2023] Open
Abstract
Insulin-dependent diabetes is a complex multifactorial disorder characterized by loss or dysfunction of β-cells resulting in failure of metabolic control. Even though type 1 and 2 diabetes differ in their pathogenesis, restoring β-cell function is the overarching goal for improved therapy of both diseases. This could be achieved either by cell-replacement therapy or by triggering intrinsic regenerative mechanisms of the pancreas. For type 1 diabetes, a combination of β-cell replacement and immunosuppressive therapy could be a curative treatment, whereas for type 2 diabetes enhancing endogenous mechanisms of β-cell regeneration might optimize blood glucose control. This review will briefly summarize recent efforts to allow β-cell regeneration where the most promising approaches are currently (1) increasing β-cell self-replication or neogenesis from ductal progenitors and (2) conversion of α-cells into β-cells.
Collapse
Affiliation(s)
- Adriana Migliorini
- Institute of Stem Cell Research, Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Am Parkring 11, D-85748, Business Campus Garching, Germany
| | - Erik Bader
- Institute of Stem Cell Research, Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Am Parkring 11, D-85748, Business Campus Garching, Germany
- Research Unit of Molecular Epidemiology Institute of Epidemiology II, Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
| | - Heiko Lickert
- Institute of Stem Cell Research, Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764 Neuherberg, Germany
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Am Parkring 11, D-85748, Business Campus Garching, Germany
- German Center for Diabetes Research (DZD), Germany
- Corresponding author at: Helmholtz Zentrum München, Am Parkring 11, D-85748, Business Campus Garching, Germany. Tel.: +49 89 3187 3760; fax: +49 89 3187 2060.
| |
Collapse
|
36
|
Huang Y, Chang Y. Regulation of pancreatic islet beta-cell mass by growth factor and hormone signaling. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2014; 121:321-49. [PMID: 24373242 DOI: 10.1016/b978-0-12-800101-1.00010-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Dysfunction and destruction of pancreatic islet beta cells is a hallmark of diabetes. Better understanding of cellular signals in beta cells will allow development of therapeutic strategies for diabetes, such as preservation and expansion of beta-cell mass and improvement of beta-cell function. During the past several decades, the number of studies analyzing the molecular mechanisms, including growth factor/hormone signaling pathways that impact islet beta-cell mass and function, has increased exponentially. Notably, somatolactogenic hormones including growth hormone (GH), prolactin (PRL), and insulin-like growth factor-1 (IGF-1) and their receptors (GHR, PRLR, and IGF-1R) are critically involved in beta-cell growth, survival, differentiation, and insulin secretion. In this chapter, we focus more narrowly on GH, PRL, and IGF-1 signaling, and GH-IGF-1 cross talk. We also discuss how these signaling aspects contribute to the regulation of beta-cell proliferation and apoptosis. In particular, our novel findings of GH-induced formation of GHR-JAK2-IGF-1R protein complex and synergistic effects of GH and IGF-1 on beta-cell signaling, proliferation, and antiapoptosis lead to a new concept that IGF-1R may serve as a proximal component of GH/GHR signaling.
Collapse
Affiliation(s)
- Yao Huang
- Department of Obstetrics and Gynecology, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| | - Yongchang Chang
- Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| |
Collapse
|
37
|
Boerner BP, George NM, Targy NM, Sarvetnick NE. TGF-β superfamily member Nodal stimulates human β-cell proliferation while maintaining cellular viability. Endocrinology 2013; 154:4099-112. [PMID: 23970788 PMCID: PMC3800770 DOI: 10.1210/en.2013-1197] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
In an effort to expand human islets and enhance allogeneic islet transplant for the treatment of type 1 diabetes, identifying signaling pathways that stimulate human β-cell proliferation is paramount. TGF-β superfamily members, in particular activin-A, are likely involved in islet development and may contribute to β-cell proliferation. Nodal, another TGF-β member, is present in both embryonic and adult rodent islets. Nodal, along with its coreceptor, Cripto, are pro-proliferative factors in certain cell types. Although Nodal stimulates apoptosis of rat insulinoma cells (INS-1), Nodal and Cripto signaling have not been studied in the context of human islets. The current study investigated the effects of Nodal and Cripto on human β-cell proliferation, differentiation, and viability. In the human pancreas and isolated human islets, we observed Nodal mRNA and protein expression, with protein expression observed in β and α-cells. Cripto expression was absent from human islets. Furthermore, in cultured human islets, exogenous Nodal stimulated modest β-cell proliferation and inhibited α-cell proliferation with no effect on cellular viability, apoptosis, or differentiation. Nodal stimulated the phosphorylation of mothers against decapentaplegic (SMAD)-2, with no effect on AKT or MAPK signaling, suggesting phosphorylated SMAD signaling was involved in β-cell proliferation. Cripto had no effect on human islet cell proliferation, differentiation, or viability. In conclusion, Nodal stimulates human β-cell proliferation while maintaining cellular viability. Nodal signaling warrants further exploration to better understand and enhance human β-cell proliferative capacity.
Collapse
Affiliation(s)
- Brian P Boerner
- MD, and Nora E. Sarvetnick, PhD, University of Nebraska Medical Center, 985965 Nebraska Medical Center, Omaha, Nebraska 68198-5965. ; or
| | | | | | | |
Collapse
|
38
|
Abstract
The pituitary lactogenic hormone prolactin (PRL) exerts various physiological actions in humans and rodents via its binding to a membrane receptor. Beside its role in lactation and reproduction, accumulating evidence suggests that PRL has a crucial impact on energy balance by acting on two key players, the pancreas and the adipose tissue. Adipose tissue is now recognized as an endocrine organ and its metabolic activity appears to play an important role in pathophysiology such as obesity and diabetes. White adipocytes store excess of energy in the form of triglycerides for future need while brown adipocytes metabolize lipids and glucose to produce heat, highlighting their different metabolic functionality. The plasticity of white adipose tissue, by the emergence of beige adipocytes, appears to be essential in energy homeostasis. PRL receptor deficient mice provided direct evidence that PRL signaling is involved in the regulation of adipogenesis affecting energy balance and metabolic adaptation most notably during development. Moreover, it was demonstrated that PRL signaling participates to brown adipose tissue differentiation and function, opening novel understanding of hormonal regulation of energy balance. This review summarizes our current knowledge about PRL signaling and its role on adipose tissue.
Collapse
Affiliation(s)
- Nadège Carré
- Institut National de la Santé et de la Recherche Médicale (INSERM) U693, Le Kremlin-Bicêtre, France; Université Paris-Sud, Faculté de Médecine Paris-Sud, Unité Mixte de Recherche (UMR)-S693, Le Kremlin-Bicêtre, France.
| | - Nadine Binart
- Institut National de la Santé et de la Recherche Médicale (INSERM) U693, Le Kremlin-Bicêtre, France; Université Paris-Sud, Faculté de Médecine Paris-Sud, Unité Mixte de Recherche (UMR)-S693, Le Kremlin-Bicêtre, France
| |
Collapse
|
39
|
Stein J, Milewski WM, Dey A. The negative cell cycle regulators, p27(Kip1), p18(Ink4c), and GSK-3, play critical role in maintaining quiescence of adult human pancreatic β-cells and restrict their ability to proliferate. Islets 2013; 5:156-69. [PMID: 23896637 PMCID: PMC4049839 DOI: 10.4161/isl.25605] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Adult human pancreatic β-cells are primarily quiescent (G0) yet the mechanisms controlling their quiescence are poorly understood. Here, we demonstrate, by immunofluorescence and confocal microscopy, abundant levels of the critical negative cell cycle regulators, p27(Kip1) and p18(Ink4c), 2 key members of cyclin-dependent kinase (CDK) inhibitor family, and glycogen synthase kinase-3 (GSK-3), a serine-threonine protein kinase, in islet β-cells of adult human pancreatic tissue. Our data show that p27(Kip1) localizes primarily in β-cell nuclei, whereas, p18(Ink4c) is mostly present in β-cell cytosol. Additionally, p-p27(S10), a phosphorylated form of p27(Kip1), which was shown to interact with and to sequester cyclinD-CDK4/6 in the cytoplasm, is present in substantial amounts in β-cell cytosol. Our immunofluorescence analysis displays similar distribution pattern of p27(Kip1), p-p27(S10), p18(Ink4c) and GSK-3 in islet β-cells of adult mouse pancreatic tissue. We demonstrate marked interaction of p27(Kip1) with cyclin D3, an abundant D-type cyclin in adult human islets, and vice versa as well as with its cognate kinase partners, CDK4 and CDK6. Likewise, we show marked interaction of p18(Ink4c) with CDK4. The data collectively suggest that inhibition of CDK function by p27(Kip1) and p18(Ink4c) contributes to human β-cell quiescence. Consistent with this, we have found by BrdU incorporation assay that combined treatments of small molecule GSK-3 inhibitor and mitogen/s lead to elevated proliferation of human β-cells, which is caused partly due to p27(Kip1) downregulation. The results altogether suggest that ex vivo expansion of human β-cells is achievable via increased proliferation for β-cell replacement therapy in diabetes.
Collapse
Affiliation(s)
- Jeffrey Stein
- Section of Endocrinology; Diabetes and Metabolism; Department of Medicine; University of Chicago; Chicago, IL USA
| | - Wieslawa M Milewski
- Section of Endocrinology; Diabetes and Metabolism; Department of Medicine; University of Chicago; Chicago, IL USA
| | - Arunangsu Dey
- Section of Endocrinology; Diabetes and Metabolism; Department of Medicine; University of Chicago; Chicago, IL USA
- College of Medicine; Department of Biochemistry and Molecular Genetics; University of Illinois at Chicago; Chicago, IL USA
- Correspondence to: Arunangsu Dey,
| |
Collapse
|
40
|
Abstract
Preservation and regeneration of β cell endocrine function is a long-sought goal in diabetes research. Defective insulin secretion from β cells underlies both type 1 and type 2 diabetes, thus fueling considerable interest in molecules capable of rebuilding β cell secretion capacity. Though early work in rodents suggested that regeneration might be possible, recent studies have revealed that aging powerfully restricts cell cycle entry of β cells, which may limit regeneration capacity. Consequently, aging has emerged as an enigmatic challenge that might limit β cell regeneration therapies. This Review summarizes recent data regarding the role of aging in β cell regeneration and proposes models explaining these phenomena.
Collapse
Affiliation(s)
- Jake A Kushner
- McNair Medical Institute, Pediatric Diabetes and Endocrinology, Baylor College of Medicine, Houston, Texas 77030, USA.
| |
Collapse
|
41
|
López-Acosta JF, Moreno-Amador JL, Jiménez-Palomares M, Díaz-Marrero AR, Cueto M, Perdomo G, Cózar-Castellano I. Epoxypukalide induces proliferation and protects against cytokine-mediated apoptosis in primary cultures of pancreatic β-cells. PLoS One 2013; 8:e52862. [PMID: 23300997 PMCID: PMC3534672 DOI: 10.1371/journal.pone.0052862] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Accepted: 11/23/2012] [Indexed: 01/09/2023] Open
Abstract
There is an urgency to find new treatments for the devastating epidemic of diabetes. Pancreatic β-cells viability and function are impaired in the two most common forms of diabetes, type 1 and type 2. Regeneration of pancreatic β-cells has been proposed as a potential therapy for diabetes. In a preliminary study, we screened a collection of marine products for β-cell proliferation. One unique compound (epoxypukalide) showed capability to induce β-cell replication in the cell line INS1 832/13 and in primary rat cell cultures. Epoxypukalide was used to study β-cell proliferation by [3H]thymidine incorporation and BrdU incorporation followed by BrdU/insulin staining in primary cultures of rat islets. AKT and ERK1/2 signalling pathways were analyzed. Cell cycle activators, cyclin D2 and cyclin E, were detected by western-blot. Apoptosis was studied by TUNEL and cleaved caspase 3. β-cell function was measured by glucose-stimulated insulin secretion. Epoxypukalide induced 2.5-fold increase in β-cell proliferation; this effect was mediated by activation of ERK1/2 signalling pathway and upregulation of the cell cycle activators, cyclin D2 and cyclin E. Interestingly, epoxypukalide showed protection from basal (40% lower versus control) and cytokine-induced apoptosis (80% lower versus control). Finally, epoxypukalide did not impair β-cell function when measured by glucose-stimulated insulin secretion. In conclusion, epoxypukalide induces β-cell proliferation and protects against basal and cytokine-mediated β-cell death in primary cultures of rat islets. These findings may be translated into new treatments for diabetes.
Collapse
Affiliation(s)
- José Francisco López-Acosta
- Instituto de Genética y Biología Molecular (IBGM)-Universidad de Valladolid, Valladolid, Spain
- Unidad de Investigación, Hospital Universitario Puerta del Mar, Cádiz, Spain
| | | | | | - Ana R. Díaz-Marrero
- Instituto de Productos Naturales y Agrobiología del CSIC, La Laguna, Tenerife, Spain
| | - Mercedes Cueto
- Instituto de Productos Naturales y Agrobiología del CSIC, La Laguna, Tenerife, Spain
| | - Germán Perdomo
- Unidad de Investigación, Hospital Universitario Puerta del Mar, Cádiz, Spain
| | - Irene Cózar-Castellano
- Instituto de Genética y Biología Molecular (IBGM)-Universidad de Valladolid, Valladolid, Spain
- Unidad de Investigación, Hospital Universitario Puerta del Mar, Cádiz, Spain
- * E-mail:
| |
Collapse
|
42
|
Orime K, Shirakawa J, Togashi Y, Tajima K, Inoue H, Ito Y, Sato K, Nakamura A, Aoki K, Goshima Y, Terauchi Y. Trefoil factor 2 promotes cell proliferation in pancreatic β-cells through CXCR-4-mediated ERK1/2 phosphorylation. Endocrinology 2013. [PMID: 23183167 DOI: 10.1210/en.2012-1814] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Decreased β-cell mass is a hallmark of type 2 diabetes, and therapeutic approaches to increase the pancreatic β-cell mass have been expected. In recent years, gastrointestinal incretin peptides have been shown to exert a cell-proliferative effect in pancreatic β-cells. Trefoil factor 2 (TFF2), which is predominantly expressed in the surface epithelium of the stomach, plays a role in antiapoptosis, migration, and proliferation. The TFF family is expressed in pancreatic β-cells, whereas the role of TFF2 in pancreatic β-cells has been obscure. In this study, we investigated the mechanism by which TFF2 enhances pancreatic β-cell proliferation. The effects of TFF2 on cell proliferation were evaluated in INS-1 cells, MIN6 cells, and mouse islets using an adenovirus vector containing TFF2 or a recombinant TFF2 peptide. The forced expression of TFF2 led to an increase in bromodeoxyuridine (BrdU) incorporation in both INS-1 cells and islets, without any alteration in insulin secretion. TFF2 significantly increased the mRNA expression of cyclin A2, D1, D2, D3, and E1 in islets. TFF2 peptide increased ERK1/2 phosphorylation and BrdU incorporation in MIN6 cells. A MAPK kinase inhibitor (U0126) abrogated the TFF2 peptide-mediated proliferation of MIN6 cells. A CX-chemokine receptor-4 antagonist also prevented the TFF2 peptide-mediated increase in ERK1/2 phosphorylation and BrdU incorporation in MIN6 cells. These results indicated that TFF2 is involved in β-cell proliferation at least partially via CX-chemokine receptor-4-mediated ERK1/2 phosphorylation, suggesting TFF2 may be a novel target for inducing β-cell proliferation.
Collapse
Affiliation(s)
- Kazuki Orime
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, 3-9 Fuku-ura, Kanazawa-ku, Yokohama 236-0004, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Davis NE, Hamilton D, Fontaine MJ. Harnessing the immunomodulatory and tissue repair properties of mesenchymal stem cells to restore β cell function. Curr Diab Rep 2012; 12:612-22. [PMID: 22869154 PMCID: PMC3767573 DOI: 10.1007/s11892-012-0305-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Islet cell transplantation has therapeutic potential to cure type 1 diabetes (T1D), which is characterized by autoimmune-mediated destruction of insulin-producing β cells. However, current success rates are limited by long-term decline in islet graft function resulting partially from poor revascularization and immune destruction. Mesenchymal stem cells (MSCs) have the potential to enhance islet transplantation and prevent disease progression by a multifaceted approach. MSCs have been shown to be effective at inhibiting inflammatory-mediated immune responses and at promoting tissue regeneration. The immunomodulatory and tissue repairing properties of MSCs may benefit β cell regeneration in the context of T1D. This review will elucidate how MSCs can minimize β cell damage by providing survival signals and simultaneously modulate the immune response by inhibiting activation, and proliferation of several immune cell types. In addition, MSCs can enhance islet graft revascularization, maintaining long-term β cell viability and function.
Collapse
Affiliation(s)
| | - Diana Hamilton
- Department of Pathology Stanford University School of Medicine
| | | |
Collapse
|
44
|
Chen S, Shimoda M, Chen J, Matsumoto S, Grayburn PA. Transient overexpression of cyclin D2/CDK4/GLP1 genes induces proliferation and differentiation of adult pancreatic progenitors and mediates islet regeneration. Cell Cycle 2012; 11:695-705. [PMID: 22373529 DOI: 10.4161/cc.11.4.19120] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The molecular mechanism of β-cell regeneration remains poorly understood. Cyclin D2/CDK4 expresses in normal β cells and maintains adult β-cell growth. We hypothesized that gene therapy with cyclin D2/CDK4/GLP-1 plasmids targeted to the pancreas of STZ-treated rats by ultrasound-targeted microbubble destruction (UTMD) would force cell cycle re-entry of residual G(0)-phase islet cells into G(1)/S phase to regenerate β cells. A single UTMD treatment induced β-cell regeneration with reversal of diabetes for 6 mo without evidence of toxicity. We observed that this β-cell regeneration was not mediated by self-replication of pre-existing β cells. Instead, cyclin D2/CDK4/GLP-1 initiated robust proliferation of adult pancreatic progenitor cells that exist within islets and terminally differentiate to mature islets with β cells and α cells.
Collapse
Affiliation(s)
- Shuyuan Chen
- Baylor Research Institute, Baylor University Medical Center, Dallas, TX, USA
| | | | | | | | | |
Collapse
|
45
|
Sugiyama C, Yamamoto M, Kotani T, Kikkawa F, Murata Y, Hayashi Y. Fertility and pregnancy-associated ß-cell proliferation in mice deficient in proglucagon-derived peptides. PLoS One 2012; 7:e43745. [PMID: 22928026 PMCID: PMC3426535 DOI: 10.1371/journal.pone.0043745] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Accepted: 07/23/2012] [Indexed: 12/15/2022] Open
Abstract
Proglucagon, which is encoded by the glucagon gene (Gcg), is the precursor of several peptide hormones, including glucagon and glucagon-like peptide 1 (GLP-1). Whereas glucagon stimulates hepatic glycogenolysis and gluconeogenesis, GLP-1 stimulates insulin secretion to lower blood glucose and also supports ß-cell proliferation and protection from apoptotic stimuli. Pregnancy is a strong inducer of change in islet function, however the roles of proglucagon-derived peptides in pregnancy are only partially understood. In the present study, we analyzed fertility and pregnancy-associated changes in homozygous glucagon-green fluorescent protein (gfp) knock-in mice (Gcggfp/gfp), which lack all the peptides derived from proglucagon. Female Gcggfp/gfp mice could deliver and raise Gcggfp/gfp pups to weaning and Gcggfp/gfp pups from Gcggfp/gfp dams were viable and fertile. Pregnancy induced ß-cell proliferation in Gcggfp/gfp mice as well as in control mice. However, serum insulin levels in pregnant Gcggfp/gfp females were lower than those in control pregnant females under ad libitum feeding, and blood glucose levels in pregnant Gcggfp/gfp females were higher after gestational day 12. Gcggfp/gfp females showed a decreased pregnancy rate and smaller litter size. The rate of successful breeding was significantly lower in Gcggfp/gfp females and was not improved by experience of breeding. Taken together, proglucagon-derived peptides are not required for pregnancy-associated ß-cell proliferation, however, are required for regulation of blood glucose levels and normal reproductive capacity. Gcggfp/gfp mice may serve as a novel model to analyze the effect of mild hyperglycemia during late gestational periods.
Collapse
Affiliation(s)
- Chisato Sugiyama
- Department of Genetics, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Michiyo Yamamoto
- Department of Genetics, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
| | - Tomomi Kotani
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Fumitaka Kikkawa
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshiharu Murata
- Department of Genetics, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
| | - Yoshitaka Hayashi
- Department of Genetics, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
- * E-mail:
| |
Collapse
|
46
|
Kondegowda NG, Mozar A, Chin C, Otero A, Garcia-Ocaña A, Vasavada RC. Lactogens protect rodent and human beta cells against glucolipotoxicity-induced cell death through Janus kinase-2 (JAK2)/signal transducer and activator of transcription-5 (STAT5) signalling. Diabetologia 2012; 55:1721-32. [PMID: 22382519 DOI: 10.1007/s00125-012-2501-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Accepted: 01/11/2012] [Indexed: 01/09/2023]
Abstract
AIMS/HYPOTHESIS A leading cause of type 2 diabetes is a reduction in functional beta cell mass partly due to increased beta cell death, triggered by stressors such as glucolipotoxicity (GLT). This study evaluates the hypothesis that lactogens can protect beta cells against GLT and examines the mechanism behind the pro-survival effect. METHODS The effect of exogenous treatment or endogenous expression of lactogens on GLT-induced beta cell death was examined in INS-1 cells, and in rodent and human islets. The mechanism behind the pro-survival effect of lactogens was determined using an inhibitor, siRNAs, a dominant negative (DN) mutant, and Cre-lox-mediated gene deletion analysis. RESULTS Lactogens significantly protect INS-1 and primary rodent beta cells against GLT-induced cell death. The pro-survival effect of lactogens in rodent beta cells is mediated through activation of the Janus kinase-2 (JAK2)/signal transducer and activator of transcription-5 (STAT5) signalling pathway. Lactogen-induced increase in the anti-apoptotic B cell lymphoma-extra large (BCLXL) protein is required to mediate its pro-survival effects in both INS-1 cells and primary rodent beta cells. Most importantly, lactogens significantly protect human beta cells against GLT-induced cell death, and their pro-survival effect is also mediated through the JAK2/STAT5 pathway. CONCLUSIONS/INTERPRETATION These studies, together with previous work, clearly demonstrate the pro-survival nature of lactogens and identify the JAK2/STAT5 pathway as an important mediator of this effect in both rodent and human beta cells. Future studies will determine the effectiveness of this peptide in vivo in the pathophysiology of type 2 diabetes.
Collapse
Affiliation(s)
- N Guthalu Kondegowda
- Division of Endocrinology, University of Pittsburgh, 200 Lothrop St, BST-E1157, Pittsburgh, PA 15261, USA
| | | | | | | | | | | |
Collapse
|
47
|
Estil les E, Téllez N, Escoriza J, Montanya E. Increased β-cell replication and β-cell mass regeneration in syngeneically transplanted rat islets overexpressing insulin-like growth factor II. Cell Transplant 2012; 21:2119-29. [PMID: 22507193 DOI: 10.3727/096368912x638955] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Insulin-like growth factor II (IGF2) is a growth-promoting peptide that increases β-cell proliferation and survival. The aim of the study was to determine the effect of IGF2 overexpression on β-cell mass in transplanted islets. Islets infected with adenovirus encoding for IGF2 (Ad-IGF2 group), for luciferase (Ad-Luc control group), or with uninfected islets (control group) were syngeneically transplanted to streptozotocin-diabetic Lewis rats. Eight hundred islets, a minimal mass model to restore normoglycemia, or 500 islets, a clearly insufficient mass, were transplanted. Rats transplanted with 800 Ad-IGF2 islets showed a better metabolic evolution than control groups. As expected, rats transplanted with 500 Ad-IGF2 or control islets maintained similar hyperglycemia throughout the study, ensuring comparable metabolic conditions among both groups. β-Cell replication was higher in Ad-IGF2 group than in control group on days 3 [1.45% (IQR: 0.26) vs. 0.58% (IQR: 0.18), p = 0.006], 10 [1.58% (IQR: 1.40) vs. 0.90% (IQR: 0.61), p = 0.035], and 28 [1.35% (IQR: 0.35) vs. 0.64% (IQR: 0.28), p = 0.004] after transplantation. β-Cell mass was similarly reduced on day 3 after transplantation in Ad-IGF2 and control group [0.36 mg (IQR: 0.26) vs. 0.38 mg (IQR: 0.19)], it increased on day 10, and on day 28 it was higher in Ad-IGF2 than in control group [0.63 mg (IQR: 0.38) vs. 0.42 mg (IQR: 0.31), p = 0.008]. Apoptosis was similarly increased in Ad-IGF2 and control islets after transplantation. No differences in insulin secretion were found between Ad-IGF2 and uninfected control islets. In summary, IGF2 overexpression in transplanted islets increased β-cell replication, induced the regeneration of the transplanted β-cell mass, and had a beneficial effect on the metabolic outcome reducing the β-cell mass needed to achieve normoglycemia.
Collapse
Affiliation(s)
- Elisabet Estil les
- Laboratory of Diabetes and Experimental Endocrinology, Department of Clinical Sciences, IDIBELL-University of Barcelona, Barcelona, Spain
| | | | | | | |
Collapse
|
48
|
Melnik BC. Excessive Leucine-mTORC1-Signalling of Cow Milk-Based Infant Formula: The Missing Link to Understand Early Childhood Obesity. J Obes 2012; 2012:197653. [PMID: 22523661 PMCID: PMC3317169 DOI: 10.1155/2012/197653] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2011] [Accepted: 01/09/2012] [Indexed: 01/22/2023] Open
Abstract
Increased protein supply by feeding cow-milk-based infant formula in comparison to lower protein content of human milk is a well-recognized major risk factor of childhood obesity. However, there is yet no conclusive biochemical concept explaining the mechanisms of formula-induced childhood obesity. It is the intention of this article to provide the biochemical link between leucine-mediated signalling of mammalian milk proteins and adipogenesis as well as early adipogenic programming. Leucine has been identified as the predominant signal transducer of mammalian milk, which stimulates the nutrient-sensitive kinase mammalian target of rapamycin complex 1 (mTORC1). Leucine thus functions as a maternal-neonatal relay for mTORC1-dependent neonatal β-cell proliferation and insulin secretion. The mTORC1 target S6K1 plays a pivotal role in stimulation of mesenchymal stem cells to differentiate into adipocytes and to induce insulin resistance. It is of most critical concern that infant formulas provide higher amounts of leucine in comparison to human milk. Exaggerated leucine-mediated mTORC1-S6K1 signalling induced by infant formulas may thus explain increased adipogenesis and generation of lifelong elevated adipocyte numbers. Attenuation of mTORC1 signalling of infant formula by leucine restriction to physiologic lower levels of human milk offers a great chance for the prevention of childhood obesity and obesity-related metabolic diseases.
Collapse
Affiliation(s)
- Bodo C. Melnik
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, Sedanstrasse 115, 49090 Osnabrück, Germany
| |
Collapse
|
49
|
Melnik BC. Leucine signaling in the pathogenesis of type 2 diabetes and obesity. World J Diabetes 2012; 3:38-53. [PMID: 22442749 PMCID: PMC3310004 DOI: 10.4239/wjd.v3.i3.38] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2011] [Revised: 02/29/2012] [Accepted: 03/09/2012] [Indexed: 02/06/2023] Open
Abstract
Epidemiological evidence points to increased dairy and meat consumption, staples of the Western diet, as major risk factors for the development of type 2 diabetes (T2D). This paper presents a new concept and comprehensive review of leucine-mediated cell signaling explaining the pathogenesis of T2D and obesity by leucine-induced over-stimulation of mammalian target of rapamycin complex 1 (mTORC1). mTORC1, a pivotal nutrient-sensitive kinase, promotes growth and cell proliferation in response to glucose, energy, growth factors and amino acids. Dairy proteins and meat stimulate insulin/insulin-like growth factor 1 signaling and provide high amounts of leucine, a primary and independent stimulator for mTORC1 activation. The downstream target of mTORC1, the kinase S6K1, induces insulin resistance by phosphorylation of insulin receptor substrate-1, thereby increasing the metabolic burden of β-cells. Moreover, leucine-mediated mTORC1-S6K1-signaling plays an important role in adipogenesis, thus increasing the risk of obesity-mediated insulin resistance. High consumption of leucine-rich proteins explains exaggerated mTORC1-dependent insulin secretion, increased β-cell growth and β-cell proliferation promoting an early onset of replicative β-cell senescence with subsequent β-cell apoptosis. Disturbances of β-cell mass regulation with increased β-cell proliferation and apoptosis as well as insulin resistance are hallmarks of T2D, which are all associated with hyperactivation of mTORC1. In contrast, the anti-diabetic drug metformin antagonizes leucine-mediated mTORC1 signaling. Plant-derived polyphenols and flavonoids are identified as natural inhibitors of mTORC1 and exert anti-diabetic and anti-obesity effects. Furthermore, bariatric surgery in obesity reduces increased plasma levels of leucine and other branched-chain amino acids. Attenuation of leucine-mediated mTORC1 signaling by defining appropriate upper limits of the daily intake of leucine-rich animal and dairy proteins may offer a great chance for the prevention of T2D and obesity, as well as other epidemic diseases of civilization with increased mTORC1 signaling, especially cancer and neurodegenerative diseases, which are frequently associated with T2D.
Collapse
Affiliation(s)
- Bodo C Melnik
- Bodo C Melnik, Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, D-49090 Osnabrück, Germany
| |
Collapse
|
50
|
Adenosine kinase inhibition selectively promotes rodent and porcine islet β-cell replication. Proc Natl Acad Sci U S A 2012; 109:3915-20. [PMID: 22345561 DOI: 10.1073/pnas.1201149109] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Diabetes is a pathological condition characterized by relative insulin deficiency, persistent hyperglycemia, and, consequently, diffuse micro- and macrovascular disease. One therapeutic strategy is to amplify insulin-secretion capacity by increasing the number of the insulin-producing β cells without triggering a generalized proliferative response. Here, we present the development of a small-molecule screening platform for the identification of molecules that increase β-cell replication. Using this platform, we identify a class of compounds [adenosine kinase inhibitors (ADK-Is)] that promote replication of primary β cells in three species (mouse, rat, and pig). Furthermore, the replication effect of ADK-Is is cell type-selective: treatment of islet cell cultures with ADK-Is increases replication of β cells but not that of α cells, PP cells, or fibroblasts. Short-term in vivo treatment with an ADK-I also increases β-cell replication but not exocrine cell or hepatocyte replication. Therefore, we propose ADK inhibition as a strategy for the treatment of diabetes.
Collapse
|