1
|
Martino M, Galderisi A, Evans-Molina C, Dayan C. Revisiting the Pattern of Loss of β-Cell Function in Preclinical Type 1 Diabetes. Diabetes 2024; 73:1769-1779. [PMID: 39106185 DOI: 10.2337/db24-0163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 07/25/2024] [Indexed: 08/09/2024]
Abstract
Type 1 diabetes (T1D) results from β-cell destruction due to autoimmunity. It has been proposed that β-cell loss is relatively quiescent in the early years after seroconversion to islet antibody positivity (stage 1), with accelerated β-cell loss only developing around 6-18 months prior to clinical diagnosis. This construct implies that immunointervention in this early stage will be of little benefit, since there is little disease activity to modulate. Here, we argue that the apparent lack of progression in early-stage disease may be an artifact of the modality of assessment used. When substantial β-cell function remains, the standard assessment, the oral glucose tolerance test, represents a submaximal stimulus and underestimates the residual function. In contrast, around the time of diagnosis, glucotoxicity exerts a deleterious effect on insulin secretion, giving the impression of disease acceleration. Once glucotoxicity is relieved by insulin therapy, β-cell function partially recovers (the honeymoon effect). However, evidence from recent trials suggests that glucose control has little effect on the underlying disease process. We therefore hypothesize that the autoimmune destruction of β-cells actually progresses at a more or less constant rate through all phases of T1D and that early-stage immunointervention will be both beneficial and desirable. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Mariangela Martino
- Diabetes Research Group, Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, U.K
- PhD Program in Immunology, Molecular Medicine, and Applied Biotechnologies, University of Rome "Tor Vergata," Rome, Italy
| | | | - Carmella Evans-Molina
- Indiana University School of Medicine, Indianapolis, IN
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN
- Department of Pediatrics and the Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN
- Richard L. Roudebush VA Medical Center, Indianapolis, IN
| | - Colin Dayan
- Diabetes Research Group, Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, U.K
| |
Collapse
|
2
|
Martin-Vega A, Cobb MH. Navigating the ERK1/2 MAPK Cascade. Biomolecules 2023; 13:1555. [PMID: 37892237 PMCID: PMC10605237 DOI: 10.3390/biom13101555] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 10/16/2023] [Accepted: 10/18/2023] [Indexed: 10/29/2023] Open
Abstract
The RAS-ERK pathway is a fundamental signaling cascade crucial for many biological processes including proliferation, cell cycle control, growth, and survival; common across all cell types. Notably, ERK1/2 are implicated in specific processes in a context-dependent manner as in stem cells and pancreatic β-cells. Alterations in the different components of this cascade result in dysregulation of the effector kinases ERK1/2 which communicate with hundreds of substrates. Aberrant activation of the pathway contributes to a range of disorders, including cancer. This review provides an overview of the structure, activation, regulation, and mutational frequency of the different tiers of the cascade; with a particular focus on ERK1/2. We highlight the importance of scaffold proteins that contribute to kinase localization and coordinate interaction dynamics of the kinases with substrates, activators, and inhibitors. Additionally, we explore innovative therapeutic approaches emphasizing promising avenues in this field.
Collapse
Affiliation(s)
- Ana Martin-Vega
- Department of Pharmacology, UT Southwestern Medical Center, 6001 Forest Park Rd., Dallas, TX 75390, USA;
| | - Melanie H. Cobb
- Department of Pharmacology, UT Southwestern Medical Center, 6001 Forest Park Rd., Dallas, TX 75390, USA;
- Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, 6001 Forest Park Rd., Dallas, TX 75390, USA
| |
Collapse
|
3
|
Cao R, Tian H, Zhang Y, Liu G, Xu H, Rao G, Tian Y, Fu X. Signaling pathways and intervention for therapy of type 2 diabetes mellitus. MedComm (Beijing) 2023; 4:e283. [PMID: 37303813 PMCID: PMC10248034 DOI: 10.1002/mco2.283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 04/18/2023] [Accepted: 04/27/2023] [Indexed: 06/13/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) represents one of the fastest growing epidemic metabolic disorders worldwide and is a strong contributor for a broad range of comorbidities, including vascular, visual, neurological, kidney, and liver diseases. Moreover, recent data suggest a mutual interplay between T2DM and Corona Virus Disease 2019 (COVID-19). T2DM is characterized by insulin resistance (IR) and pancreatic β cell dysfunction. Pioneering discoveries throughout the past few decades have established notable links between signaling pathways and T2DM pathogenesis and therapy. Importantly, a number of signaling pathways substantially control the advancement of core pathological changes in T2DM, including IR and β cell dysfunction, as well as additional pathogenic disturbances. Accordingly, an improved understanding of these signaling pathways sheds light on tractable targets and strategies for developing and repurposing critical therapies to treat T2DM and its complications. In this review, we provide a brief overview of the history of T2DM and signaling pathways, and offer a systematic update on the role and mechanism of key signaling pathways underlying the onset, development, and progression of T2DM. In this content, we also summarize current therapeutic drugs/agents associated with signaling pathways for the treatment of T2DM and its complications, and discuss some implications and directions to the future of this field.
Collapse
Affiliation(s)
- Rong Cao
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduSichuanChina
| | - Huimin Tian
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China Medical School, West China HospitalSichuan UniversityChengduSichuanChina
| | - Yu Zhang
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China Medical School, West China HospitalSichuan UniversityChengduSichuanChina
| | - Geng Liu
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduSichuanChina
| | - Haixia Xu
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduSichuanChina
| | - Guocheng Rao
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China Medical School, West China HospitalSichuan UniversityChengduSichuanChina
| | - Yan Tian
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduSichuanChina
| | - Xianghui Fu
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan University and Collaborative Innovation Center of BiotherapyChengduSichuanChina
- Department of Endocrinology and MetabolismState Key Laboratory of Biotherapy and Cancer CenterWest China Medical School, West China HospitalSichuan UniversityChengduSichuanChina
| |
Collapse
|
4
|
Yang Y, Fan X, Liu Y, Ye D, Liu C, Yang H, Su Z, Zhang Y, Liu Y. Function and Inhibition of DYRK1A: emerging roles of treating multiple human diseases. Biochem Pharmacol 2023; 212:115521. [PMID: 36990324 DOI: 10.1016/j.bcp.2023.115521] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 03/19/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023]
Abstract
Dual-specificity tyrosine phosphorylation-regulated kinase 1A (DYRK1A) is an evolutionarily conserved protein kinase and the most studied member of the Dual-specificity tyrosine-regulated kinase (DYRK) family. It has been shown that it participates in the development of plenty of diseases, and both the low or high expression of DYRK1A protein could lead to disorder. Thus, DYRK1A is recognized as a key target for the therapy for these diseases, and the studies on natural or synthetic DYRK1A inhibitors have become more and more popular. Here, we provide a comprehensive review for DYRK1A from the structure and function of DYRK1A, the roles of DYRK1A in various types of diseases, including diabetes mellitus, neurodegenerative diseases, and kinds of cancers, and the studies of its natural and synthetic inhibitors.
Collapse
|
5
|
Al-Romaiyan A, Masocha W, Oyedemi S, Marafie SK, Huang GC, Jones PM, Persaud SJ. Commiphora myrrha stimulates insulin secretion from β-cells through activation of atypical protein kinase C and mitogen-activated protein kinase. JOURNAL OF ETHNOPHARMACOLOGY 2023; 302:115937. [PMID: 36410575 DOI: 10.1016/j.jep.2022.115937] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 08/22/2022] [Accepted: 11/11/2022] [Indexed: 06/16/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ayurvedic medicine has been used in the treatment of diabetes mellitus for centuries. In Arabia and some areas of Africa, Commiphora myrrha (CM) has been extensively used as a plant-based remedy. We have previously shown that an aqueous CM resin solution directly stimulates insulin secretion from MIN6 cells, a mouse β-cell line, and isolated mouse and human islets. However, the signaling pathways involved in CM-induced insulin secretion are completely unknown. Insulin secretion is normally triggered by elevations in intracellular Ca2+ ([Ca2+]i) through voltage gated Ca2+ channels (VGCC) and activation of protein kinases. Protein and lipid kinases such as protein kinase A (PKA), Ca2+-calmodulin dependent protein kinase II (CaMKII), phosphoinositide 3-kinases (PI3Ks), protein kinase C (PKC) and mitogen-activated protein kinase (MAPK), specifically extracellular signal-regulated kinases (ERK1/2), may be involved in receptor-operated insulin secretion. Therefore, we hypothesized that CM may induce insulin secretion by modulating the activity of VGCC and/or one or more of the above kinases. AIM OF THE STUDY To investigate the possible molecular mechanism of action of CM-induced insulin secretion. The effects of aqueous CM resin extract on [Ca2+]i and protein kinase activation from β-cells were examined. METHODS The effect of aqueous CM resin solution on [Ca2+]i was assessed using Ca2+ microfluorimetry. The involvement of VGCC in CM-induced insulin secretion was investigated using static and perifusion insulin secretion experiments in the presence of either EGTA, a Ca2+ chelator, or nifedipine, a blocker of VGCC. The involvement of kinase activation in the stimulatory effect of CM on insulin secretion was examined by using static and perifusion insulin secretion experiments in the presence of known pharmacological inhibitors and/or downregulation of specific kinases. The effects of CM on phosphorylation of PKCζ and ERK1/2 were also assessed using the Wes™ capillary-based protein electrophoresis. RESULTS Ca2+ microfluorimetry measurements showed that exposing MIN6 cells to CM (0.5-2 mg/mL) was not associated with changes in [Ca2+]i. Similarly, incubating MIN6 cells and mouse islets with EGTA and nifedipine, respectively, did not attenuate the insulin secretion induced by CM. However, incubating mouse and human islets with CM in the presence of staurosporine, a non-selective protein kinase inhibitor, completely blocked the effect of CM on insulin secretion. Exposing mouse islets to CM in the presence of H89, KN62 and LY294002, inhibitors of PKA, CaMKII and PI3K, respectively, did not reduce CM-induced insulin secretion. However, incubating mouse and human islets with CM in the presence of Ro 31-8220, a pan-PKC inhibitor, diminished insulin secretion stimulated by CM, whereas inhibiting the action of typical PKC (with Go6976) and PLCβ (with U73122) did not affect CM-stimulated insulin secretion. Similarly, downregulating typical and novel PKC by chronic exposure of mouse islets to phorbol 12-myristate 13-acetate (PMA) was also not associated with a decrease in the stimulatory effect of CM on insulin secretion. Interestingly, CM-induced insulin secretion from mouse islets was inhibited in the presence of the PKCζ inhibitor ZIP and a MAPK inhibitor PD 98059. In addition, Wes™ capillary-based protein electrophoresis indicated that expression of the phosphorylated forms of PKCζ and ERK1/2, a MAPK, was significantly increased following exposure of INS-1832/13 cells, a rat insulinoma cell line, to CM. CONCLUSIONS Our data indicate that CM directly stimulates insulin secretion through activating known downstream effectors of insulin-stimulus secretion coupling. Indeed, the increase in insulin secretion seen with CM is independent of changes in [Ca2+]i and does not involve activation of VGCC. Instead, the CM stimulatory effect on insulin secretion is completely dependent on protein kinase activation. Our findings indicate that CM could induce insulin exocytosis by stimulating the phosphorylation and activation of PKCζ, which in turn phosphorylates and activates ERK1/2.
Collapse
Affiliation(s)
- Altaf Al-Romaiyan
- Department of Pharmacology & Therapeutics, Faculty of Pharmacy, Kuwait University, Kuwait.
| | - Willias Masocha
- Department of Pharmacology & Therapeutics, Faculty of Pharmacy, Kuwait University, Kuwait.
| | - Sunday Oyedemi
- School of Science and Technology, Nottingham Trent University, Clifton Lane, Nottingham, NG11 8NS, UK.
| | - Sulaiman K Marafie
- Biochemistry and Molecular Biology Department, Dasman Diabetes Institute, Kuwait.
| | - Guo-Cai Huang
- Department of Diabetes, School of Cardiovascular Medicine &Sciences, Faculty of Life Sciences and Medicine, King's College London, UK.
| | - Peter M Jones
- Department of Diabetes, School of Cardiovascular Medicine &Sciences, Faculty of Life Sciences and Medicine, King's College London, UK.
| | - Shanta J Persaud
- Department of Diabetes, School of Cardiovascular Medicine &Sciences, Faculty of Life Sciences and Medicine, King's College London, UK.
| |
Collapse
|
6
|
Darden CM, Vasu S, Mattke J, Liu Y, Rhodes CJ, Naziruddin B, Lawrence MC. Calcineurin/NFATc2 and PI3K/AKT signaling maintains β-cell identity and function during metabolic and inflammatory stress. iScience 2022; 25:104125. [PMID: 35402865 PMCID: PMC8983383 DOI: 10.1016/j.isci.2022.104125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 12/02/2021] [Accepted: 03/16/2022] [Indexed: 11/19/2022] Open
Abstract
Pancreatic islets respond to metabolic and inflammatory stress by producing hormones and other factors that induce adaptive cellular and systemic responses. Here we show that intracellular Ca2+ ([Ca2+]i) and ROS signals generated by high glucose and cytokine-induced ER stress activate calcineurin (CN)/NFATc2 and PI3K/AKT to maintain β-cell identity and function. This was attributed in part by direct induction of the endocrine differentiation gene RFX6 and suppression of several β-cell "disallowed" genes, including MCT1. CN/NFATc2 targeted p300 and HDAC1 to RFX6 and MCT1 promoters to induce and suppress gene transcription, respectively. In contrast, prolonged exposure to stress, hyperstimulated [Ca2+]i, or perturbation of CN/NFATc2 resulted in downregulation of RFX6 and induction of MCT1. These findings reveal that CN/NFATc2 and PI3K/AKT maintain β-cell function during acute stress, but β-cells dedifferentiate to a dysfunctional state upon loss or exhaustion of Ca2+/CN/NFATc2 signaling. They further demonstrate the utility of targeting CN/NFATc2 to restore β-cell function.
Collapse
Affiliation(s)
- Carly M. Darden
- Islet Cell Laboratory, Baylor Scott & White Research Institute, Dallas, TX 75204, USA
- Institute of Biomedical Studies, Baylor University, Waco, TX 76706, USA
| | - Srividya Vasu
- Islet Cell Laboratory, Baylor Scott & White Research Institute, Dallas, TX 75204, USA
| | - Jordan Mattke
- Islet Cell Laboratory, Baylor Scott & White Research Institute, Dallas, TX 75204, USA
- Institute of Biomedical Studies, Baylor University, Waco, TX 76706, USA
| | - Yang Liu
- Annette C. and Harold C. Simmons Transplant Institute, Baylor University Medical Center, Dallas, TX 75246, USA
| | - Christopher J. Rhodes
- Kovler Diabetes Center, Department of Medicine, Section of Endocrinology, Diabetes & Metabolism, University of Chicago, Chicago, IL 60637, USA
- Research and Early Development, Cardiovascular, Renal and Metabolic Diseases, BioPharmaceuticals R&D, AstraZeneca Ltd, Gaithersburg, MD 20878, USA
| | - Bashoo Naziruddin
- Annette C. and Harold C. Simmons Transplant Institute, Baylor University Medical Center, Dallas, TX 75246, USA
| | - Michael C. Lawrence
- Islet Cell Laboratory, Baylor Scott & White Research Institute, Dallas, TX 75204, USA
| |
Collapse
|
7
|
TXNIP: A Double-Edged Sword in Disease and Therapeutic Outlook. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7805115. [PMID: 35450411 PMCID: PMC9017576 DOI: 10.1155/2022/7805115] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 02/17/2022] [Accepted: 03/21/2022] [Indexed: 12/15/2022]
Abstract
Thioredoxin-interacting protein (TXNIP) was originally named vitamin D3 upregulated protein-1 (VDUP1) because of its ability to bind to thioredoxin (TRX) and inhibit TRX function and expression. TXNIP is an alpha-arrestin protein that is essential for redox homeostasis in the human body. TXNIP may act as a double-edged sword in the cell. The balance of TXNIP is crucial. A study has shown that TXNIP can travel between diverse intracellular locations and bind to different proteins to play different roles under oxidative stress. The primary function of TXNIP is to induce apoptosis or pyroptosis under oxidative stress. TXNIP also inhibits proliferation and migration in cancer cells, although TXNIP levels decrease, and function diminishes in various cancers. In this review, we summarized the main structure, binding proteins, pathways, and the role of TXNIP in diseases, aiming to explore the double-edged sword role of TXNIP, and expect it to be helpful for future treatment using TXNIP as a therapeutic target.
Collapse
|
8
|
Zhao R, Lu J, Li Q, Xiong F, Zhang Y, Zhu J, Peng G, Yang J. Single-cell heterogeneity analysis and CRISPR screens in MIN6 cell line reveal transcriptional regulators of insulin. Cell Cycle 2021; 20:2053-2065. [PMID: 34494921 DOI: 10.1080/15384101.2021.1969204] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Diabetes mellitus is caused by either insulin resistance or insulin deficiency. The pancreatic β cells are the primary producers of insulin. Large-scale CRISPR screens combined with single-cell RNA sequencing (scRNA-seq) on β cells has identified novel insulin regulators and revealed the presence of a highly complex inner network. Here, we performed pooled CRISPR delivery with single-cell transcriptome analysis on the MIN6 cell line, a pancreatic β-cell line. We have presented the scRNA-seq readout and demonstrated that the MIN6 cell line might develop genetic heterogeneity with increasing passage number based on GO and KEGG pathway analysis. Both computational and biological analyses revealed that the function of MIN6 cell lines could be divided into five clusters, including endocrine cells, basal cells, epithelial cells, and neuroendocrine cells. The fifth cluster was different from the other four clusters due to the differentially expressed insulin transcription and was called the lncRNA-enriched cluster. The experiments also confirmed that uncharacterized lncRNAs GM26917 and Cenpw were associated with insulin transcription. This study provides information that can be used to systematically characterize insulin regulator genes and other genes that control protein folding and vesicle trafficking.
Collapse
Affiliation(s)
- Ruxuan Zhao
- Beijing Key Laboratory of Diabetes Research and Care, Beijing Tongren Hospital, Capital Medical University, Beijing China
| | - Jing Lu
- Beijing Key Laboratory of Diabetes Research and Care, Beijing Tongren Hospital, Capital Medical University, Beijing China
| | - Qi Li
- Beijing Key Laboratory of Diabetes Research and Care, Beijing Tongren Hospital, Capital Medical University, Beijing China
| | - Fengran Xiong
- Beijing Key Laboratory of Diabetes Research and Care, Beijing Tongren Hospital, Capital Medical University, Beijing China
| | - Yingchao Zhang
- Beijing Key Laboratory of Diabetes Research and Care, Beijing Tongren Hospital, Capital Medical University, Beijing China
| | - Juanjuan Zhu
- Beijing Key Laboratory of Diabetes Research and Care, Beijing Tongren Hospital, Capital Medical University, Beijing China
| | - Gongxin Peng
- Center for Bioinformatics, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Jinkui Yang
- Beijing Key Laboratory of Diabetes Research and Care, Beijing Tongren Hospital, Capital Medical University, Beijing China
| |
Collapse
|
9
|
The MEK Inhibitor Trametinib Suppresses Major Histocompatibility Antigen-mismatched Rejection Following Pancreatic Islet Transplantation. Transplant Direct 2020; 6:e591. [PMID: 32851124 PMCID: PMC7423917 DOI: 10.1097/txd.0000000000001045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 06/19/2020] [Accepted: 07/04/2020] [Indexed: 11/26/2022] Open
Abstract
Background. Potential adverse effects, such as functional impairment of islets, render conventional immunosuppressive drugs unsuitable for use in islet transplantation. In addition, as a single therapy, they cannot prolong islet allograft survival. Here, we investigated the utility of the mitogen-activated protein kinase inhibitor trametinib and asked whether it ameliorates acute rejection of transplanted islets without the need for conventional immunosuppressants. Methods. Islets from fully major histocompatibility complex-mismatched BALB/c mice were transplanted into streptozotocin-induced diabetic C57BL/6 mice via the portal vein. These mice received trametinib or vehicle (orally) for 28 days. Isolated islets from BALB/c mice were incubated in vitro with different concentrations of trametinib to determine viability and function. Results. Trametinib (0.1 and 0.3 mg/kg) prolonged graft survival significantly (P = 0.0007 and P = 0.005, respectively) when compared with vehicle. Histologic analyses revealed that cellular infiltration of the graft by lymphocytes was inhibited significantly on day 7 (P < 0.05). In addition, trametinib suppressed functional differentiation of naive CD4+ T cells in recipients. Expression of mRNA encoding inflammatory cytokines interleukin (IL)-2, tumor necrosis factor α, and interferon γ in recipients treated with trametinib was also inhibited (P < 0.001, P < 0.05, and P < 0.01, respectively). Trametinib also increased production of IL-4 and IL-10 (P < 0.05 and P = 0.20, respectively). In vitro, islets incubated with different concentrations of trametinib exhibited no harmful effects with respect to viability and function. Conclusions. Trametinib delayed islet graft rejection by inhibiting functional differentiation of naive CD4+ T cells and regulating inflammatory cytokines. Trametinib might be a promising candidate for maintenance immunosuppressive therapy after allogeneic islet transplantation.
Collapse
|
10
|
Abstract
Glucose-induced (physiological) insulin secretion from the islet β-cell involves interplay between cationic (i.e., changes in intracellular calcium) and metabolic (i.e., generation of hydrophobic and hydrophilic second messengers) events. A large body of evidence affirms support for novel regulation, by G proteins, of specific intracellular signaling events, including actin cytoskeletal remodeling, transport of insulin-containing granules to the plasma membrane for fusion, and secretion of insulin into the circulation. This article highlights the following aspects of GPCR-G protein biology of the islet. First, it overviews our current understanding of the identity of a wide variety of G protein regulators and their modulatory roles in GPCR-G protein-effector coupling, which is requisite for optimal β-cell function under physiological conditions. Second, it describes evidence in support of novel, noncanonical, GPCR-independent mechanisms of activation of G proteins in the islet. Third, it highlights the evidence indicating that abnormalities in G protein function lead to islet β-cell dysregulation and demise under the duress of metabolic stress and diabetes. Fourth, it summarizes observations of potential beneficial effects of GPCR agonists in preventing/halting metabolic defects in the islet β-cell under various pathological conditions (e.g., metabolic stress and inflammation). Lastly, it identifies knowledge gaps and potential avenues for future research in this evolving field of translational islet biology. Published 2020. Compr Physiol 10:453-490, 2020.
Collapse
Affiliation(s)
- Anjaneyulu Kowluru
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Center for Translational Research in Diabetes, Biomedical Research Service, John D. Dingell VA Medical Center, Wayne State University, Detroit, Michigan, USA
| |
Collapse
|
11
|
Fan L, Shan A, Su Y, Cheng Y, Ji H, Yang Q, Lei Y, Liu B, Wang W, Ning G, Cao Y, Jiang X. MiR-221/222 Inhibit Insulin Production of Pancreatic β-Cells in Mice. Endocrinology 2020; 161:5639771. [PMID: 31761936 DOI: 10.1210/endocr/bqz027] [Citation(s) in RCA: 7] [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] [Received: 09/17/2019] [Accepted: 11/23/2019] [Indexed: 11/19/2022]
Abstract
Microribonucleic acids (miRNAs) are essential for the regulation of development, proliferation, and functions of pancreatic β-cells. The conserved miR-221/222 cluster is an important regulator in multiple cellular processes. Here we investigated the functional role of miR-221/222 in the regulation of β-cell proliferation and functions in transgenic mouse models. We generated 2 pancreatic β-cell-specific-miR-221/222 transgenic mouse models on a C57BL/6J background. The glucose metabolic phenotypes, β-cell mass, and β-cell functions were analyzed in the mouse models. Adenovirus-mediated overexpression of miR-221/222 was performed on β-cells and mouse insulinoma 6 (MIN6) cells to explore the effect and mechanisms of miR-221/222 on β-cell proliferation and functions. Luciferase reporter assay, histological analysis, and quantitative polymerase chain reaction (PCR) were carried out to study the direct target genes of miR-221/222 in β-cells. The expression of miR-221/222 was significantly upregulated in β-cells from the high-fat diet (HFD)-fed mice and db/db mice. Overexpression of miR-221/222 impaired the insulin production and secretion of β-cells and resulted in glucose intolerance in vivo. The β-cell mass and proliferation were increased by miR-221/222 expression via Cdkn1b and Cdkn1c. MiR-221/222 repressed insulin transcription activity through targeting Nfatc3 and lead to reduction of insulin in β-cells. Our findings demonstrate that miR-221/222 are important regulators of β-cell proliferation and insulin production. The expression of miR-221/222 in β-cells could regulate glucose metabolism in physiological and pathological processes.
Collapse
Affiliation(s)
- Liwen Fan
- National Clinical Research Centre for Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Key Laboratory for Endocrine Tumors, Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Aijing Shan
- National Clinical Research Centre for Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Key Laboratory for Endocrine Tumors, Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Yutong Su
- National Clinical Research Centre for Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Key Laboratory for Endocrine Tumors, Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Yulong Cheng
- National Clinical Research Centre for Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Key Laboratory for Endocrine Tumors, Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - He Ji
- National Clinical Research Centre for Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Key Laboratory for Endocrine Tumors, Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Qi Yang
- National Clinical Research Centre for Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Key Laboratory for Endocrine Tumors, Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Ying Lei
- National Clinical Research Centre for Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Key Laboratory for Endocrine Tumors, Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Bei Liu
- National Clinical Research Centre for Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Key Laboratory for Endocrine Tumors, Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Weiqing Wang
- National Clinical Research Centre for Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Key Laboratory for Endocrine Tumors, Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Guang Ning
- National Clinical Research Centre for Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Key Laboratory for Endocrine Tumors, Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Yanan Cao
- National Clinical Research Centre for Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Key Laboratory for Endocrine Tumors, Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| | - Xiuli Jiang
- National Clinical Research Centre for Endocrine and Metabolic Diseases, Shanghai Clinical Center for Endocrine and Metabolic Diseases, Shanghai Key Laboratory for Endocrine Tumors, Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine, Shanghai, China
| |
Collapse
|
12
|
Billert M, Kołodziejski PA, Strowski MZ, Nowak KW, Skrzypski M. Phoenixin-14 stimulates proliferation and insulin secretion in insulin producing INS-1E cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1866:118533. [DOI: 10.1016/j.bbamcr.2019.118533] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 08/13/2019] [Accepted: 08/14/2019] [Indexed: 12/25/2022]
|
13
|
Hu L, He F, Luo Y, Luo H, Hai L, Li Y, Zhou Z, Liu F, Dai YS. Reduced Compensatory β-Cell Proliferation in Nfatc3-Deficient Mice Fed on High-Fat Diet. Exp Clin Endocrinol Diabetes 2019; 129:651-660. [PMID: 31546271 DOI: 10.1055/a-1008-9110] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND High-fat-diet induces pancreatic β-cell compensatory proliferation, and impairments in pancreatic β-cell proliferation and function can lead to defects in insulin secretion and diabetes. NFATc3 is important for HFD-induced adipose tissue inflammation. But it is unknown whether NFATc3 is required for β cell compensatory growth in mice fed with HFD. METHODS NFATc3 mRNA and protein expression levels were quantified by RT-qPCR and Western blotting, respectively, in pancreatic islets of WT mice fed on HFD for 12-20 weeks. Adenoviral-mediated overexpression of NFATc3 were conducted in Min6 cells and cultured primary mouse islets. NFATc3-/- mice and WT control mice were fed with HFD and metabolic and functional parameters were measured. RESULTS We observed that the NFATc3 expression level was reduced in the islets of high-fat-diet (HFD)-fed mice. Adenovirus-mediated overexpression of NFATc3 enhanced glucose-stimulated insulin secretion and β-cell gene expression in cultured primary mouse islets. Nfatc3-/- mice initially developed similar glucose tolerance at 2-4 weeks after HFD feeding than HFD-fed WT mice, but Nfatc3-/- mice developed improved glucose tolerance and insulin sensitivity after 8 weeks of HFD feeding compared to Nfatc3+/+fed with HFD. Furthermore, Nfatc3-/- mice on HFD exhibited decreased β-cell mass and reduced expression of genes important for β-cell proliferation and function compared to Nfatc3+/+mice on HFD. CONCLUSIONS The findings suggested that NFATc3 plays a role in maintaining the pancreatic β-cell compensatory growth and gene expression in response to obesity.
Collapse
Affiliation(s)
- Li Hu
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fengli He
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yan Luo
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hairong Luo
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Luo Hai
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yabin Li
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhiguang Zhou
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Feng Liu
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Yan-Shan Dai
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Metabolic Disease, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Bristol-Myers Squibb Company, Princeton, NJ 08540, United States
| |
Collapse
|
14
|
Tan Q, Li Y, Li X, Zhang S. Hyperinsulinemia impairs functions of circulating endothelial progenitor cells. Acta Diabetol 2019; 56:785-795. [PMID: 30859314 DOI: 10.1007/s00592-019-01314-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 02/21/2019] [Indexed: 02/06/2023]
Abstract
AIMS Circulating endothelial progenitor cells (EPCs) play a key role in maintaining endothelial function. Dysfunction of EPCs is associated with the cardiovascular complication of diabetes. The purpose of this study is to investigate the direct effects of hyperinsulinemia on EPCs and the underlying mechanisms. METHODS EPCs isolated from healthy adults were cultured with various concentrations of insulin (control group, without insulin; physiological insulin group, 10 nM insulin and hyperinsulinemia group, 100 nM insulin) with or without phosphatidylinositol-3-kinase (PI3-K) inhibitor (LY294002, 5 µM), endothelial nitric oxide synthase (eNOS) inhibitor (L-NG-nitro-arginine methyl ester (L-NAME), 100 µM), sodium nitroprusside (SNP, 25 µM), p38 mitogen-activated protein kinase(MAPK) inhibitor (SB203580, 5 µM) or extracellular signal-regulated kinases (ERK) 1/2 inhibitor (PD98059, 10 µM). Proliferation, tube formation, and apoptosis of EPCs were determined. Expressions of eNOS, PI3-K, protein kinase B (Akt), p38 MAPK, and ERK 1/2 were assessed. RESULTS Hyperinsulinemia caused a significant decrease in proliferation and tube formation abilities than control group. Hyperinsulinemia increased apoptosis rate of EPCs than control group. Furthermore, hyperinsulinemia downregulated phosphorylation of eNOS, PI3-K and Akt, and upregulated phosphorylation of p38 MAPK and ERK. SNP could restore impaired tube formation induced by hyperinsulinemia. P38 MAPK inhibitor but not ERK inhibitor could decrease apoptosis induced by hyperinsulinemia. CONCLUSION Hyperinsulinemia impaired EPCs' tube formation ability by downregulation of PI-3K/Akt/eNOS pathway. Hyperinsulinemia induced apoptosis of EPCs via upregulation of p38 MAPK.
Collapse
Affiliation(s)
- Qiang Tan
- Department of Cardiology, The First Hospital of Qinhuangdao, Hebei Medical University, Qinhuangdao, Hebei, China.
| | - Yang Li
- Department of Cardiology, The First Hospital of Qinhuangdao, Hebei Medical University, Qinhuangdao, Hebei, China
| | - Xuan Li
- Department of Cardiology, The First Hospital of Qinhuangdao, Hebei Medical University, Qinhuangdao, Hebei, China
| | - Shuangyue Zhang
- Department of Cardiology, The First Hospital of Qinhuangdao, Hebei Medical University, Qinhuangdao, Hebei, China
| |
Collapse
|
15
|
Billert M, Sassek M, Wojciechowicz T, Jasaszwili M, Strowski MZ, Nowak KW, Skrzypski M. Neuropeptide B stimulates insulin secretion and expression but not proliferation in rat insulin‑producing INS‑1E cells. Mol Med Rep 2019; 20:2030-2038. [PMID: 31257494 DOI: 10.3892/mmr.2019.10415] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 05/24/2019] [Indexed: 11/06/2022] Open
Abstract
Neuropeptide B (NPB) regulates food intake, body weight and energy homeostasis by interacting with NPBW1/NPBW2 in humans and NPBW1 in rodents. NPB and NPBW1 are widely expressed in the central nervous system and peripheral tissues including pancreatic islets. Although previous studies have demonstrated a prominent role for NPB and NPBW1 in controlling glucose and energy homeostasis, it remains unknown as to whether NPB modulates pancreatic β‑cell functions. Therefore, the aim of the present study was to investigate the effects of NPB on insulin expression and secretion in vitro. Furthermore, the role of NPB in the modulation of INS‑1E cell growth, viability and death was examined. Gene expression was assessed by reverse transcription‑quantitative PCR. Cell proliferation and viability were determined by BrdU or MTT tests, respectively. Apoptotic cell death was evaluated by relative quantification histone‑complexed DNA fragments (mono‑and oligonucleosomes). Insulin secretion was studied using an ELISA test. Protein phosphorylation was assessed by western blot analysis. NPB and NPBW1 mRNA was expressed in INS‑1E cells and rat pancreatic islets. In INS‑1E cells, NPB enhanced insulin 1 mRNA expression via an ERK1/2‑dependent mechanism. Furthermore, NPB stimulated insulin secretion from INS‑1E cells and rat pancreatic islets. By contrast, NPB failed to affect INS‑1E cell growth or death. We conclude that NPB may regulate insulin secretion and expression in INS‑1E cells and insulin secretion in rat pancreatic islets.
Collapse
Affiliation(s)
- Maria Billert
- Department of Animal Physiology and Biochemistry, Poznań University of Life Sciences, 60‑637 Poznań, Poland
| | - Maciej Sassek
- Department of Animal Physiology and Biochemistry, Poznań University of Life Sciences, 60‑637 Poznań, Poland
| | - Tatiana Wojciechowicz
- Department of Animal Physiology and Biochemistry, Poznań University of Life Sciences, 60‑637 Poznań, Poland
| | - Mariami Jasaszwili
- Department of Animal Physiology and Biochemistry, Poznań University of Life Sciences, 60‑637 Poznań, Poland
| | - Mathias Z Strowski
- Department of Hepatology and Gastroenterology, Charité‑University Medicine Berlin, D‑13353 Berlin, Germany
| | - Krzysztof W Nowak
- Department of Animal Physiology and Biochemistry, Poznań University of Life Sciences, 60‑637 Poznań, Poland
| | - Marek Skrzypski
- Department of Animal Physiology and Biochemistry, Poznań University of Life Sciences, 60‑637 Poznań, Poland
| |
Collapse
|
16
|
Sabatini PV, Speckmann T, Lynn FC. Friend and foe: β-cell Ca 2+ signaling and the development of diabetes. Mol Metab 2019; 21:1-12. [PMID: 30630689 PMCID: PMC6407368 DOI: 10.1016/j.molmet.2018.12.007] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 12/03/2018] [Accepted: 12/19/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The divalent cation Calcium (Ca2+) regulates a wide range of processes in disparate cell types. Within insulin-producing β-cells, increases in cytosolic Ca2+ directly stimulate insulin vesicle exocytosis, but also initiate multiple signaling pathways. Mediated through activation of downstream kinases and transcription factors, Ca2+-regulated signaling pathways leverage substantial influence on a number of critical cellular processes within the β-cell. Additionally, there is evidence that prolonged activation of these same pathways is detrimental to β-cell health and may contribute to Type 2 Diabetes pathogenesis. SCOPE OF REVIEW This review aims to briefly highlight canonical Ca2+ signaling pathways in β-cells and how β-cells regulate the movement of Ca2+ across numerous organelles and microdomains. As a main focus, this review synthesizes experimental data from in vitro and in vivo models on both the beneficial and detrimental effects of Ca2+ signaling pathways for β-cell function and health. MAJOR CONCLUSIONS Acute increases in intracellular Ca2+ stimulate a number of signaling cascades, resulting in (de-)phosphorylation events and activation of downstream transcription factors. The short-term stimulation of these Ca2+ signaling pathways promotes numerous cellular processes critical to β-cell function, including increased viability, replication, and insulin production and secretion. Conversely, chronic stimulation of Ca2+ signaling pathways increases β-cell ER stress and results in the loss of β-cell differentiation status. Together, decades of study demonstrate that Ca2+ movement is tightly regulated within the β-cell, which is at least partially due to its dual roles as a potent signaling molecule.
Collapse
Affiliation(s)
- Paul V Sabatini
- Diabetes Research Group, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada; Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada; Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Thilo Speckmann
- Diabetes Research Group, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada; Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Francis C Lynn
- Diabetes Research Group, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada; Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
17
|
Endothelin-converting enzyme-1 regulates glucagon-like peptide-1 receptor signalling and resensitisation. Biochem J 2019; 476:513-533. [PMID: 30626614 DOI: 10.1042/bcj20180853] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 01/07/2019] [Accepted: 01/08/2019] [Indexed: 02/08/2023]
Abstract
Following nutrient ingestion, glucagon-like peptide 1 (GLP-1) is secreted from intestinal L-cells and mediates anti-diabetic effects, most notably stimulating glucose-dependent insulin release from pancreatic β-cells but also inhibiting glucagon release, promoting satiety and weight reduction and potentially enhancing or preserving β-cell mass. These effects are mediated by the GLP-1 receptor (GLP-1R), which is a therapeutic target in type 2 diabetes. Although agonism at the GLP-1R has been well studied, desensitisation and resensitisation are perhaps less well explored. An understanding of these events is important, particularly in the design and use of novel receptor ligands. Here, using either HEK293 cells expressing the recombinant human GLP-1R or the pancreatic β-cell line, INS-1E with endogenous expressesion of the GLP-1R, we demonstrate GLP-1R desensitisation and subsequent resensitisation following removal of extracellular GLP-1 7-36 amide. Resensitisation is dependent on receptor internalisation, endosomal acidification and receptor recycling. Resensitisation is also regulated by endothelin-converting enzyme-1 (ECE-1) activity, most likely through proteolysis of GLP-1 in endosomes and the facilitation of GLP-1R dephosphorylation and recycling. Inhibition of ECE-1 activity also increases GLP-1-induced activation of extracellular signal-regulated kinase and generation of cAMP, suggesting processes dependent upon the lifetime of the internalised ligand-receptor complex.
Collapse
|
18
|
Abdolazimi Y, Zhao Z, Lee S, Xu H, Allegretti P, Horton TM, Yeh B, Moeller HP, Nichols RJ, McCutcheon D, Shalizi A, Smith M, Armstrong NA, Annes JP. CC-401 Promotes β-Cell Replication via Pleiotropic Consequences of DYRK1A/B Inhibition. Endocrinology 2018; 159:3143-3157. [PMID: 29514186 PMCID: PMC6287593 DOI: 10.1210/en.2018-00083] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 02/27/2018] [Indexed: 12/23/2022]
Abstract
Pharmacologic expansion of endogenous β cells is a promising therapeutic strategy for diabetes. To elucidate the molecular pathways that control β-cell growth we screened ∼2400 bioactive compounds for rat β-cell replication-modulating activity. Numerous hit compounds impaired or promoted rat β-cell replication, including CC-401, an advanced clinical candidate previously characterized as a c-Jun N-terminal kinase inhibitor. Surprisingly, CC-401 induced rodent (in vitro and in vivo) and human (in vitro) β-cell replication via dual-specificity tyrosine phosphorylation-regulated kinase (DYRK) 1A and 1B inhibition. In contrast to rat β cells, which were broadly growth responsive to compound treatment, human β-cell replication was only consistently induced by DYRK1A/B inhibitors. This effect was enhanced by simultaneous glycogen synthase kinase-3β (GSK-3β) or activin A receptor type II-like kinase/transforming growth factor-β (ALK5/TGF-β) inhibition. Prior work emphasized DYRK1A/B inhibition-dependent activation of nuclear factor of activated T cells (NFAT) as the primary mechanism of human β-cell-replication induction. However, inhibition of NFAT activity had limited effect on CC-401-induced β-cell replication. Consequently, we investigated additional effects of CC-401-dependent DYRK1A/B inhibition. Indeed, CC-401 inhibited DYRK1A-dependent phosphorylation/stabilization of the β-cell-replication inhibitor p27Kip1. Additionally, CC-401 increased expression of numerous replication-promoting genes normally suppressed by the dimerization partner, RB-like, E2F and multivulval class B (DREAM) complex, which depends upon DYRK1A/B activity for integrity, including MYBL2 and FOXM1. In summary, we present a compendium of compounds as a valuable resource for manipulating the signaling pathways that control β-cell replication and leverage a DYRK1A/B inhibitor (CC-401) to expand our understanding of the molecular pathways that control β-cell growth.
Collapse
Affiliation(s)
- Yassan Abdolazimi
- Department of Medicine, Division of Endocrinology, Stanford University,
Stanford, California
| | - Zhengshan Zhao
- Biomedical Institute for Regenerative Research, Texas A&M University,
Commerce, Texas
| | - Sooyeon Lee
- Department of Medicine, Division of Endocrinology, Stanford University,
Stanford, California
| | - Haixia Xu
- Department of Medicine, Division of Endocrinology, Stanford University,
Stanford, California
| | - Paul Allegretti
- Department of Medicine, Division of Endocrinology, Stanford University,
Stanford, California
- Chemistry, Engineering and Medicine for Human Health Research Institute,
Stanford University, Stanford, California
| | - Timothy M Horton
- Department of Medicine, Division of Endocrinology, Stanford University,
Stanford, California
- Chemistry, Engineering and Medicine for Human Health Research Institute,
Stanford University, Stanford, California
- Department of Chemistry, Stanford University, Stanford, California
| | - Benjamin Yeh
- Department of Medicine, Division of Endocrinology, Stanford University,
Stanford, California
| | - Hannah P Moeller
- Department of Medicine, Division of Endocrinology, Stanford University,
Stanford, California
| | - Robert J Nichols
- Department of Genetics, Stanford University, Stanford, California
| | - David McCutcheon
- Department of Medicine, Division of Endocrinology, Stanford University,
Stanford, California
- Chemistry, Engineering and Medicine for Human Health Research Institute,
Stanford University, Stanford, California
| | - Aryaman Shalizi
- Department of Pathology, Stanford University, Stanford, California
| | - Mark Smith
- Chemistry, Engineering and Medicine for Human Health Research Institute,
Stanford University, Stanford, California
- Medicinal Chemistry Knowledge Center, Chemistry, Engineering and Medicine for
Human Health, Stanford University, Stanford, California
| | - Neali A Armstrong
- Department of Medicine, Division of Endocrinology, Stanford University,
Stanford, California
| | - Justin P Annes
- Department of Medicine, Division of Endocrinology, Stanford University,
Stanford, California
- Chemistry, Engineering and Medicine for Human Health Research Institute,
Stanford University, Stanford, California
| |
Collapse
|
19
|
Isx9 Regulates Calbindin D28K Expression in Pancreatic β Cells and Promotes β Cell Survival and Function. Int J Mol Sci 2018; 19:ijms19092542. [PMID: 30150605 PMCID: PMC6165483 DOI: 10.3390/ijms19092542] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 08/13/2018] [Accepted: 08/19/2018] [Indexed: 11/21/2022] Open
Abstract
Pancreatic β-cell dysfunction and death contribute to the onset of diabetes, and novel strategies of β-cell function and survival under diabetogenic conditions need to be explored. We previously demonstrated that Isx9, a small molecule based on the isoxazole scaffold, drives neuroendocrine phenotypes by increasing the expression of genes required for β-cell function and improves glycemia in a model of β cell regeneration. We further investigated the role of Isx9 in β-cell survival. We find that Isx9 drives the expression of Calbindin-D28K (D28K), a key regulator of calcium homeostasis, and plays a cytoprotective role through its calcium buffering capacity in β cells. Isx9 increased the activity of the calcineurin (CN)/cytoplasmic nuclear factor of the activated T-cells (NFAT) transcription factor, a key regulator of D28K, and improved the recruitment of NFATc1, cAMP response element-binding protein (CREB), and p300 to the D28K promoter. We found that nutrient stimulation increased D28K plasma membrane enrichment and modulated calcium channel activity in order to regulate glucose-induced insulin secretion. Isx9-mediated expression of D28K protected β cells against chronic stress induced by serum withdrawal or chronic inflammation by reducing caspase 3 activity. Consequently, Isx9 improved human islet function after transplantation in NOD-SCID mice in a streptozotocin-induced diabetes model. In summary, Isx9 significantly regulates expression of genes relevant to β cell survival and function, and may be an attractive therapy to treat diabetes and improve islet function post-transplantation.
Collapse
|
20
|
Pramme-Steinwachs I, Jastroch M, Ussar S. Extracellular calcium modulates brown adipocyte differentiation and identity. Sci Rep 2017; 7:8888. [PMID: 28827782 PMCID: PMC5567186 DOI: 10.1038/s41598-017-09025-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 07/19/2017] [Indexed: 11/24/2022] Open
Abstract
Brown adipocytes are important in regulating non-shivering thermogenesis, whole body glucose and lipid homeostasis. Increasing evidence supports an important role of metabolites as well as macro- and micronutrients in brown adipocyte differentiation and function. Calcium is one of the most abundant ions in the body regulating multiple cellular processes. We observed that increasing extracellular calcium concentration during brown adipocyte differentiation blocks lipid accumulation and suppresses induction of major adipogenic transcription factors such as PPARγ and C/EBPα. In contrast, the depletion of calcium in the medium enhances adipogenesis and expression of brown adipocyte selective genes, such as UCP1. Mechanistically, we show that elevated extracellular calcium inhibits C/EBPβ activity through hyperactivation of ERK, a process that is independent of intracellular calcium levels and reversibly halts differentiation. Moreover, increased extracellular calcium solely after the induction phase of differentiation specifically suppresses gene expression of UCP1, PRDM16 and PGC1-α. Notably, depleting extracellular calcium provokes opposite effects. Together, we show that modulating extracellular calcium concentration controls brown adipocyte differentiation and thermogenic gene expression, highlighting the importance of tissue microenvironment on brown adipocyte heterogeneity and function.
Collapse
Affiliation(s)
- Ines Pramme-Steinwachs
- JRG Adipocytes & Metabolism, Institute for Diabetes & Obesity, Helmholtz Center Munich, 85748, Garching, Germany.,German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany
| | - Martin Jastroch
- German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany.,Institute for Diabetes & Obesity, Helmholtz Center Munich, 85748, Garching, Germany
| | - Siegfried Ussar
- JRG Adipocytes & Metabolism, Institute for Diabetes & Obesity, Helmholtz Center Munich, 85748, Garching, Germany. .,German Center for Diabetes Research (DZD), 85764, Neuherberg, Germany.
| |
Collapse
|
21
|
Nandipati KC, Subramanian S, Agrawal DK. Protein kinases: mechanisms and downstream targets in inflammation-mediated obesity and insulin resistance. Mol Cell Biochem 2016; 426:27-45. [PMID: 27868170 DOI: 10.1007/s11010-016-2878-8] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 11/07/2016] [Indexed: 12/23/2022]
Abstract
Obesity-induced low-grade inflammation (metaflammation) impairs insulin receptor signaling. This has been implicated in the development of insulin resistance. Insulin signaling in the target tissues is mediated by stress kinases such as p38 mitogen-activated protein kinase, c-Jun NH2-terminal kinase, inhibitor of NF-kB kinase complex β (IKKβ), AMP-activated protein kinase, protein kinase C, Rho-associated coiled-coil containing protein kinase, and RNA-activated protein kinase. Most of these kinases phosphorylate several key regulators in glucose homeostasis. The phosphorylation of serine residues in the insulin receptor and IRS-1 molecule results in diminished enzymatic activity in the phosphatidylinositol 3-kinase (PI3K)/Akt pathway. This has been one of the key mechanisms observed in the tissues that are implicated in insulin resistance especially in type 2 diabetes mellitus (T2-DM). Identifying the specific protein kinases involved in obesity-induced chronic inflammation may help in developing the targeted drug therapies to minimize the insulin resistance. This review is focused on the protein kinases involved in the inflammatory cascade and molecular mechanisms and their downstream targets with special reference to obesity-induced T2-DM.
Collapse
Affiliation(s)
- Kalyana C Nandipati
- Department of Surgery, Creighton University School of Medicine, 601 N. 30th Street, Suite # 3700, Omaha, NE, 68131, USA.
- Department of Clinical & Translational Science, Creighton University School of Medicine, 2500, California Plaza, Room # 510, Criss II, Omaha, NE, 68131, USA.
| | - Saravanan Subramanian
- Department of Clinical & Translational Science, Creighton University School of Medicine, 2500, California Plaza, Room # 510, Criss II, Omaha, NE, 68131, USA
| | - Devendra K Agrawal
- Department of Clinical & Translational Science, Creighton University School of Medicine, 2500, California Plaza, Room # 510, Criss II, Omaha, NE, 68131, USA
| |
Collapse
|
22
|
Meerson A, Yehuda H. Leptin and insulin up-regulate miR-4443 to suppress NCOA1 and TRAF4, and decrease the invasiveness of human colon cancer cells. BMC Cancer 2016; 16:882. [PMID: 27842582 PMCID: PMC5109693 DOI: 10.1186/s12885-016-2938-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 10/27/2016] [Indexed: 01/17/2023] Open
Abstract
Background Obesity is a risk factor for colorectal cancer (CRC). Normal and tumor cells respond to metabolic hormones, such as leptin and insulin. Thus, obesity-associated resistance to these hormones likely leads to changes in gene expression and behavior of tumor cells. However, the mechanisms affected by leptin and insulin signaling in CRC cells remain mostly unknown. Methods We hypothesized that microRNAs (miRNAs) are involved in the regulation of tumorigenesis-related gene expression in CRC cells by leptin and insulin. To test this hypothesis, miRNA levels in the CRC-derived cell lines HCT-116, HT-29 and DLD-1 were profiled, following leptin and insulin treatment. Candidate miRNAs were validated by RT-qPCR. Predicted miRNA targets with known roles in cancer, were validated by immunoblots and reporter assays in HCT-116 cells. Transfection of HCT-116 cells with candidate miRNA mimic was used to test in vitro effects on proliferation and invasion. Results Of ~800 miRNAs profiled, miR-4443 was consistently up-regulated by leptin and insulin in HCT-116 and HT-29, but not in DLD-1, which lacked normal leptin receptor expression. Dose response experiments showed that leptin at 100 ng/ml consistently up-regulated miR-4443 in HCT-116 cells, concomitantly with a significant decrease in cell invasion ability. Transfection with miR-4443 mimic decreased invasion and proliferation of HCT-116 cells. Moreover, leptin and miR-4443 transfection significantly down-regulated endogenous NCOA1 and TRAF4, both predicted targets of miR-4443 with known roles in cancer metastasis. miR-4443 was found to directly regulate TRAF4 and NCOA1, as validated by a reporter assay. The up-regulation of miR-4443 by leptin or insulin was attenuated by the inhibition of MEK1/2. Conclusions Our findings suggest that miR-4443 acts in a tumor-suppressive manner by down-regulating TRAF4 and NCOA1 downstream of MEK-C/EBP-mediated leptin and insulin signaling, and that insulin and/or leptin resistance (e.g. in obesity) may suppress this pathway and increase the risk of metastatic CRC. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2938-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ari Meerson
- Molecular Biology of Chronic Diseases, MIGAL Galilee Research Institute, PO Box 831, Kiryat Shmona, 11016, Israel. .,Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, Copenhagen, DK-2200, Denmark.
| | - Hila Yehuda
- Molecular Biology of Chronic Diseases, MIGAL Galilee Research Institute, PO Box 831, Kiryat Shmona, 11016, Israel
| |
Collapse
|
23
|
Kinase Signaling in Apoptosis Induced by Saturated Fatty Acids in Pancreatic β-Cells. Int J Mol Sci 2016; 17:ijms17091400. [PMID: 27626409 PMCID: PMC5037680 DOI: 10.3390/ijms17091400] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 08/18/2016] [Accepted: 08/22/2016] [Indexed: 12/12/2022] Open
Abstract
Pancreatic β-cell failure and death is considered to be one of the main factors responsible for type 2 diabetes. It is caused by, in addition to hyperglycemia, chronic exposure to increased concentrations of fatty acids, mainly saturated fatty acids. Molecular mechanisms of apoptosis induction by saturated fatty acids in β-cells are not completely clear. It has been proposed that kinase signaling could be involved, particularly, c-Jun N-terminal kinase (JNK), protein kinase C (PKC), p38 mitogen-activated protein kinase (p38 MAPK), extracellular signal-regulated kinase (ERK), and Akt kinases and their pathways. In this review, we discuss these kinases and their signaling pathways with respect to their possible role in apoptosis induction by saturated fatty acids in pancreatic β-cells.
Collapse
|
24
|
Bedinger DH, Adams SH. Metabolic, anabolic, and mitogenic insulin responses: A tissue-specific perspective for insulin receptor activators. Mol Cell Endocrinol 2015; 415:143-56. [PMID: 26277398 DOI: 10.1016/j.mce.2015.08.013] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 08/05/2015] [Accepted: 08/09/2015] [Indexed: 12/17/2022]
Abstract
Insulin acts as the major regulator of the fasting-to-fed metabolic transition by altering substrate metabolism, promoting energy storage, and helping activate protein synthesis. In addition to its glucoregulatory and other metabolic properties, insulin can also act as a growth factor. The metabolic and mitogenic responses to insulin are regulated by divergent post-receptor signaling mechanisms downstream from the activated insulin receptor (IR). However, the anabolic and growth-promoting properties of insulin require tissue-specific inter-relationships between the two pathways, and the nature and scope of insulin-regulated processes vary greatly across tissues. Understanding the nuances of this interplay between metabolic and growth-regulating properties of insulin would have important implications for development of novel insulin and IR modulator therapies that stimulate insulin receptor activation in both pathway- and tissue-specific manners. This review will provide a unique perspective focusing on the roles of "metabolic" and "mitogenic" actions of insulin signaling in various tissues, and how these networks should be considered when evaluating selective pharmacologic approaches to prevent or treat metabolic disease.
Collapse
Affiliation(s)
| | - Sean H Adams
- Arkansas Children's Nutrition Center and University of Arkansas for Medical Sciences, Department of Pediatrics, Little Rock, AR, USA
| |
Collapse
|
25
|
Wauson EM, Guerra ML, Dyachok J, McGlynn K, Giles J, Ross EM, Cobb MH. Differential Regulation of ERK1/2 and mTORC1 Through T1R1/T1R3 in MIN6 Cells. Mol Endocrinol 2015; 29:1114-22. [PMID: 26168033 DOI: 10.1210/me.2014-1181] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The MAPKs ERK1/2 respond to nutrients and other insulin secretagogues in pancreatic β-cells and mediate nutrient-dependent insulin gene transcription. Nutrients also stimulate the mechanistic target of rapamycin complex 1 (mTORC1) to regulate protein synthesis. We showed previously that activation of both ERK1/2 and mTORC1 in the MIN6 pancreatic β-cell-derived line by extracellular amino acids (AAs) is at least in part mediated by the heterodimeric T1R1/T1R3, a G protein-coupled receptor. We show here that AAs differentially activate these two signaling pathways in MIN6 cells. Pretreatment with pertussis toxin did not prevent the activation of either ERK1/2 or mTORC1 by AAs, indicating that G(I) is not central to either pathway. Although glucagon-like peptide 1, an agonist for a G(s-)coupled receptor, activated ERK1/2 well and mTORC1 to a small extent, AAs had no effect on cytosolic cAMP accumulation. Ca(2+) entry is required for ERK1/2 activation by AAs but is dispensable for AA activation of mTORC1. Pretreatment with UBO-QIC, a selective G(q) inhibitor, reduced the activation of ERK1/2 but had little effect on the activation of mTORC1 by AAs, suggesting a differential requirement for G(q). Inhibition of G(12/13) by the overexpression of the regulator of G protein signaling domain of p115 ρ-guanine nucleotide exchange factor had no effect on mTORC1 activation by AAs, suggesting that these G proteins are also not involved. We conclude that AAs regulate ERK1/2 and mTORC1 through distinct signaling pathways.
Collapse
Affiliation(s)
- Eric M Wauson
- Department of Pharmacology (E.M.W., M.L.G., J.D., K.M., E.M.R., M.H.C.) and the Green Center for Systems Biology (J.D., E.M.R.), University of Texas Southwestern Medical Center, Dallas, Texas 75390-9041; and Department of Physiology and Pharmacology (E.M.W., J.G.), Des Moines University, Des Moines, Iowa 50312
| | - Marcy L Guerra
- Department of Pharmacology (E.M.W., M.L.G., J.D., K.M., E.M.R., M.H.C.) and the Green Center for Systems Biology (J.D., E.M.R.), University of Texas Southwestern Medical Center, Dallas, Texas 75390-9041; and Department of Physiology and Pharmacology (E.M.W., J.G.), Des Moines University, Des Moines, Iowa 50312
| | - Julia Dyachok
- Department of Pharmacology (E.M.W., M.L.G., J.D., K.M., E.M.R., M.H.C.) and the Green Center for Systems Biology (J.D., E.M.R.), University of Texas Southwestern Medical Center, Dallas, Texas 75390-9041; and Department of Physiology and Pharmacology (E.M.W., J.G.), Des Moines University, Des Moines, Iowa 50312
| | - Kathleen McGlynn
- Department of Pharmacology (E.M.W., M.L.G., J.D., K.M., E.M.R., M.H.C.) and the Green Center for Systems Biology (J.D., E.M.R.), University of Texas Southwestern Medical Center, Dallas, Texas 75390-9041; and Department of Physiology and Pharmacology (E.M.W., J.G.), Des Moines University, Des Moines, Iowa 50312
| | - Jennifer Giles
- Department of Pharmacology (E.M.W., M.L.G., J.D., K.M., E.M.R., M.H.C.) and the Green Center for Systems Biology (J.D., E.M.R.), University of Texas Southwestern Medical Center, Dallas, Texas 75390-9041; and Department of Physiology and Pharmacology (E.M.W., J.G.), Des Moines University, Des Moines, Iowa 50312
| | - Elliott M Ross
- Department of Pharmacology (E.M.W., M.L.G., J.D., K.M., E.M.R., M.H.C.) and the Green Center for Systems Biology (J.D., E.M.R.), University of Texas Southwestern Medical Center, Dallas, Texas 75390-9041; and Department of Physiology and Pharmacology (E.M.W., J.G.), Des Moines University, Des Moines, Iowa 50312
| | - Melanie H Cobb
- Department of Pharmacology (E.M.W., M.L.G., J.D., K.M., E.M.R., M.H.C.) and the Green Center for Systems Biology (J.D., E.M.R.), University of Texas Southwestern Medical Center, Dallas, Texas 75390-9041; and Department of Physiology and Pharmacology (E.M.W., J.G.), Des Moines University, Des Moines, Iowa 50312
| |
Collapse
|
26
|
Bilodeau MS, Arguin G, Gendron FP. C/EBPβ regulates P2X7 receptor expression in response to glucose challenge in intestinal epithelial cells. Biochem Cell Biol 2015; 93:38-46. [DOI: 10.1139/bcb-2014-0098] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Activation of the ATP-dependent P2X7 receptor modulates glucose transport in intestinal epithelial cells through the downregulation of glucose transporter GLUT2. In the present study, we show that an increase in glucose concentration stimulates P2X7 receptor transcription via modulation of CCAAT/enhancer binding proteins (C/EBPs) α and β expression. The described human P2X7 receptor promoter region (GenBank Y12851) was cloned upstream of a luciferase reporter gene in pGL4.10 plasmid and used to determine whether C/EBPs, namely C/EBPα and C/EBPβ, are able to stimulate the transcription of P2X7 receptor. Results show that C/EBPβ was the main regulator of P2X7 receptor expression in response to a glucose challenge. Chromatin immunoprecipitation (ChIP) assays further revealed that C/EBPβ occupied the –213 to +6 nt P2X7 promoter region. Surprisingly, C/EBPα was also able to bind this region as revealed by ChIP assays, but without inducing receptor transcription. In fact, C/EBPα and the C/EBPβ-LIP isoform blocked the C/EBPβ-dependent regulation of P2X7 receptor transcription. These findings suggest that glucose is not only the major source of energy for cell function but may also act as a signaling molecule to stimulate the expression of regulatory proteins.
Collapse
Affiliation(s)
- Maude S. Bilodeau
- Department of Anatomy and Cell Biology, Faculté de Médecine et des Sciences de la Santé, Pavillon de Recherche Appliquée sur le Cancer, Université de Sherbrooke, Sherbrooke, 3201 Jean-Mignault, QC J1E 4K8, Canada
| | - Guillaume Arguin
- Department of Anatomy and Cell Biology, Faculté de Médecine et des Sciences de la Santé, Pavillon de Recherche Appliquée sur le Cancer, Université de Sherbrooke, Sherbrooke, 3201 Jean-Mignault, QC J1E 4K8, Canada
| | - Fernand-Pierre Gendron
- Department of Anatomy and Cell Biology, Faculté de Médecine et des Sciences de la Santé, Pavillon de Recherche Appliquée sur le Cancer, Université de Sherbrooke, Sherbrooke, 3201 Jean-Mignault, QC J1E 4K8, Canada
| |
Collapse
|
27
|
Huang HH, Stehno-Bittel L. Differences in insulin biosynthesis pathway between small and large islets do not correspond to insulin secretion. Islets 2015; 7:e1129097. [PMID: 26752360 PMCID: PMC4878277 DOI: 10.1080/19382014.2015.1129097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
In a variety of mammalian species, small islets secrete more insulin per volume than large islets. This difference may be due to diffusional limitations of large islets, or inherent differences in the insulin production pathways. The purpose of this study was to identify possible differences in the early phase of glucose-stimulated insulin biosynthesis between large and small islets. Isolated small and large rat islets were challenged with 30 minutes of high glucose. The expression of insulin gene transcription factors (MafA, NeuroD/ Beta2, and PDX-1), preproinsulin mRNA, proinsulin and insulin were compared between large and small islets. Under basal (low glucose) conditions, MafA and NeuroD had higher mRNA levels and greater protein amounts in large islets compared to small when normalized to GAPDH levels. 30 minutes of high glucose stimulation failed to alter the mRNA or subsequent protein levels of either gene. However, 30 minutes of high glucose suppressed activated PDX-1 protein levels in both small and large islets. High glucose stimulation did not statistically alter the preproinsulin mRNA (insulin 1 and insulin 2) levels. At the translational level, high glucose increased the proinsulin levels, and large islets showed a higher proinsulin content per cell than small islets. Insulin content per cell was not significantly different between small and large islets under basal or high glucose levels. The results fail to explain the higher level of insulin secretion noted in small versus large islets and may suggest that possible differences lie downstream in the secretory pathway rather than insulin biosynthesis.
Collapse
Affiliation(s)
- Han-Hung Huang
- Department of Physical Therapy Angelo State University; Texas Tech System; San Angelo, TX USA
| | - Lisa Stehno-Bittel
- Department of Physical Therapy and Rehabilitation Science; University of Kansas Medical Center; Kansas City, KS USA
- Likarda, LLC; Kansas City, KS USA
- Correspondence to: Lisa Stehno-Bittel;
| |
Collapse
|
28
|
Alejandro EU, Gregg B, Blandino-Rosano M, Cras-Méneur C, Bernal-Mizrachi E. Natural history of β-cell adaptation and failure in type 2 diabetes. Mol Aspects Med 2014; 42:19-41. [PMID: 25542976 DOI: 10.1016/j.mam.2014.12.002] [Citation(s) in RCA: 163] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 11/04/2014] [Accepted: 12/05/2014] [Indexed: 02/07/2023]
Abstract
Type 2 diabetes mellitus (T2D) is a complex disease characterized by β-cell failure in the setting of insulin resistance. The current evidence suggests that genetic predisposition, and environmental factors can impair the capacity of the β-cells to respond to insulin resistance and ultimately lead to their failure. However, genetic studies have demonstrated that known variants account for less than 10% of the overall estimated T2D risk, suggesting that additional unidentified factors contribute to susceptibility of this disease. In this review, we will discuss the different stages that contribute to the development of β-cell failure in T2D. We divide the natural history of this process in three major stages: susceptibility, β-cell adaptation and β-cell failure, and provide an overview of the molecular mechanisms involved. Further research into mechanisms will reveal key modulators of β-cell failure and thus identify possible novel therapeutic targets and potential interventions to protect against β-cell failure.
Collapse
Affiliation(s)
- Emilyn U Alejandro
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, Brehm Center for Diabetes Research, University of Michigan, Ann Arbor, MI, USA
| | - Brigid Gregg
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - Manuel Blandino-Rosano
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, Brehm Center for Diabetes Research, University of Michigan, Ann Arbor, MI, USA
| | - Corentin Cras-Méneur
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, Brehm Center for Diabetes Research, University of Michigan, Ann Arbor, MI, USA
| | - Ernesto Bernal-Mizrachi
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, Brehm Center for Diabetes Research, University of Michigan, Ann Arbor, MI, USA; VA Ann Arbor Healthcare System, Ann Arbor, MI, USA.
| |
Collapse
|
29
|
Lawrence MC, Borenstein-Auerbach N, McGlynn K, Kunnathodi F, Shahbazov R, Syed I, Kanak M, Takita M, Levy MF, Naziruddin B. NFAT targets signaling molecules to gene promoters in pancreatic β-cells. Mol Endocrinol 2014; 29:274-88. [PMID: 25496032 DOI: 10.1210/me.2014-1066] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Nuclear factor of activated T cells (NFAT) is activated by calcineurin in response to calcium signals derived by metabolic and inflammatory stress to regulate genes in pancreatic islets. Here, we show that NFAT targets MAPKs, histone acetyltransferase p300, and histone deacetylases (HDACs) to gene promoters to differentially regulate insulin and TNF-α genes. NFAT and ERK associated with the insulin gene promoter in response to glucagon-like peptide 1, whereas NFAT formed complexes with p38 MAPK (p38) and Jun N-terminal kinase (JNK) upon promoters of the TNF-α gene in response to IL-1β. Translocation of NFAT and MAPKs to gene promoters was calcineurin/NFAT dependent, and complex stability required MAPK activity. Knocking down NFATc2 expression, eliminating NFAT DNA binding sites, or interfering with NFAT nuclear import prevented association of MAPKs with gene promoters. Inhibiting p38 and JNK activity increased NFAT-ERK association with promoters, which repressed TNF-α and enhanced insulin gene expression. Moreover, inhibiting p38 and JNK induced a switch from NFAT-p38/JNK-histone acetyltransferase p300 to NFAT-ERK-HDAC3 complex formation upon the TNF-α promoter, which resulted in gene repression. Histone acetyltransferase/HDAC exchange was reversed on the insulin gene by p38/JNK inhibition in the presence of glucagon-like peptide 1, which enhanced gene expression. Overall, these data indicate that NFAT directs signaling enzymes to gene promoters in islets, which contribute to protein-DNA complex stability and promoter regulation. Furthermore, the data suggest that TNF-α can be repressed and insulin production can be enhanced by selectively targeting signaling components of NFAT-MAPK transcriptional/signaling complex formation in pancreatic β-cells. These findings have therapeutic potential for suppressing islet inflammation while preserving islet function in diabetes and islet transplantation.
Collapse
Affiliation(s)
- Michael C Lawrence
- Islet Cell Laboratory (M.C.L., N.B.-A., F.K., R.S., I.S., M.T.), Baylor Research Institute, Dallas, Texas 75226; Department of Pharmacology (K.M.), University of Texas Southwestern Medical Center, Dallas, Texas 75390; Institute of Biomedical Studies (M.K.), Baylor University, Waco, Texas 76798; and Annette C. and Harold C. Simmons Transplant Institute (M.F.L., B.N.), Baylor University Medical Center, Dallas, Texas 75246
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Luo Y, He F, Hu L, Hai L, Huang M, Xu Z, Zhang J, Zhou Z, Liu F, Dai YS. Transcription factor Ets1 regulates expression of thioredoxin-interacting protein and inhibits insulin secretion in pancreatic β-cells. PLoS One 2014; 9:e99049. [PMID: 24897113 PMCID: PMC4045976 DOI: 10.1371/journal.pone.0099049] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 05/08/2014] [Indexed: 11/18/2022] Open
Abstract
Long-term activation of extracellular-regulated kinase (ERK1/2) pathway has been shown to cause glucotoxicity and inhibit insulin gene expression in β-cells. Transcription factor Ets1 is activated by ERK1/2-mediated phosphorylation at the Thr38 residue. We hypothesize that Ets1 plays an important role in mediating ERK1/2 induced glucotoxicity in β-cells. We determined the role of Ets1 in Min6 cells and isolated mouse islets using overexpression and siRNA mediated knockdown of Ets1. The results show that Ets1 was localized in insulin-staining positive cells but not in glucagon-staining positive cells. Overexpression of Ets1 reduced glucose-stimulated insulin secretion in primary mouse islets. Overexpression of Ets1 in Min6 β-cells and mouse islets increased expression of thioredoxin-interacting protein (TXNIP). Conversely, knockdown of Ets1 by siRNA reduced expression of TXNIP in Min6 cells. Ets1 was associated with the txnip promoter in min6 cells and transfection of 293 cells with Ets1 and p300 synergistically increased txnip promoter reporter activity. Moreover, overexpression of Ets1 inhibited Min6 cell proliferation. Our results suggest that Ets1, by promoting TXNIP expression, negatively regulates β-cell function. Thus, over-activation of Ets1 may contribute to diet-induced β-cell dysfunction.
Collapse
Affiliation(s)
- Yan Luo
- Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fengli He
- Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Li Hu
- Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Luo Hai
- Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Meifeng Huang
- Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhipeng Xu
- Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jingjing Zhang
- Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhiguang Zhou
- Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Feng Liu
- Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Yan-Shan Dai
- Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- * E-mail:
| |
Collapse
|
31
|
Kanak MA, Takita M, Kunnathodi F, Lawrence MC, Levy MF, Naziruddin B. Inflammatory response in islet transplantation. Int J Endocrinol 2014; 2014:451035. [PMID: 24883060 PMCID: PMC4021753 DOI: 10.1155/2014/451035] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Revised: 02/04/2014] [Accepted: 02/12/2014] [Indexed: 12/23/2022] Open
Abstract
Islet cell transplantation is a promising beta cell replacement therapy for patients with brittle type 1 diabetes as well as refractory chronic pancreatitis. Despite the vast advancements made in this field, challenges still remain in achieving high frequency and long-term successful transplant outcomes. Here we review recent advances in understanding the role of inflammation in islet transplantation and development of strategies to prevent damage to islets from inflammation. The inflammatory response associated with islets has been recognized as the primary cause of early damage to islets and graft loss after transplantation. Details on cell signaling pathways in islets triggered by cytokines and harmful inflammatory events during pancreas procurement, pancreas preservation, islet isolation, and islet infusion are presented. Robust control of pre- and peritransplant islet inflammation could improve posttransplant islet survival and in turn enhance the benefits of islet cell transplantation for patients who are insulin dependent. We discuss several potent anti-inflammatory strategies that show promise for improving islet engraftment. Further understanding of molecular mechanisms involved in the inflammatory response will provide the basis for developing potent therapeutic strategies for enhancing the quality and success of islet transplantation.
Collapse
Affiliation(s)
- Mazhar A. Kanak
- Institute for Biomedical Studies, Baylor University, Waco, TX 76712, USA
| | - Morihito Takita
- Islet Cell Laboratory, Baylor Research Institute, Dallas, TX 75204, USA
| | - Faisal Kunnathodi
- Islet Cell Laboratory, Baylor Research Institute, Dallas, TX 75204, USA
| | | | - Marlon F. Levy
- Baylor Annette C. and Harold C. Simmons Transplant Institute, 3410 Worth Street, Dallas, TX 75246, USA
| | - Bashoo Naziruddin
- Baylor Annette C. and Harold C. Simmons Transplant Institute, 3410 Worth Street, Dallas, TX 75246, USA
| |
Collapse
|
32
|
Setyowati Karolina D, Sepramaniam S, Tan HZ, Armugam A, Jeyaseelan K. miR-25 and miR-92a regulate insulin I biosynthesis in rats. RNA Biol 2014; 10:1365-78. [PMID: 24084692 DOI: 10.4161/rna.25557] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The 3' UTR of insulin has been identified as a critical region that confers mRNA stability, which is crucial for promoting transcription in response to glucose challenge. miRNAs are endogenously encoded non-coding RNAs that function as regulators of gene expression. This regulatory function is generally mediated by complementary binding to the 3'UTR of its mRNA targets that affects subsequent translational process. Genes involved in the regulation of glucose homeostasis, particularly in insulin production, have been found as targets of several miRNAs. Yet, no direct miRNA-based regulators of insulin biosynthesis have been identified. In this study, identification of possible miRNA-based regulators of insulin production is explored. Members of a miRNA family, miR-25 and miR-92a, are found as direct modulators of insulin expression. Overexpression of miR-25 or miR-92a reduced insulin expression while inhibition of miR-25 and miR-92a expression using corresponding antagomiRs promoted insulin expression and ultimately enhanced glucose-induced insulin secretion. Furthermore, suppression of insulin secretion by pre miR-9 could be attenuated by treatment with anti-miR-25 or miR-92a. Interestingly, we found the binding site of miR-25 and miR-92a to overlap with that of PTBP1, an important RNA binding molecule that stabilizes insulin mRNA for translation. Despite the increase in PTBP1 protein in the pancreas of diabetic rats, we observed insulin expression to be reduced alongside upregulation of miR-25 and miR-92a, suggesting an intricate regulation of insulin (bio)synthesis at its mRNA level.
Collapse
Affiliation(s)
- Dwi Setyowati Karolina
- Department of Biochemistry; Yong Loo Lin School of Medicine; National University Health System; Singapore
| | | | | | | | | |
Collapse
|
33
|
Osborne JK, Guerra ML, Gonzales JX, McMillan EA, Minna JD, Cobb MH. NeuroD1 mediates nicotine-induced migration and invasion via regulation of the nicotinic acetylcholine receptor subunits in a subset of neural and neuroendocrine carcinomas. Mol Biol Cell 2014; 25:1782-92. [PMID: 24719457 PMCID: PMC4038504 DOI: 10.1091/mbc.e13-06-0316] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Nicotine up-regulates NeuroD1 in bronchial epithelial cells and certain undifferentiated carcinomas. NeuroD1 enhances expression of nicotinic acetylcholine receptor subunits. Increased invasion in Matrigel depends on these receptor subunits. Nicotine may induce positive feedback through NeuroD1 and increased expression of its own receptor. Cigarette smoking is a major risk factor for acquisition of small cell lung cancer (SCLC). A role has been demonstrated for the basic helix-loop-helix transcription factor NeuroD1 in the pathogenesis of neural and neuroendocrine lung cancer, including SCLC. In the present study we investigate the possible function of NeuroD1 in established tumors, as well as actions early on in pathogenesis, in response to nicotine. We demonstrate that nicotine up-regulates NeuroD1 in immortalized normal bronchial epithelial cells and a subset of undifferentiated carcinomas. Increased expression of NeuroD1 subsequently leads to regulation of expression and function of the nicotinic acetylcholine receptor subunit cluster of α3, α5, and β4. In addition, we find that coordinated expression of these subunits by NeuroD1 leads to enhanced nicotine-induced migration and invasion, likely through changes in intracellular calcium. These findings suggest that aspects of the pathogenesis of neural and neuroendocrine lung cancers may be affected by a nicotine- and NeuroD1-induced positive feedback loop.
Collapse
Affiliation(s)
- Jihan K Osborne
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9041
| | - Marcy L Guerra
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9041
| | - Joshua X Gonzales
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9041
| | - Elizabeth A McMillan
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9041
| | - John D Minna
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9041Hamon Cancer Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX 75390-9041
| | - Melanie H Cobb
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9041
| |
Collapse
|
34
|
Guerra ML, Wauson EM, McGlynn K, Cobb MH. Muscarinic control of MIN6 pancreatic β cells is enhanced by impaired amino acid signaling. J Biol Chem 2014; 289:14370-9. [PMID: 24695728 DOI: 10.1074/jbc.m114.565069] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We have shown recently that the class C G protein-coupled receptor T1R1/T1R3 taste receptor complex is an early amino acid sensor in MIN6 pancreatic β cells. Amino acids are unable to activate ERK1/2 in β cells in which T1R3 has been depleted. The muscarinic receptor agonist carbachol activated ERK1/2 better in T1R3-depleted cells than in control cells. Ligands that activate certain G protein-coupled receptors in pancreatic β cells potentiate glucose-stimulated insulin secretion. Among these is the M3 muscarinic acetylcholine receptor, the major muscarinic receptor in β cells. We found that expression of M3 receptors increased in T1R3-depleted MIN6 cells and that calcium responses were altered. To determine whether these changes were related to impaired amino acid signaling, we compared responses in cells exposed to reduced amino acid concentrations. M3 receptor expression was increased, and some, but not all, changes in calcium signaling were mimicked. These findings suggest that M3 acetylcholine receptors are increased in β cells as a mechanism to compensate for amino acid deficiency.
Collapse
Affiliation(s)
- Marcy L Guerra
- From the Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390-9041
| | - Eric M Wauson
- From the Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390-9041
| | - Kathleen McGlynn
- From the Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390-9041
| | - Melanie H Cobb
- From the Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390-9041
| |
Collapse
|
35
|
Sidharthan NP, Minchin RF, Butcher NJ. Cytosolic sulfotransferase 1A3 is induced by dopamine and protects neuronal cells from dopamine toxicity: role of D1 receptor-N-methyl-D-aspartate receptor coupling. J Biol Chem 2013; 288:34364-74. [PMID: 24136195 DOI: 10.1074/jbc.m113.493239] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Dopamine neurotoxicity is associated with several neurodegenerative diseases, and neurons utilize several mechanisms, including uptake and metabolism, to protect them from injury. Metabolism of dopamine involves three enzymes: monoamine oxidase, catechol O-methyltransferase, and sulfotransferase. In primates but not lower order animals, a sulfotransferase (SULT1A3) is present that can rapidly metabolize dopamine to dopamine sulfate. Here, we show that SULT1A3 and a closely related protein SULT1A1 are highly inducible by dopamine. This involves activation of the D1 and NMDA receptors. Both ERK1/2 phosphorylation and calcineurin activation are required for induction. Pharmacological agents that inhibited induction or siRNA targeting SULT1A3 significantly increased the susceptibility of cells to dopamine toxicity. Taken together, these results show that dopamine can induce its own metabolism and protect neuron-like cells from damage, suggesting that SULT1A3 activity may be a risk factor for dopamine-dependent neurodegenerative diseases.
Collapse
Affiliation(s)
- Neelima P Sidharthan
- From the School of Biomedical Sciences, University of Queensland, Brisbane, Queensland, Australia 4072
| | | | | |
Collapse
|
36
|
NeuroD1 regulation of migration accompanies the differential sensitivity of neuroendocrine carcinomas to TrkB inhibition. Oncogenesis 2013; 2:e63. [PMID: 23958853 PMCID: PMC3759124 DOI: 10.1038/oncsis.2013.24] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2013] [Revised: 06/07/2013] [Accepted: 06/13/2013] [Indexed: 01/09/2023] Open
Abstract
The developmental transcription factor NeuroD1 is anomalously expressed in a subset of aggressive neuroendocrine tumors. Previously, we demonstrated that TrkB and neural cell adhesion molecule (NCAM) are downstream targets of NeuroD1 that contribute to the actions of neurogenic differentiation 1 (NeuroD1) in neuroendocrine lung. We found that several malignant melanoma and prostate cell lines express NeuroD1 and TrkB. Inhibition of TrkB activity decreased invasion in several neuroendocrine pigmented melanoma but not in prostate cell lines. We also found that loss of the tumor suppressor p53 increased NeuroD1 expression in normal human bronchial epithelial cells and cancer cells with neuroendocrine features. Although we found that a major mechanism of action of NeuroD1 is by the regulation of TrkB, effective targeting of TrkB to inhibit invasion may depend on the cell of origin. These findings suggest that NeuroD1 is a lineage-dependent oncogene acting through its downstream target, TrkB, across multiple cancer types, which may provide new insights into the pathogenesis of neuroendocrine cancers.
Collapse
|
37
|
Osman MA, Sarkar FH, Rodriguez-Boulan E. A molecular rheostat at the interface of cancer and diabetes. Biochim Biophys Acta Rev Cancer 2013; 1836:166-76. [PMID: 23639840 DOI: 10.1016/j.bbcan.2013.04.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Accepted: 04/23/2013] [Indexed: 12/17/2022]
Abstract
Epidemiology studies revealed the connection between several types of cancer and type 2 diabetes (T2D) and suggested that T2D is both a symptom and a risk factor of pancreatic cancer. High level of circulating insulin (hyperinsulinemia) in obesity has been implicated in promoting aggressive types of cancers. Insulin resistance, a symptom of T2D, pressures pancreatic β-cells to increase insulin secretion, leading to hyperinsulinemia, which in turn leads to a gradual loss of functional β-cell mass, thus indicating a fine balance and interplay between β-cell function and mass. While the mechanisms of these connections are unclear, the mTORC1-Akt signaling pathway has been implicated in controlling β-cell function and mass, and in mediating the link of cancer and T2D. However, incomplete understating of how the pathway is regulated and how it integrates body metabolism has hindered its efficacy as a clinical target. The IQ motif containing GTPase activating protein 1 (IQGAP1)-Exocyst axis is a growth factor- and nutrient-sensor that couples cell growth and division. Here we discuss how IQGAP1-Exocyst, through differential interactions with Rho-type of small guanosine triphosphatases (GTPases), acts as a rheostat that modulates the mTORC1-Akt and MAPK signals, and integrates β-cell function and mass with insulin signaling, thus providing a molecular mechanism for cancer initiation in diabetes. Delineating this regulatory pathway may have the potential of contributing to optimizing the efficacy and selectivity of future therapies for cancer and diabetes.
Collapse
Affiliation(s)
- Mahasin A Osman
- Warren Alpert Medical School, Division of Biology and Medicine, Molecular Pharmacology, Physiology and Biotechnology, Brown University, Providence, RI 02912, USA.
| | | | | |
Collapse
|
38
|
NeuroD1 regulates survival and migration of neuroendocrine lung carcinomas via signaling molecules TrkB and NCAM. Proc Natl Acad Sci U S A 2013; 110:6524-9. [PMID: 23553831 DOI: 10.1073/pnas.1303932110] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Small-cell lung cancer and other aggressive neuroendocrine cancers are often associated with early dissemination and frequent metastases. We demonstrate that neurogenic differentiation 1 (NeuroD1) is a regulatory hub securing cross talk among survival and migratory-inducing signaling pathways in neuroendocrine lung carcinomas. We find that NeuroD1 promotes tumor cell survival and metastasis in aggressive neuroendocrine lung tumors through regulation of the receptor tyrosine kinase tropomyosin-related kinase B (TrkB). Like TrkB, the prometastatic signaling molecule neural cell adhesion molecule (NCAM) is a downstream target of NeuroD1, whose impaired expression mirrors loss of NeuroD1. TrkB and NCAM may be therapeutic targets for aggressive neuroendocrine cancers that express NeuroD1.
Collapse
|
39
|
Goodyer WR, Gu X, Liu Y, Bottino R, Crabtree GR, Kim SK. Neonatal β cell development in mice and humans is regulated by calcineurin/NFAT. Dev Cell 2012; 23:21-34. [PMID: 22814600 DOI: 10.1016/j.devcel.2012.05.014] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2011] [Revised: 01/04/2012] [Accepted: 05/19/2012] [Indexed: 11/29/2022]
Abstract
Little is known about the mechanisms governing neonatal growth and maturation of organs. Here we demonstrate that calcineurin/Nuclear Factor of Activated T cells (Cn/NFAT) signaling regulates neonatal pancreatic development in mouse and human islets. Inactivation of calcineurin b1 (Cnb1) in mouse islets impaired dense core granule biogenesis, decreased insulin secretion, and reduced cell proliferation and mass, culminating in lethal diabetes. Pancreatic β cells lacking Cnb1 failed to express genes revealed to be direct NFAT targets required for replication, insulin storage, and secretion. In contrast, glucokinase activation stimulated Cn-dependent expression of these genes. Calcineurin inhibitors, such as tacrolimus, used for human immunosuppression, induce diabetes. Tacrolimus exposure reduced Cn/NFAT-dependent expression of factors essential for insulin dense core granule formation and secretion and neonatal β cell proliferation, consistent with our genetic studies. Discovery of conserved pathways regulating β cell maturation and proliferation suggests new strategies for controlling β cell growth or replacement in human islet diseases.
Collapse
Affiliation(s)
- William R Goodyer
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | | | | | | | |
Collapse
|
40
|
Costa MM, Violato NM, Taboga SR, Góes RM, Bosqueiro JR. Reduction of insulin signalling pathway IRS-1/IRS-2/AKT/mTOR and decrease of epithelial cell proliferation in the prostate of glucocorticoid-treated rats. Int J Exp Pathol 2012; 93:188-95. [PMID: 22583132 DOI: 10.1111/j.1365-2613.2012.00817.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Previous studies by our research group using a model of insulin resistance induced by dexamethasone (DEX) showed that in the rat ventral prostate there was epithelial and smooth muscle cell atrophy and there were also alterations in fibroblasts. Proteins of the insulin signalling pathway are known to be very important for cell proliferation and development. Thus, we investigated the insulin signalling pathway and epithelial proliferation in the rat ventral prostate in this model and correlated the findings with expression of glucocorticoid (GR) and androgen (AR) receptors. Insulin resistance was induced in adult male Wistar rats by injection of DEX (1 mg/kg, ip for 5 consecutive days), whereas control (CTL) rats received saline. DEX treatment resulted in a significant decrease in body weight, but not in prostate weight. Reductions in insulin receptor 1 (IRS-1) (CTL 1.11 ± 0.06; DEX 0.85 ± 0.03), IRS-2 (CTL 0.95 ± 0.05; DEX 0.49 ± 0.04), AKT (CTL 0.98 ± 0.03; DEX 0.78 ± 0.02), mammalian target of rapamycin (mTOR; CTL 0.65 ± 0.08; DEX 0.22 ± 0.05), GR (CTL 1.30 ± 0.09; DEX 0.57 ± 0.10) and AR (CTL 1.83 ± 0.16; DEX 0.55 ± 0.08) protein levels were observed in the prostate of DEX-treated rats. The expression of the IRα-subunit, phosphoinositide 3-kinase, p-AKT, p70(S6K) , extracellular signal-regulated kinase (ERK) and p-ERK was not altered. The frequency of AR-positive cells in the epithelium of the prostate decreased in the glucocorticoid-treated group, and the intensity of the reaction for this receptor in the cell nuclei was lower in this group. Furthermore, the treatment with DEX reduced the frequency of proliferating cell nuclear antigen-positive (PCNA) cells 30-fold. This study suggests that the reduction in the insulin signalling pathway proteins IRS-1/IRS-2/AKT/mTOR in the prostate of DEX-treated rats may be associated with the morphological alterations observed previously.
Collapse
Affiliation(s)
- Maitê M Costa
- Department of Physical Education, Faculty of Sciences, São Paulo State University-Unesp, Bauru, São Paulo, Brazil
| | | | | | | | | |
Collapse
|
41
|
Neuronal Cbl controls biosynthesis of insulin-like peptides in Drosophila melanogaster. Mol Cell Biol 2012; 32:3610-23. [PMID: 22778134 DOI: 10.1128/mcb.00592-12] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The Cbl family proteins function as both E3 ubiquitin ligases and adaptor proteins to regulate various cellular signaling events, including the insulin/insulin-like growth factor 1 (IGF1) and epidermal growth factor (EGF) pathways. These pathways play essential roles in growth, development, metabolism, and survival. Here we show that in Drosophila melanogaster, Drosophila Cbl (dCbl) regulates longevity and carbohydrate metabolism through downregulating the production of Drosophila insulin-like peptides (dILPs) in the brain. We found that dCbl was highly expressed in the brain and knockdown of the expression of dCbl specifically in neurons by RNA interference increased sensitivity to oxidative stress or starvation, decreased carbohydrate levels, and shortened life span. Insulin-producing neuron-specific knockdown of dCbl resulted in similar phenotypes. dCbl deficiency in either the brain or insulin-producing cells upregulated the expression of dilp genes, resulting in elevated activation of the dILP pathway, including phosphorylation of Drosophila Akt and Drosophila extracellular signal-regulated kinase (dERK). Genetic interaction analyses revealed that blocking Drosophila epidermal growth factor receptor (dEGFR)-dERK signaling in pan-neurons or insulin-producing cells by overexpressing a dominant-negative form of dEGFR abolished the effect of dCbl deficiency on the upregulation of dilp genes. Furthermore, knockdown of c-Cbl in INS-1 cells, a rat β-cell line, also increased insulin biosynthesis and glucose-stimulated secretion in an ERK-dependent manner. Collectively, these results suggest that neuronal dCbl regulates life span, stress responses, and metabolism by suppressing dILP production and the EGFR-ERK pathway mediates the dCbl action. Cbl suppression of insulin biosynthesis is evolutionarily conserved, raising the possibility that Cbl may similarly exert its physiological actions through regulating insulin production in β cells.
Collapse
|
42
|
Lipopolysaccharides impair insulin gene expression in isolated islets of Langerhans via Toll-Like Receptor-4 and NF-κB signalling. PLoS One 2012; 7:e36200. [PMID: 22558381 PMCID: PMC3338606 DOI: 10.1371/journal.pone.0036200] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2012] [Accepted: 04/03/2012] [Indexed: 12/12/2022] Open
Abstract
Background Type 2 diabetes is characterized by pancreatic β-cell dysfunction and is associated with low-grade inflammation. Recent observations suggest that the signalling cascade activated by lipopolysaccharides (LPS) binding to Toll-Like Receptor 4 (TLR4) exerts deleterious effects on pancreatic β-cell function; however, the molecular mechanisms of these effects are incompletely understood. In this study, we tested the hypothesis that LPS alters insulin gene expression via TLR4 and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) in islets. Methodology/Principal Findings A 24-h exposure of isolated human, rat and mouse islets of Langerhans to LPS dose-dependently reduced insulin gene expression. This was associated in mouse and rat islets with decreased mRNA expression of pancreas-duodenum homebox-1 (PDX-1) and mammalian homologue of avian MafA/l-Maf (MafA). Accordingly, LPS exposure also decreased glucose-induced insulin secretion. LPS repression of insulin, PDX-1 and MafA expression, as well as its inhibition of insulin secretion, were not observed in islets from TLR4-deficient mice. LPS inhibition of β-cell gene expression in rat islets was prevented by inhibition of the NF-κB pathway, but not the p38 mitogen-activated protein kinase (p38 MAPK) pathway. Conclusions/Significance Our findings demonstrate that LPS inhibit β-cell gene expression in a TLR4-dependent manner and via NF-κB signaling in pancreatic islets, suggesting a novel mechanism by which the gut microbiota might affect pancreatic β-cell function.
Collapse
|
43
|
Li X, Xue B, Wang X, Sun L, Zhang T, Qu L, Zou X, Mu Y. Reduced expression of the LRP16 gene in mouse insulinoma (MIN6) cells exerts multiple effects on insulin content, proliferation and apoptosis. ACTA ACUST UNITED AC 2012; 32:190-198. [PMID: 22528219 DOI: 10.1007/s11596-012-0034-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2011] [Indexed: 01/12/2023]
Abstract
This study assessed the effects of leukemia-related protein 16 (LRP16) on the regulation of pancreatic functions in mouse insulinoma (MIN6) cells. Cells with down-regulated expression of LRP16 were obtained by a shRNA interference strategy. Insulin content and glucose-stimulated insulin secretion (GSIS) were examined by radioimmunoassay. Western blotting was applied to detect protein expression. Glucose-stimulated sub-cellular localization of PDX-1 was immunocytochemically determined. Cell proliferation and apoptosis were detected by flow cytometry. Our results showed that LRP16 regulated insulin content in MIN6 cells by controlling expression of insulin and insulin transcription factors. LRP16 gene silence in MIN6 cells led to reduced cell proliferation and increased apoptosis. The observation of phosphorylation of serine-473 Akt and the localization of PDX-1 to the nucleus under glucose-stimulation exhibited that LRP16 was a component mediating Akt signaling in MIN6 cells. These results suggest that LRP16 plays a key role in maintaining pancreatic β-cell functions and may help us to understand the protective effects of estrogen on the functions of pancreatic β-cells.
Collapse
Affiliation(s)
- Xiaojin Li
- Department of Endocrinology, Chinese PLA General Hospital, Beijing, 100853, China
| | - Bing Xue
- Department of Endocrinology, Chinese PLA General Hospital, Beijing, 100853, China
| | - Xuan Wang
- Department of Endocrinology, Chinese PLA General Hospital, Beijing, 100853, China
| | - Lianqing Sun
- Department of Endocrinology, Chinese PLA General Hospital, Beijing, 100853, China
| | - Tingting Zhang
- Department of Endocrinology, Chinese PLA General Hospital, Beijing, 100853, China
| | - Ling Qu
- Department of Endocrinology, Chinese PLA General Hospital, Beijing, 100853, China
| | - Xiaoman Zou
- Department of Endocrinology, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yiming Mu
- Department of Endocrinology, Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
44
|
Selway J, Rigatti R, Storey N, Lu J, Willars GB, Herbert TP. Evidence that Ca2+ within the microdomain of the L-type voltage gated Ca2+ channel activates ERK in MIN6 cells in response to glucagon-like peptide-1. PLoS One 2012; 7:e33004. [PMID: 22412973 PMCID: PMC3296766 DOI: 10.1371/journal.pone.0033004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Accepted: 02/08/2012] [Indexed: 11/27/2022] Open
Abstract
Glucagon like peptide-1 (GLP-1) is released from intestinal L-cells in response to nutrient ingestion and acts upon pancreatic β-cells potentiating glucose-stimulated insulin secretion and stimulating β-cell proliferation, differentiation, survival and gene transcription. These effects are mediated through the activation of multiple signal transduction pathways including the extracellular regulated kinase (ERK) pathway. We have previously reported that GLP-1 activates ERK through a mechanism dependent upon the influx of extracellular Ca2+ through L-type voltage gated Ca2+ channels (VGCC). However, the mechanism by which L-type VGCCs couple to the ERK signalling pathway in pancreatic β-cells is poorly understood. In this report, we characterise the relationship between L-type VGCC mediated changes in intracellular Ca2+ concentration ([Ca2+]i) and the activation of ERK, and demonstrate that the sustained activation of ERK (up to 30 min) in response to GLP-1 requires the continual activation of the L-type VGCC yet does not require a sustained increase in global [Ca2+]i or Ca2+ efflux from the endoplasmic reticulum. Moreover, sustained elevation of [Ca2+]i induced by ionomycin is insufficient to stimulate the prolonged activation of ERK. Using the cell permeant Ca2+ chelators, EGTA-AM and BAPTA-AM, to determine the spatial dynamics of L-type VGCC-dependent Ca2+ signalling to ERK, we provide evidence that a sustained increase in Ca2+ within the microdomain of the L-type VGCC is sufficient for signalling to ERK and that this plays an important role in GLP-1- stimulated ERK activation.
Collapse
Affiliation(s)
- Joanne Selway
- Department of Cell Physiology and Pharmacology, Henry Wellcome Building, University of Leicester, Leicester, United Kingdom
| | | | | | | | | | | |
Collapse
|
45
|
Lombardi A, Ulianich L, Treglia AS, Nigro C, Parrillo L, Lofrumento DD, Nicolardi G, Garbi C, Beguinot F, Miele C, Di Jeso B. Increased hexosamine biosynthetic pathway flux dedifferentiates INS-1E cells and murine islets by an extracellular signal-regulated kinase (ERK)1/2-mediated signal transmission pathway. Diabetologia 2012; 55:141-53. [PMID: 22006246 DOI: 10.1007/s00125-011-2315-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Accepted: 08/25/2011] [Indexed: 11/26/2022]
Abstract
AIMS/HYPOTHESIS Beta cell failure is caused by loss of cell mass, mostly by apoptosis, but also by simple dysfunction (decline of glucose-stimulated insulin secretion, downregulation of specific gene expression). Apoptosis and dysfunction are caused, at least in part, by lipoglucotoxicity. The mechanisms implicated are oxidative stress, increase in the hexosamine biosynthetic pathway (HBP) flux and endoplasmic reticulum (ER) stress. Oxidative stress plays a role in glucotoxicity-induced beta cell dedifferentiation, while glucotoxicity-induced ER stress has been mostly linked to beta cell apoptosis. We sought to clarify whether ER stress caused by increased HBP flux participates in a dedifferentiating response of beta cells, in the absence of relevant apoptosis. METHODS We used INS-1E cells and murine islets. We analysed the unfolded protein response and the expression profile of beta cells by real-time RT-PCR and western blot. The signal transmission pathway elicited by ER stress was investigated by real-time RT-PCR and immunofluorescence. RESULTS Glucosamine and high glucose induced ER stress, but did not decrease cell viability in INS-1E cells. ER stress caused dedifferentiation of beta cells, as shown by downregulation of beta cell markers and of the transcription factor, pancreatic and duodenal homeobox 1. Glucose-stimulated insulin secretion was inhibited. These effects were prevented by the chemical chaperone, 4-phenyl butyric acid. The extracellular signal-regulated kinase (ERK) signal transmission pathway was implicated, since its inhibition prevented the effects induced by glucosamine and high glucose. CONCLUSIONS/INTERPRETATION Glucotoxic ER stress dedifferentiates beta cells, in the absence of apoptosis, through a transcriptional response. These effects are mediated by the activation of ERK1/2.
Collapse
Affiliation(s)
- A Lombardi
- Dipartimento di Scienze e Tecnologie Biologiche e Ambientali, Università degli Studi del Salento, 73100 Lecce, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Osborne JK, Zaganjor E, Cobb MH. Signal control through Raf: in sickness and in health. Cell Res 2011; 22:14-22. [PMID: 22143568 DOI: 10.1038/cr.2011.193] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The extracellular signal-regulated kinase 1/2 (ERK1/2) cascade is the prototype mammalian mitogen-activated protein kinase (MAPK) signaling cascade that regulates a number of processes, including proliferation, differentiation, survival, migration, stress responses and apoptosis. How this seemingly linear cascade is modulated to achieve a specific cellular function has been a main focus of the field. In this review, we describe new as well as old findings in the regulation of the ERK1/2 pathway in normal and disease states via MAP3Ks.
Collapse
Affiliation(s)
- Jihan K Osborne
- Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390-9041, USA
| | | | | |
Collapse
|
47
|
A small molecule differentiation inducer increases insulin production by pancreatic β cells. Proc Natl Acad Sci U S A 2011; 108:20713-8. [PMID: 22143803 DOI: 10.1073/pnas.1118526109] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
New drugs for preserving and restoring pancreatic β-cell function are critically needed for the worldwide epidemic of type 2 diabetes and the cure for type 1 diabetes. We previously identified a family of neurogenic 3,5-disubstituted isoxazoles (Isx) that increased expression of neurogenic differentiation 1 (NeuroD1, also known as BETA2); this transcription factor functions in neuronal and pancreatic β-cell differentiation and is essential for insulin gene transcription. Here, we probed effects of Isx on human cadaveric islets and MIN6 pancreatic β cells. Isx increased the expression and secretion of insulin in islets that made little insulin after prolonged ex vivo culture and increased expression of neurogenic differentiation 1 and other regulators of islet differentiation and insulin gene transcription. Within the first few hours of exposure, Isx caused biphasic activation of ERK1/2 and increased bulk histone acetylation. Although there was little effect on histone deacetylase activity, Isx increased histone acetyl transferase activity in nuclear extracts. Reconstitution assays indicated that Isx increased the activity of the histone acetyl transferase p300 through an ERK1/2-dependent mechanism. In summary, we have identified a small molecule with antidiabetic activity, providing a tool for exploring islet function and a possible lead for therapeutic intervention in diabetes.
Collapse
|
48
|
Li HQ, Wang BP, Deng XL, Zhang JY, Wang YB, Zheng J, Xia WF, Zeng TS, Chen LL. Insulin improves β-cell function in glucose-intolerant rat models induced by feeding a high-fat diet. Metabolism 2011; 60:1566-74. [PMID: 21550078 DOI: 10.1016/j.metabol.2011.01.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2009] [Revised: 01/11/2011] [Accepted: 01/29/2011] [Indexed: 01/09/2023]
Abstract
Insulin therapy has been shown to contribute to extended glycemia remission in newly diagnosed patients with type 2 diabetes mellitus. This study investigated the effects of insulin treatment on pancreatic lipid content, and β-cell apoptosis and proliferation in glucose-intolerant rats to explore the protective role of insulin on β-cell function. A rat glucose-intolerant model was induced by streptozotocin and a high-fat diet. Plasma and pancreatic triglycerides, free fatty acids, and insulin were measured; and pancreatic β-cell cell apoptosis and proliferation were detected by a propidium iodide cell death assay and immunofluorescence for proliferating cell nuclear antigen. Relative β-cell area was determined by immunohistochemistry for insulin, whereas insulin production in pancreas was assessed by reverse transcriptase polymerase chain reaction. Islet β-cell secreting function was assessed by the index ΔI30/ΔG30. Glucose-intolerant rats had higher pancreatic lipid content, more islet β-cell apoptosis, lower β-cell proliferation, and reduced β-cell area in pancreas when compared with controls. Insulin therapy reduced blood glucose, inhibited pancreatic lipid accumulation and islet β-cell apoptosis, and increased β-cell proliferation and β-cell area in glucose-intolerant rats. Furthermore, impaired insulin secretion and insulin production in glucose-intolerant rats were improved by insulin therapy. Insulin can preserve β-cell function by protecting islets from glucotoxicity and lipotoxicity. It can also ameliorate β-cell area by enhancing β-cell proliferation and reducing β-cell apoptosis.
Collapse
MESH Headings
- Animals
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/drug therapy
- Diabetes Mellitus, Experimental/physiopathology
- Diet, High-Fat
- Disease Models, Animal
- Drug Evaluation, Preclinical
- Eating/physiology
- Fatty Acids, Nonesterified/analysis
- Fatty Acids, Nonesterified/blood
- Fatty Acids, Nonesterified/metabolism
- Glucose Intolerance/drug therapy
- Glucose Intolerance/etiology
- Glucose Intolerance/genetics
- Glucose Intolerance/physiopathology
- Insulin/analysis
- Insulin/genetics
- Insulin/pharmacology
- Insulin/therapeutic use
- Insulin-Secreting Cells/drug effects
- Insulin-Secreting Cells/metabolism
- Insulin-Secreting Cells/physiology
- Male
- Pancreas/chemistry
- Pancreas/metabolism
- Rats
- Rats, Wistar
- Streptozocin
- Triglycerides/analysis
- Triglycerides/blood
- Triglycerides/metabolism
Collapse
Affiliation(s)
- Hui-qing Li
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Alejandro EU, Lim GE, Mehran AE, Hu X, Taghizadeh F, Pelipeychenko D, Baccarini M, Johnson JD. Pancreatic β-cell Raf-1 is required for glucose tolerance, insulin secretion, and insulin 2 transcription. FASEB J 2011; 25:3884-95. [PMID: 21817126 DOI: 10.1096/fj.10-180349] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Regulation of glucose homeostasis by insulin depends on pancreatic β-cell growth, survival, and function. Raf-1 kinase is a major downstream target of several growth factors that promote proliferation and survival of many cell types, including the pancreatic β cells. We have previously reported that insulin protects β cells from apoptosis and promotes proliferation by activating Raf-1 signaling in cultured human islets, mouse islets, and MIN6 cells. As Raf-1 activity is critical for basal apoptosis and insulin secretion in vitro, we hypothesized that Raf-1 may play an important role in glucose homeostasis in vivo. To test this hypothesis, we utilized the Cre-loxP recombination system to obtain a pancreatic β-cell-specific ablation of Raf-1 kinase gene (RIPCre(+/+):Raf-1(flox/flox)) and a complete set of littermate controls (RIPCre(+/+):Raf-1(wt/wt)). RIPCre(+/+):Raf-1(flox/flox) mice were viable, and no effects on weight gain were observed. RIPCre(+/+):Raf-1(flox/flox) mice had increased fasting blood glucose levels and impaired glucose tolerance but normal insulin tolerance compared to littermate controls. Insulin secretion in vivo and in isolated islets was markedly impaired, but there was no apparent effect on the exocytosis machinery. However, islet insulin protein and insulin 2 mRNA, but not insulin 1 mRNA, were dramatically reduced in Raf-1-knockout mice. Analysis of insulin 2 knockout mice demonstrated that this reduction in mRNA was sufficient to impair in vivo insulin secretion. Our data further indicate that Raf-1 specifically and acutely regulates insulin 2 mRNA via negative action on Foxo1, which has been shown to selectively control the insulin 2 gene. This work provides the first direct evidence that Raf-1 signaling is essential for the regulation of basal insulin transcription and the supply of releasable insulin in vivo.
Collapse
Affiliation(s)
- Emilyn U Alejandro
- Laboratory of Molecular Signalling in Diabetes, Diabetes Research Group, Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Øzbay LA, Smidt K, Mortensen DM, Carstens J, Jørgensen KA, Rungby J. Cyclosporin and tacrolimus impair insulin secretion and transcriptional regulation in INS-1E beta-cells. Br J Pharmacol 2011; 162:136-46. [PMID: 20825407 DOI: 10.1111/j.1476-5381.2010.01018.x] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND AND PURPOSE Introducing the calcineurin inhibitors cyclosporin (CsA) and tacrolimus (Tac) has improved the outcome of organ transplants, but complications such as new onset diabetes mellitus after transplantation (NODAT) decrease survival rates. EXPERIMENTAL APPROACH We sought, in a beta-cell culture model, to elucidate the pathogenic mechanisms behind NODAT and the relative contribution of the calcineurin inhibitors. INS-1E cells were incubated at basal and stimulatory glucose concentrations, while exposed to pharmacologically relevant doses of CsA, Tac and vehicle for 6 or 24 h. RESULTS Tac inhibited basal (P < 0.05), but not glucose-stimulated insulin secretion (GSIS) after 6 h of exposure. After 24 h, both agents inhibited basal and GSIS (P < 0.05). Calcineurin phosphatase activity was decreased by both drugs during all conditions. Apoptosis was only seen with CsA treatment, which also induced a slight suppression of calcineurin and insulin mRNA, as well as increased levels of the sterol receptor element binding protein (SREBP)-1c, a transcription factor thought to suppress genes essential for beta-cell function and induce insulin resistance. Expression levels of nuclear factor of activated T-cells (NFAT)-c1, -c2, -c3 and -c4 were not decreased notably by either drug. CONCLUSIONS AND IMPLICATIONS Tac had acute inhibitory effects on basal insulin secretion, but prolonged exposure (24 h) to Tac or CsA revealed similar suppression of insulin secretion. These prolonged effects were mirrored by a total inhibition of calcineurin activity in beta-cells. CsA showed greater inhibition of beta-cell survival and transcriptional markers, essential for beta-cell function.
Collapse
Affiliation(s)
- L A Øzbay
- Department of Nephrology, Aarhus University Hospital, Skejby, Aarhus, Denmark.
| | | | | | | | | | | |
Collapse
|