1
|
Yamada Y, Urakawa N, Tamiya H, Sakamoto S, Takahashi H, Harada N, Kitakaze T, Izawa T, Matsumua S, Yoshihara E, Inui H, Mashimo T, Yamaji R. Nrf2- and p53-inducible REDD2/DDiT4L/Rtp801L confers pancreatic β-cell dysfunction, leading to glucose intolerance in high-fat diet-fed mice. J Biol Chem 2025:110271. [PMID: 40409543 DOI: 10.1016/j.jbc.2025.110271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 05/07/2025] [Accepted: 05/11/2025] [Indexed: 05/25/2025] Open
Abstract
Pancreatic β-cells play a critical role in glucose homeostasis by secreting insulin. Chronic oxidative stress causes β-cell dysfunction, including β-cell loss; however, the underlying mechanisms remain unclear. Here, we demonstrate the critical role of the regulated in development and DNA damage response 2 (REDD2/DDiT4L/Rtp801L) in β-cell dysfunction. In INS-1 β-cells, Redd2 was induced by high glucose/palmitate or streptozotocin (STZ) exposure. Knockdown of Redd2 attenuated STZ-induced loss of cell viability, while REDD2 overexpression reduced cell viability and p70S6K phosphorylation, suggesting the involvement of suppression of mTORC1 activation. STZ also activated the transcription factors nuclear factor erythroid 2-related factor 2 (Nrf2) and p53, and overexpression of these transcription factors synergistically induced Redd2 expression. Reporter assays using the Redd2 promoter (-2328/-1) and chromatin immunoprecipitation identified the functional binding sites for Nrf2 (EpRE2, -349/-340) and p53 (p53RE1, -90/-81) on the Redd2 promoter. Purified recombinant p53 and Nrf2 bound directly. There were no noticeable changes in male global Redd2-knockout mice (C57BL/6J background), except for inguinal adipose tissue decrease when the mice were fed a standard diet. In contrast, when the mice were fed a high-fat diet (HFD), Redd2-knockout mice exhibited improved glucose tolerance relative to littermate controls. Redd2-knockout in HFD-fed mice increased β-cell mass due to reduced β-cell apoptosis and elevated plasma insulin concentrations, whereas insulin sensitivity remained unaffected. In both STZ-induced male and female and HFD-fed male models, β-cell specific Redd2-knockout improved glucose tolerance without affecting insulin sensitivity. Our results identify REDD2 as a novel regulator of β-cell dysfunction under oxidative stress.
Collapse
Affiliation(s)
- Yukiho Yamada
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Osaka Metropolitan University, Sakai, Osaka, Japan
| | - Natsuho Urakawa
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai, Osaka, Japan
| | - Hisato Tamiya
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai, Osaka, Japan
| | - Shuya Sakamoto
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai, Osaka, Japan
| | - Hiroki Takahashi
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Osaka Metropolitan University, Sakai, Osaka, Japan
| | - Naoki Harada
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Osaka Metropolitan University, Sakai, Osaka, Japan; Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai, Osaka, Japan; The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA.
| | - Tomoya Kitakaze
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Osaka Metropolitan University, Sakai, Osaka, Japan; Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai, Osaka, Japan
| | - Takeshi Izawa
- Graduate School of Veterinary Science, Osaka Metropolitan University, Izumisano, Osaka, Japan
| | - Shigenobu Matsumua
- Department of Nutrition, Graduate School of Human Life and Ecology, Osaka Metropolitan University, Habikino, Osaka, Japan
| | - Eiji Yoshihara
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA; David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, USA
| | - Hiroshi Inui
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Osaka Metropolitan University, Sakai, Osaka, Japan; Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai, Osaka, Japan; Department of Health and Nutrition, Otemae University, Osaka, Japan
| | - Tomoji Mashimo
- Division of Animal Genetics, Laboratory Animal Research Center, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo, Japan
| | - Ryoichi Yamaji
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Osaka Metropolitan University, Sakai, Osaka, Japan; Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai, Osaka, Japan; Center for Research and Development of Bioresources, Osaka Metropolitan University, Sakai, Osaka, Japan
| |
Collapse
|
2
|
Donis R, Patel KA, Wakeling MN, Johnson MB, Amoli MM, Yildiz M, Akçay T, Aspi I, Yong J, Yaghootkar H, Weedon MN, Hattersley AT, Flanagan SE, De Franco E. A homozygous TARS2 variant is a novel cause of syndromic neonatal diabetes. Diabet Med 2025; 42:e15471. [PMID: 39509107 PMCID: PMC11823299 DOI: 10.1111/dme.15471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 10/21/2024] [Accepted: 10/23/2024] [Indexed: 11/15/2024]
Abstract
AIMS Neonatal diabetes is a monogenic condition which can be the presenting feature of complex syndromes. The aim of this study was to identify novel genetic causes of neonatal diabetes with neurological features including developmental delay and epilepsy. METHODS We performed genome sequencing in 27 individuals with neonatal diabetes plus epilepsy and/or developmental delay of unknown genetic cause. Replication studies were performed in 123 individuals with diabetes diagnosed aged ≤1 year without a known genetic cause using targeted next-generation sequencing. RESULTS Three individuals, all diagnosed with diabetes in the first week of life, shared a rare homozygous missense variant, p.(Arg327Gln), in TARS2. Replication studies identified the same homozygous variant in a fourth individual diagnosed with diabetes at 1 year. One proband had epilepsy, one had development delay and two had both. Biallelic TARS2 variants cause a mitochondrial encephalopathy (COXPD-21) characterised by severe hypotonia, epilepsy and developmental delay. Diabetes is not a known feature of COXPD-21. Current evidence suggests that the p.(Arg327Gln) variant disrupts TARS2's regulation of the mTORC1 pathway which is essential for β-cells. CONCLUSIONS Our findings establish the homozygous p.(Arg327Gln) TARS2 variant as a novel cause of syndromic neonatal diabetes and uncover a role for TARS2 in pancreatic β-cells.
Collapse
Affiliation(s)
- Russell Donis
- Department of Clinical and Biomedical ScienceUniversity of Exeter Faculty of Health and Life SciencesExeterUK
| | - Kashyap A. Patel
- Department of Clinical and Biomedical ScienceUniversity of Exeter Faculty of Health and Life SciencesExeterUK
| | - Matthew N. Wakeling
- Department of Clinical and Biomedical ScienceUniversity of Exeter Faculty of Health and Life SciencesExeterUK
| | - Matthew B. Johnson
- Department of Clinical and Biomedical ScienceUniversity of Exeter Faculty of Health and Life SciencesExeterUK
| | - Masha M. Amoli
- Metabolic Disorders Research Centre, Endocrinology and Metabolism Molecular‐Cellular Sciences InstituteTehran University of Medical SciencesTehranIran
| | - Melek Yildiz
- Department of Paediatric Endocrinology, İstanbul University, İstanbul Faculty of MedicineİstanbulTurkey
| | - Teoman Akçay
- Department of Paediatric EndocrinologyBakırköy Dr. Sadi Konuk Education and Research HospitalİstanbulTurkey
| | - Irani Aspi
- Nanavati Super Speciality HospitalMumbaiIndia
- Juvenile Diabetes Foundation, Maharashtra ChapterMumbaiIndia
| | - James Yong
- Children and Young People's Diabetes TeamSt James's University HospitalLeedsUK
| | - Hanieh Yaghootkar
- College of Health and ScienceUniversity of Lincoln, Joseph Banks LaboratoriesLincolnUK
| | - Michael N. Weedon
- Department of Clinical and Biomedical ScienceUniversity of Exeter Faculty of Health and Life SciencesExeterUK
| | - Andrew T. Hattersley
- Department of Clinical and Biomedical ScienceUniversity of Exeter Faculty of Health and Life SciencesExeterUK
| | - Sarah E. Flanagan
- Department of Clinical and Biomedical ScienceUniversity of Exeter Faculty of Health and Life SciencesExeterUK
| | - Elisa De Franco
- Department of Clinical and Biomedical ScienceUniversity of Exeter Faculty of Health and Life SciencesExeterUK
| |
Collapse
|
3
|
Popa AD, Gherasim A, Mihalache L, Arhire LI, Graur M, Niță O. Fasting Mimicking Diet for Metabolic Syndrome: A Narrative Review of Human Studies. Metabolites 2025; 15:150. [PMID: 40137116 PMCID: PMC11943686 DOI: 10.3390/metabo15030150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 02/19/2025] [Accepted: 02/21/2025] [Indexed: 03/27/2025] Open
Abstract
Metabolic syndrome (MetS) is an association of risk factors that share insulin resistance (IR), exerting a super cumulative effect on the risk of developing cardiometabolic diseases. Lifestyle optimization is a key element in the prevention and non-pharmacological therapy of MetS. Certain studies have concluded that some dietary patterns could be more beneficial as an adjunctive treatment for MetS. Fasting mimicking diet (FMD) is a form of periodic fasting in which caloric intake is restricted for 5 days each month. It has been studied for its beneficial effects not only in patients with neoplasia and neurodegenerative diseases but also for its effects on IR and metabolism. In this narrative review, the effects of FMD in patients with MetS were analyzed, focusing on its impact on key metabolic components and summarizing findings from human studies. FMD has demonstrated beneficial effects on MetS by reducing BMI and waist circumference, preserving lean mass, and improving the metabolic profile. Moreover, individuals with a higher BMI or a greater number of MetS components appear to derive greater benefits from this intervention. However, limitations such as high dropout rates, small sample sizes, and methodological constraints restrict the generalizability of current findings. Further large-scale studies are needed to confirm these effects and establish FMD as a viable non-pharmacological strategy for managing MetS.
Collapse
Affiliation(s)
- Alina Delia Popa
- Internal Medicine II Department, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania; (A.D.P.); (L.M.); (L.I.A.); (O.N.)
| | - Andreea Gherasim
- Internal Medicine II Department, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania; (A.D.P.); (L.M.); (L.I.A.); (O.N.)
| | - Laura Mihalache
- Internal Medicine II Department, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania; (A.D.P.); (L.M.); (L.I.A.); (O.N.)
| | - Lidia Iuliana Arhire
- Internal Medicine II Department, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania; (A.D.P.); (L.M.); (L.I.A.); (O.N.)
| | - Mariana Graur
- Faculty of Medicine and Biological Sciences, University “Ștefan cel Mare” of Suceava, 720229 Suceava, Romania;
| | - Otilia Niță
- Internal Medicine II Department, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania; (A.D.P.); (L.M.); (L.I.A.); (O.N.)
| |
Collapse
|
4
|
Wu Y, Wang H, Xu H. Autophagy-lysosome pathway in insulin & glucagon homeostasis. Front Endocrinol (Lausanne) 2025; 16:1541794. [PMID: 39996055 PMCID: PMC11847700 DOI: 10.3389/fendo.2025.1541794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Accepted: 01/22/2025] [Indexed: 02/26/2025] Open
Abstract
Lysosome, a highly dynamic organelle, is an important nutrient sensing center. They utilize different ion channels and transporters to complete the mission in degradation, trafficking, nutrient sensing and integration of various metabolic pathways to maintain cellular homeostasis. Glucose homeostasis relies on tightly regulated insulin secretion by pancreatic β cells, and their dysfunction is a hallmark of type 2 diabetes. Glucagon also plays an important role in hyperglycemia in diabetic patients. Currently, lysosome has been recognized as a nutrient hub to regulate the homeostasis of insulin and other hormones. In this review, we will discuss recent advances in understanding lysosome-mediated autophagy and lysosomal proteins involved in maintaining insulin and glucagon homeostasis, as well as their contributions to the etiology of diabetes.
Collapse
Affiliation(s)
- Yi Wu
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, China
- Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
- Shanghai Key Laboratory of Molecular Imaging, School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Hui Wang
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Huoyan Xu
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, China
| |
Collapse
|
5
|
Kong X, Yan D, Shao L, Li B, Lv S, Tu Y, Zhang Y, Shu X, Ying Y, Ma X. Increased COX6A2 Promotes Pancreatic β-Cell Apoptosis and Is Suppressed in Diabetic GK Rats After Roux-en-Y Gastric Bypass. Diabetes 2025; 74:175-187. [PMID: 39546664 DOI: 10.2337/db24-0301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 11/01/2024] [Indexed: 11/17/2024]
Abstract
ARTICLE HIGHLIGHTS Cytochrome c oxidase subunit 6A2 (COX6A2) expression is increased in diabetic islets. Increased COX6A2 promotes β-cell apoptosis via modulation of cyclophilin D-mediated cytochrome c release from mitochondria to the cytoplasm. Carbohydrate-responsive element-binding protein epigenetically regulates COX6A2 expression in β-cells. Roux-en-Y gastric bypass reduces COX6A2 expression by regulating the glucagon-like peptide 1/cAMP-dependent protein kinase/carbohydrate-responsive element-binding protein signaling pathway.
Collapse
Affiliation(s)
- Xiangchen Kong
- Shenzhen University Diabetes Institute, Shenzhen Key Laboratory of Metabolism and Cardiovascular Homeostasis, Shenzhen University Medical School, Shenzhen, China
| | - Dan Yan
- Shenzhen University Diabetes Institute, Shenzhen Key Laboratory of Metabolism and Cardiovascular Homeostasis, Shenzhen University Medical School, Shenzhen, China
| | - Lianqi Shao
- Shenzhen University Diabetes Institute, Shenzhen Key Laboratory of Metabolism and Cardiovascular Homeostasis, Shenzhen University Medical School, Shenzhen, China
| | - Bingfeng Li
- Shenzhen University Diabetes Institute, Shenzhen Key Laboratory of Metabolism and Cardiovascular Homeostasis, Shenzhen University Medical School, Shenzhen, China
- Department of Surgery, Li Kai Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Simian Lv
- Shenzhen University Diabetes Institute, Shenzhen Key Laboratory of Metabolism and Cardiovascular Homeostasis, Shenzhen University Medical School, Shenzhen, China
- School of Basic Medicine, Jiamusi University, Jiamusi, China
| | - Yifan Tu
- Shenzhen University Diabetes Institute, Shenzhen Key Laboratory of Metabolism and Cardiovascular Homeostasis, Shenzhen University Medical School, Shenzhen, China
| | - Yingqi Zhang
- Shenzhen University Diabetes Institute, Shenzhen Key Laboratory of Metabolism and Cardiovascular Homeostasis, Shenzhen University Medical School, Shenzhen, China
| | - Xingsheng Shu
- Shenzhen University Diabetes Institute, Shenzhen Key Laboratory of Metabolism and Cardiovascular Homeostasis, Shenzhen University Medical School, Shenzhen, China
| | - Ying Ying
- Shenzhen University Diabetes Institute, Shenzhen Key Laboratory of Metabolism and Cardiovascular Homeostasis, Shenzhen University Medical School, Shenzhen, China
| | - Xiaosong Ma
- Shenzhen University Diabetes Institute, Shenzhen Key Laboratory of Metabolism and Cardiovascular Homeostasis, Shenzhen University Medical School, Shenzhen, China
| |
Collapse
|
6
|
Chau GC, Lim JE, Moon K, Kim IS, Um SH. The stimulatory effect of HI 129, a novel indole derivative, on glucose-induced insulin secretion. Biochem Pharmacol 2024; 230:116558. [PMID: 39326678 DOI: 10.1016/j.bcp.2024.116558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 08/29/2024] [Accepted: 09/24/2024] [Indexed: 09/28/2024]
Abstract
Indole derivatives exhibit a broad spectrum of beneficial effects, encompassing anti-inflammatory, antiviral, antimalarial, anti-diabetic, antioxidant, anti-hepatitis, and antidepressant properties. Here, we describe the potentiation of insulin secretion in pancreatic islets and INS-1 cells through methyl 2-(2-ethoxy-1-hydroxy-2-oxoethyl)-1-(pyrimidine-2-yl)-1H-indole-3-carboxylate (HI 129), a novel indole derivative. Treatment with HI 129 led to notably decreased ADP/ATP ratios in pancreatic islets and INS-1 cells compared to those in the vehicle-treated controls, indicating a shift in cellular ATP production. Moreover, the augmentation of insulin secretion by HI 129 was closely correlated with its ability to enhance the mitochondrial membrane potential and respiration, partly by reducing the phosphorylation levels of AMP-activated protein kinase (AMPK). Mechanistically, HI 129 enhanced the association between AMPK and β-arrestin-1, critical molecules for glucose-induced insulin secretion. Furthermore, β-arrestin-1 depletion attenuated the effect of HI 129 on glucose-induced insulin secretion, suggesting that HI 129 potentiates insulin secretion via β-arrestin-1/AMPK signaling. These results collectively underscore the potential of HI 129 in enhancing insulin secretion as a novel candidate for improving glucose homeostasis in type 2 diabetes.
Collapse
Affiliation(s)
- Gia Cac Chau
- Department of Molecular Cell Biology, School of Medicine, Sungkyunkwan University, Suwon, Gyeonggi-do 16419, Korea
| | - Ji Eun Lim
- Department of Molecular Cell Biology, School of Medicine, Sungkyunkwan University, Suwon, Gyeonggi-do 16419, Korea
| | - Kyeongwon Moon
- School of Pharmacy, Sungkyunkwan University, Suwon, Gyeonggi-do 16419, Korea
| | - In Su Kim
- School of Pharmacy, Sungkyunkwan University, Suwon, Gyeonggi-do 16419, Korea.
| | - Sung Hee Um
- Department of Molecular Cell Biology, School of Medicine, Sungkyunkwan University, Suwon, Gyeonggi-do 16419, Korea; Biomedical Institute Convergence at Sungkyunkwan University, School of Medicine, Sungkyunkwan University, Suwon, Gyeonggi-do 16419, Korea; Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Samsung Medical Center, Sungkyunkwan University, Seoul, 06351, Korea.
| |
Collapse
|
7
|
Lewis CV, Garcia AM, Burciaga SD, Posey JN, Jordan M, Nguyen TTN, Stenmark KR, Mickael C, Sul C, Delaney C, Nozik ES. Redistribution of SOD3 expression due to R213G polymorphism affects pulmonary interstitial macrophage reprogramming in response to hypoxia. Physiol Genomics 2024; 56:776-790. [PMID: 39311838 PMCID: PMC11573264 DOI: 10.1152/physiolgenomics.00078.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 08/14/2024] [Accepted: 09/11/2024] [Indexed: 10/25/2024] Open
Abstract
The extracellular isoform of superoxide dismutase (SOD3) is decreased in patients and animals with pulmonary hypertension (PH). The human R213G single-nucleotide polymorphism (SNP) in SOD3 causes its release from tissue extracellular matrix (ECM) into extracellular fluids, without modulating enzyme activity, increasing cardiovascular disease risk in humans and exacerbating chronic hypoxic PH in mice. Given the importance of interstitial macrophages (IMs) to PH pathogenesis, this study aimed to determine whether R213G SOD3 increases IM accumulation and alters IM reprogramming in response to hypoxia. R213G mice and wild-type (WT) controls were exposed to hypobaric hypoxia for 4 or 14 days compared with normoxia. Flow cytometry demonstrated a transient increase in IMs at day 4 in both strains. Contrary to our hypothesis, the R213G SNP did not augment IM accumulation. To determine strain differences in the IM reprogramming response to hypoxia, we performed RNAsequencing on IMs isolated at each timepoint. We found that IMs from R213G mice exposed to hypoxia activated ECM-related pathways and a combination of alternative macrophage and proinflammatory signaling. Furthermore, when compared with WT responses, IMs from R213G mice lacked metabolic remodeling and demonstrated a blunted anti-inflammatory response between the early (day 4) and later (day 14) timepoints. We confirmed metabolic responses using Agilent Seahorse assays, whereby WT, but not R213G, IMs upregulated glycolysis at day 4 that returned to baseline at day 14. Finally, we identify differential regulation of several redox-sensitive upstream regulators that could be investigated in future studies.NEW & NOTEWORTHY Redistributed expression of SOD3 out of tissue ECM due to the human R213G SNP exacerbates chronic hypoxic PH. Highlighting the importance of macrophage phenotype, our findings reveal that the R213G SNP does not exacerbate pulmonary macrophage accumulation in response to hypoxia but influences their metabolic and phenotypic reprogramming. We demonstrate a deficiency in the metabolic response to hypoxic stress in R213G macrophages, associated with weakened inflammatory resolution and activation of profibrotic pathways implicated in PH.
Collapse
Affiliation(s)
- Caitlin V Lewis
- Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Section of Pediatric Critical Care Medicine, Department of Pediatrics, Children's Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Anastacia M Garcia
- Division of Cardiology, Department of Pediatrics, Children's Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Samuel D Burciaga
- Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Section of Pediatric Critical Care Medicine, Department of Pediatrics, Children's Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Janelle N Posey
- Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Section of Neonatology, Department of Pediatrics, Division of Cardiology, Children's Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Mariah Jordan
- Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Section of Neonatology, Department of Pediatrics, Division of Cardiology, Children's Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Thi-Tina N Nguyen
- Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Section of Pediatric Critical Care Medicine, Department of Pediatrics, Children's Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Kurt R Stenmark
- Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Section of Pediatric Critical Care Medicine, Department of Pediatrics, Children's Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Claudia Mickael
- Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Section of Pediatric Critical Care Medicine, Department of Pediatrics, Children's Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Christina Sul
- Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Section of Pediatric Critical Care Medicine, Department of Pediatrics, Children's Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Cassidy Delaney
- Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Section of Neonatology, Department of Pediatrics, Division of Cardiology, Children's Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Eva S Nozik
- Cardiovascular Pulmonary Research Laboratories, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Section of Pediatric Critical Care Medicine, Department of Pediatrics, Children's Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| |
Collapse
|
8
|
Wang P, Xiao H, Wu T, Fu Q, Song X, Zhao Y, Li Y, Huang J, Song Z. Activation of skeletal carbohydrate-response element binding protein (ChREBP)-mediated de novo lipogenesis increases intramuscular fat content in chickens. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2024; 18:107-118. [PMID: 39091296 PMCID: PMC11292260 DOI: 10.1016/j.aninu.2024.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 04/10/2024] [Accepted: 04/10/2024] [Indexed: 08/04/2024]
Abstract
The intracellular lipids in muscle cells of farm animals play a crucial role in determining the overall intramuscular fat (IMF) content, which has a positive impact on meat quality. However, the mechanisms underlying the deposition of lipids in muscle cells of farm animals are not yet fully understood. The purpose of this study was to determine the roles of carbohydrate-response element binding protein (ChREBP) and fructose in IMF deposition of chickens. For virus-mediated ChREBP overexpression in tibialis anterior (TA) muscle of chickens, seven 5-d-old male yellow-feather chickens were used. At 10 d after virus injection, the chickens were slaughtered to obtain TA muscles for analysis. For fructose administration trial, sixty 9-wk-old male yellow-feather chickens were randomly divided into 2 groups, with 6 replicates per group and 5 chickens per replicate. The chickens were fed either a basal diet or a basal diet supplemented with 10% fructose (purity ≥ 99%). At 4 wk later, the chickens were slaughtered, and breast and thigh muscles were collected for analysis. The results showed that the skeletal ChREBP mRNA levels were positively associated with IMF content in multiple species, including the chickens, pigs, and mice (P < 0.05). ChREBP overexpression increased lipid accumulation in both muscle cells in vitro and the TA muscles of mice and chickens in vivo (P < 0.05), by activation of the de novo lipogenesis (DNL) pathway. Moreover, activation of ChREBP by dietary fructose administration also resulted in increased IMF content in mice and notably chickens (P < 0.05). Furthermore, the lipidomics analysis revealed that ChREBP activation altered the lipid composition of chicken IMF and tented to improve the flavor profile of the meat. In conclusion, this study found that ChREBP plays a pivotal role in mediating the deposition of fat in chicken muscles in response to fructose-rich diets, which provides a novel strategy for improving meat quality in the livestock industry.
Collapse
Affiliation(s)
- Peng Wang
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning, Guangxi 530004, China
| | - Haihan Xiao
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning, Guangxi 530004, China
| | - Tian Wu
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning, Guangxi 530004, China
| | - Qinghua Fu
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning, Guangxi 530004, China
| | - Xudong Song
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning, Guangxi 530004, China
| | - Yameng Zhao
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning, Guangxi 530004, China
| | - Yan Li
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning, Guangxi 530004, China
| | - Jieping Huang
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning, Guangxi 530004, China
| | - Ziyi Song
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning, Guangxi 530004, China
| |
Collapse
|
9
|
Liu X, Guo B, Li Q, Nie J. mTOR in metabolic homeostasis and disease. Exp Cell Res 2024; 441:114173. [PMID: 39047807 DOI: 10.1016/j.yexcr.2024.114173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 07/20/2024] [Accepted: 07/21/2024] [Indexed: 07/27/2024]
Abstract
The ability to maintain cellular metabolic homeostasis is critical to life, in which mTOR plays an important role. This kinase integrates upstream nutrient signals and performs essential functions in physiology and metabolism by increasing metabolism and suppressing autophagy. Thus, dysregulation of mTOR activity leads to diseases, especially metabolic diseases such as cancer, type 2 diabetes and neurological disorders. Therefore, inhibition of overactivated mTOR becomes a rational approach to treat a variety of metabolic diseases. In this review, we discuss how mTOR responds to upstream signals and how mTOR regulates metabolic processes, including protein, nucleic acid, and lipid metabolism. Furthermore, we discuss the possible causes and consequences of dysregulated mTOR signaling activity, and summarize relevant applications, such as inhibition of mTOR activity to treat these diseases. This review will advance our comprehensive knowledge of the association between mTOR and metabolic homeostasis, which has significant ramifications for human health.
Collapse
Affiliation(s)
- Xuejia Liu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Bin Guo
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Qiye Li
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Jing Nie
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China.
| |
Collapse
|
10
|
Roohi TF, Faizan S, Parray ZA, Baig MDAI, Mehdi S, Kinattingal N, Krishna KL. Beyond Glucose: The Dual Assault of Oxidative and ER Stress in Diabetic Disorders. High Blood Press Cardiovasc Prev 2023; 30:513-531. [PMID: 38041772 DOI: 10.1007/s40292-023-00611-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 11/15/2023] [Indexed: 12/03/2023] Open
Abstract
Diabetes mellitus, a prevalent global health concern, is characterized by hyperglycemia. However, recent research reveals a more intricate landscape where oxidative stress and endoplasmic reticulum (ER) stress orchestrate a dual assault, profoundly impacting diabetic disorders. This review elucidates the interplay between these two stress pathways and their collective consequences on diabetes. Oxidative stress emanates from mitochondria, where reactive oxygen species (ROS) production spirals out of control, leading to cellular damage. We explore ROS-mediated signaling pathways, which trigger β-cell dysfunction, insulin resistance, and endothelial dysfunction the quintessential features of diabetes. Simultaneously, ER stress unravels, unveiling how protein folding disturbances activate the unfolded protein response (UPR). We dissect the UPR's dual role, oscillating between cellular adaptation and apoptosis, significantly influencing pancreatic β-cells and peripheral insulin-sensitive tissues. Crucially, this review exposes the synergy between oxidative and ER stress pathways. ROS-induced UPR activation and ER stress-induced oxidative stress create a detrimental feedback loop, exacerbating diabetic complications. Moreover, we spotlight promising therapeutic strategies that target both stress pathways. Antioxidants, molecular chaperones, and novel pharmacological agents offer potential avenues for diabetes management. As the global diabetes burden escalates, comprehending the dual assault of oxidative and ER stress is paramount. This review not only unveils the intricate molecular mechanisms governing diabetic pathophysiology but also advocates a holistic therapeutic approach. By addressing both stress pathways concurrently, we may forge innovative solutions for diabetic disorders, ultimately alleviating the burden of this pervasive health issue.
Collapse
Affiliation(s)
- Tamsheel Fatima Roohi
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysore, Karnataka, 570015, India
| | - Syed Faizan
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysore, Karnataka, 570015, India
| | - Zahoor Ahmad Parray
- Department of Chemistry, Indian Institute of Technology (IIT) Delhi, Hauz Khas Campus, New Delhi, 110016, India
| | - M D Awaise Iqbal Baig
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysore, Karnataka, 570015, India
| | - Seema Mehdi
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysore, Karnataka, 570015, India
| | - Nabeel Kinattingal
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysore, Karnataka, 570015, India
| | - K L Krishna
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysore, Karnataka, 570015, India.
| |
Collapse
|
11
|
Jomova K, Raptova R, Alomar SY, Alwasel SH, Nepovimova E, Kuca K, Valko M. Reactive oxygen species, toxicity, oxidative stress, and antioxidants: chronic diseases and aging. Arch Toxicol 2023; 97:2499-2574. [PMID: 37597078 PMCID: PMC10475008 DOI: 10.1007/s00204-023-03562-9] [Citation(s) in RCA: 651] [Impact Index Per Article: 325.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 07/24/2023] [Indexed: 08/21/2023]
Abstract
A physiological level of oxygen/nitrogen free radicals and non-radical reactive species (collectively known as ROS/RNS) is termed oxidative eustress or "good stress" and is characterized by low to mild levels of oxidants involved in the regulation of various biochemical transformations such as carboxylation, hydroxylation, peroxidation, or modulation of signal transduction pathways such as Nuclear factor-κB (NF-κB), Mitogen-activated protein kinase (MAPK) cascade, phosphoinositide-3-kinase, nuclear factor erythroid 2-related factor 2 (Nrf2) and other processes. Increased levels of ROS/RNS, generated from both endogenous (mitochondria, NADPH oxidases) and/or exogenous sources (radiation, certain drugs, foods, cigarette smoking, pollution) result in a harmful condition termed oxidative stress ("bad stress"). Although it is widely accepted, that many chronic diseases are multifactorial in origin, they share oxidative stress as a common denominator. Here we review the importance of oxidative stress and the mechanisms through which oxidative stress contributes to the pathological states of an organism. Attention is focused on the chemistry of ROS and RNS (e.g. superoxide radical, hydrogen peroxide, hydroxyl radicals, peroxyl radicals, nitric oxide, peroxynitrite), and their role in oxidative damage of DNA, proteins, and membrane lipids. Quantitative and qualitative assessment of oxidative stress biomarkers is also discussed. Oxidative stress contributes to the pathology of cancer, cardiovascular diseases, diabetes, neurological disorders (Alzheimer's and Parkinson's diseases, Down syndrome), psychiatric diseases (depression, schizophrenia, bipolar disorder), renal disease, lung disease (chronic pulmonary obstruction, lung cancer), and aging. The concerted action of antioxidants to ameliorate the harmful effect of oxidative stress is achieved by antioxidant enzymes (Superoxide dismutases-SODs, catalase, glutathione peroxidase-GPx), and small molecular weight antioxidants (vitamins C and E, flavonoids, carotenoids, melatonin, ergothioneine, and others). Perhaps one of the most effective low molecular weight antioxidants is vitamin E, the first line of defense against the peroxidation of lipids. A promising approach appears to be the use of certain antioxidants (e.g. flavonoids), showing weak prooxidant properties that may boost cellular antioxidant systems and thus act as preventive anticancer agents. Redox metal-based enzyme mimetic compounds as potential pharmaceutical interventions and sirtuins as promising therapeutic targets for age-related diseases and anti-aging strategies are discussed.
Collapse
Affiliation(s)
- Klaudia Jomova
- Department of Chemistry, Faculty of Natural Sciences, Constantine the Philosopher University in Nitra, Nitra, 949 74, Slovakia
| | - Renata Raptova
- Faculty of Chemical and Food Technology, Slovak University of Technology, Bratislava, 812 37, Slovakia
| | - Suliman Y Alomar
- Zoology Department, College of Science, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Saleh H Alwasel
- Zoology Department, College of Science, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Sciences, University of Hradec Kralove, 50005, Hradec Kralove, Czech Republic
| | - Kamil Kuca
- Department of Chemistry, Faculty of Sciences, University of Hradec Kralove, 50005, Hradec Kralove, Czech Republic
| | - Marian Valko
- Faculty of Chemical and Food Technology, Slovak University of Technology, Bratislava, 812 37, Slovakia.
| |
Collapse
|
12
|
Choi EH, Park SJ. TXNIP: A key protein in the cellular stress response pathway and a potential therapeutic target. Exp Mol Med 2023; 55:1348-1356. [PMID: 37394581 PMCID: PMC10393958 DOI: 10.1038/s12276-023-01019-8] [Citation(s) in RCA: 101] [Impact Index Per Article: 50.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/19/2023] [Accepted: 03/21/2023] [Indexed: 07/04/2023] Open
Abstract
Thioredoxin-interacting protein (TXNIP), which is also known as thioredoxin-binding protein 2 (TBP2), directly interacts with the major antioxidant protein thioredoxin (TRX) and inhibits its antioxidant function and expression. However, recent studies have demonstrated that TXNIP is a multifunctional protein with functions beyond increasing intracellular oxidative stress. TXNIP activates endoplasmic reticulum (ER) stress-mediated nucleotide-binding oligomerization domain (NOD)-like receptor protein-3 (NLRP3) inflammasome complex formation, triggers mitochondrial stress-induced apoptosis, and stimulates inflammatory cell death (pyroptosis). These newly discovered functions of TXNIP highlight its role in disease development, especially in response to several cellular stress factors. In this review, we provide an overview of the multiple functions of TXNIP in pathological conditions and summarize its involvement in various diseases, such as diabetes, chronic kidney disease, and neurodegenerative diseases. We also discuss the potential of TXNIP as a therapeutic target and TXNIP inhibitors as novel therapeutic drugs for treating these diseases.
Collapse
Affiliation(s)
- Eui-Hwan Choi
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu, 41061, South Korea
| | - Sun-Ji Park
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu, 41061, South Korea.
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|
13
|
Chang X, Tian C, Jia Y, Cai Y, Yan P. MLXIPL promotes the migration, invasion, and glycolysis of hepatocellular carcinoma cells by phosphorylation of mTOR. BMC Cancer 2023; 23:176. [PMID: 36809979 PMCID: PMC9945719 DOI: 10.1186/s12885-023-10652-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 02/15/2023] [Indexed: 02/23/2023] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is associated with a high occurrence, mortality, and poor prognosis. MLX interacting protein like (MLXIPL) is an important regulator of glucolipid metabolism and is involved in tumor progression. We aimed to clarify the role of MLXIPL in HCC and its underlying mechanisms. METHODS The level of MLXIPL was predicted using bioinformatic analysis and verified using quantitative real-time PCR (qPCR), immunohistochemical analysis, and western blot. We assessed the effects of MLXIPL on biological behaviors using the cell counting kit-8, colony formation, and Transwell assay. Glycolysis was evaluated using the Seahorse method. The interaction between MLXIPL and mechanistic target of rapamycin kinase (mTOR) was confirmed using RNA immunoprecipitation and co-immunoprecipitation. mTOR expression was detected in HCC cells using qPCR, immunofluorescence analysis, and western blot. RESULTS The results showed that MLXIPL levels were elevated in both HCC tissues and HCC cell lines. Knockdown of MLXIPL impeded HCC cell growth, invasion, migration, and glycolysis. Moreover, MLXIPL combined with mTOR to induce phosphorylation of mTOR. Activated mTOR abrogated the effects on cellular processes induced by MLXIPL. CONCLUSION MLXIPL promoted the malignant progression of HCC by activating phosphorylation of mTOR, suggesting an important role of the combination of MLXIPL and mTOR in HCC.
Collapse
Affiliation(s)
- Xiaowei Chang
- grid.508540.c0000 0004 4914 235XDepartment of General Surgery, The First Affiliated Hospital of Xi’an Medical University, No. 48, Fenghao West Road, Lianhu District, 710077 Xi’an, Shaanxi China
| | - Chang Tian
- grid.508540.c0000 0004 4914 235XDepartment of Clinical Laboratory, The First Affiliated Hospital of Xi’an Medical University, Xi’an, Shaanxi China
| | - Yuanyuan Jia
- grid.508540.c0000 0004 4914 235XDepartment of Faculty Development and Teaching Evaluation Office, The First Affiliated Hospital of Xi’an Medical University, Xi’an, Shaanxi China
| | - Yu Cai
- grid.508540.c0000 0004 4914 235XDepartment of General Surgery, The First Affiliated Hospital of Xi’an Medical University, No. 48, Fenghao West Road, Lianhu District, 710077 Xi’an, Shaanxi China
| | - Pu Yan
- Department of General Surgery, The First Affiliated Hospital of Xi'an Medical University, No. 48, Fenghao West Road, Lianhu District, 710077, Xi'an, Shaanxi, China.
| |
Collapse
|
14
|
Kim HJ, Kim DH, Um SH. The Novel Inhibitory Effect of YM976 on Adipocyte Differentiation. Cells 2023; 12:cells12020205. [PMID: 36672141 PMCID: PMC9856710 DOI: 10.3390/cells12020205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 12/25/2022] [Accepted: 12/29/2022] [Indexed: 01/05/2023] Open
Abstract
The pyrimidine derivative YM976 (4-(3-chlorophenyl)-1,7-diethylpyrido(2,3-d)-pyrimidin-2(1H)-one) exerts anti-inflammatory and anti-asthmatic effects. Considering that accumulation of lipids in adipose tissue is accompanied by inflammation, we investigated whether YM976 affects adipocyte differentiation. We found that YM976 significantly decreased lipid accumulation without cytotoxicity and reduced the expression levels of peroxisome proliferator-activated receptor γ (PPARγ) and CCAAT/enhancer-binding protein α (C/EBPα) as well as their lipogenic regulators including fatty acid synthase (FASN) and fatty acid-binding protein 4 (FABP4) in 3T3-L1 cells induced for differentiation. YM976 mainly inhibited the early stage of adipocyte differentiation. Furthermore, intracellular cAMP level was elevated by YM976 resulting in increased phosphorylation of adenosine monophosphate-activated protein kinase (AMPK). Conversely, decreasing the levels of AMPK or treatment with Compound C, an AMPK inhibitor, lessened the suppressive effects of YM976 on PPARγ transcriptional activity and adipogenesis. Thus, our results suggest YM976 as a novel potential compound for controlling lipid accumulation and formation of adipocytes in obesity.
Collapse
Affiliation(s)
- Hee Jung Kim
- Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon 16419, Republic of Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Samsung Medical Center, Sungkyunkwan University, Seoul 06351, Republic of Korea
| | - Dong-Hoon Kim
- Department of Pharmacology, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Sung Hee Um
- Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon 16419, Republic of Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Samsung Medical Center, Sungkyunkwan University, Seoul 06351, Republic of Korea
- Biomedical Institute for Convergence (BICS) at Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
15
|
Antioxidant Phytochemicals as Potential Therapy for Diabetic Complications. Antioxidants (Basel) 2023; 12:antiox12010123. [PMID: 36670985 PMCID: PMC9855127 DOI: 10.3390/antiox12010123] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/10/2022] [Accepted: 12/21/2022] [Indexed: 01/06/2023] Open
Abstract
The global prevalence of diabetes continues to increase partly due to rapid urbanization and an increase in the aging population. Consequently, this is associated with a parallel increase in the prevalence of diabetic vascular complications which significantly worsen the burden of diabetes. For these diabetic vascular complications, there is still an unmet need for safe and effective alternative/adjuvant therapeutic interventions. There is also an increasing urge for therapeutic options to come from natural products such as plants. Hyperglycemia-induced oxidative stress is central to the development of diabetes and diabetic complications. Furthermore, oxidative stress-induced inflammation and insulin resistance are central to endothelial damage and the progression of diabetic complications. Human and animal studies have shown that polyphenols could reduce oxidative stress, hyperglycemia, and prevent diabetic complications including diabetic retinopathy, diabetic nephropathy, and diabetic peripheral neuropathy. Part of the therapeutic effects of polyphenols is attributed to their modulatory effect on endogenous antioxidant systems. This review attempts to summarize the established effects of polyphenols on endogenous antioxidant systems from the literature. Moreover, potential therapeutic strategies for harnessing the potential benefits of polyphenols for diabetic vascular complications are also discussed.
Collapse
|
16
|
Lee S, Choi S, Park SH, Im GJ, Chang J. Transcriptomic Analysis of the Effect of Metformin against Cisplatin-Induced Ototoxicity: A Potential Mechanism of Metformin-Mediated Inhibition of Thioredoxin-Interacting Protein (Txnip) Gene Expression. Curr Issues Mol Biol 2022; 45:286-310. [PMID: 36661507 PMCID: PMC9857533 DOI: 10.3390/cimb45010021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/29/2022] [Accepted: 12/30/2022] [Indexed: 01/03/2023] Open
Abstract
Ototoxicity is the drug-induced damage of the inner ear, causing bilateral irreversible sensorineural hearing loss. Cisplatin is a widely used chemotherapeutic agent which causes ototoxicity as its side effect. Pretreatment with metformin prior to the application of cisplatin significantly decreased the late apoptosis and attenuated the cisplatin-induced increase in ROS. To understand the molecular mechanisms that are involved in the preventive effect of metformin, we evaluated the change of gene expression induced by cisplatin at several different time points (0 h, 6 h, 15 h, 24 h and 48 h) and the alteration of gene expression according to pretreatment with metformin in HEI-OC1 cells through microarray analysis. Cisplatin exposure induced a total of 89 DEGs (differentially expressed genes) after 6 h, with a total of 433 DEGs after 15 h, a total of 941 DEGs after 24 h, and a total of 2764 DEGs after 48 h. When cells were pretreated with metformin for 24 h, we identified a total of 105 DEGs after 6 h of cisplatin exposure, a total of 257 DEGs after 15 h, a total of 1450 DEGs after 24 h, and a total of 1463 DEGs after 48 h. The analysis was performed based on the gene expression, network analyses, and qRT-PCR, and we identified several genes (CSF2, FOS, JUN, TNFα, NFκB, Txnip, ASK1, TXN2, ATF3, TP53, IL6, and IGF1) as metformin-related preventive biomarkers in cisplatin ototoxicity.
Collapse
Affiliation(s)
- Sehee Lee
- Department of Otorhinolaryngology-Head & Neck Surgery, Hallym University College of Medicine, Kangnam Sacred Heart Hospital, Seoul 07441, Republic of Korea
- Department of Otorhinolaryngology-Head & Neck Surgery, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Sun Choi
- Department of Otorhinolaryngology-Head & Neck Surgery, Hallym University College of Medicine, Kangnam Sacred Heart Hospital, Seoul 07441, Republic of Korea
| | - Seok Hyun Park
- Department of Otorhinolaryngology-Head & Neck Surgery, Hallym University College of Medicine, Kangnam Sacred Heart Hospital, Seoul 07441, Republic of Korea
| | - Gi Jung Im
- Department of Otorhinolaryngology-Head & Neck Surgery, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Jiwon Chang
- Department of Otorhinolaryngology-Head & Neck Surgery, Hallym University College of Medicine, Kangnam Sacred Heart Hospital, Seoul 07441, Republic of Korea
- Correspondence: or ; Tel.: +82-2-6960-1270
| |
Collapse
|
17
|
Kant R, Manne RK, Anas M, Penugurti V, Chen T, Pan BS, Hsu CC, Lin HK. Deregulated transcription factors in cancer cell metabolisms and reprogramming. Semin Cancer Biol 2022; 86:1158-1174. [PMID: 36244530 PMCID: PMC11220368 DOI: 10.1016/j.semcancer.2022.10.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 09/10/2022] [Accepted: 10/11/2022] [Indexed: 01/27/2023]
Abstract
Metabolic reprogramming is an important cancer hallmark that plays a key role in cancer malignancies and therapy resistance. Cancer cells reprogram the metabolic pathways to generate not only energy and building blocks but also produce numerous key signaling metabolites to impact signaling and epigenetic/transcriptional regulation for cancer cell proliferation and survival. A deeper understanding of the mechanisms by which metabolic reprogramming is regulated in cancer may provide potential new strategies for cancer targeting. Recent studies suggest that deregulated transcription factors have been observed in various human cancers and significantly impact metabolism and signaling in cancer. In this review, we highlight the key transcription factors that are involved in metabolic control, dissect the crosstalk between signaling and transcription factors in metabolic reprogramming, and offer therapeutic strategies targeting deregulated transcription factors for cancer treatment.
Collapse
Affiliation(s)
- Rajni Kant
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston-Salem, NC 27101, USA
| | - Rajesh Kumar Manne
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston-Salem, NC 27101, USA
| | - Mohammad Anas
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston-Salem, NC 27101, USA
| | - Vasudevarao Penugurti
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston-Salem, NC 27101, USA
| | - Tingjin Chen
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston-Salem, NC 27101, USA
| | - Bo-Syong Pan
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston-Salem, NC 27101, USA
| | - Che-Chia Hsu
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston-Salem, NC 27101, USA
| | - Hui-Kuan Lin
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston-Salem, NC 27101, USA.
| |
Collapse
|
18
|
Kim HJ, Lee DE, Park EC, Ra MJ, Jung SM, Yu JN, Um SH, Kim KH. Anti-Adipogenic Effects of Salicortin from the Twigs of Weeping Willow (Salix pseudolasiogyne) in 3T3-L1 Cells. Molecules 2022; 27:molecules27206954. [PMID: 36296558 PMCID: PMC9609119 DOI: 10.3390/molecules27206954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 10/03/2022] [Accepted: 10/10/2022] [Indexed: 11/16/2022] Open
Abstract
Salix pseudolasiogyne (Salicaceae), the “weeping willow,” has been used in traditional Korean medicine to treat pain and fever due to its high concentrations of salicylic acid and salicin. The present study investigated bioactive compounds from S. pseudolasiogyne twigs to discover bioactive natural products. Phytochemical investigation of the ethanol (EtOH) extract of S. pseudolasiogyne twigs followed by liquid chromatography–mass spectrometry (LC/MS)-based analysis led to the isolation of two salicin derivatives, salicortinol and salicortin, the structures of which were determined by interpretation of their NMR spectra and data from the LC/MS analysis. To the best of our knowledge, this is the first report of salicortinol isolated from S. pseudolasiogyne. The isolated compounds were evaluated for their anti-adipogenic effects in 3T3-L1 cells. Both salicortinol and salicortin were found to significantly inhibit adipocyte differentiation in 3T3-L1 cells. In particular, salicortin exhibited a strong inhibitory effect on lipid accumulation. Furthermore, salicortin inhibited the expression of lipogenic and adipogenic transcription factors, including FASN, FABP4, C/EBPα, C/EBPβ, and PPARγ, without inducing cytotoxicity. These results suggest that salicortin could be a potential therapeutic compound for the prevention or treatment of metabolic disorders such as obesity.
Collapse
Affiliation(s)
- Hee Jung Kim
- Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon 16419, Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Samsung Medical Center, Sungkyunkwan University, Seoul 06351, Korea
| | - Da Eun Lee
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea
| | - Eon Chung Park
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea
| | - Moon-Jin Ra
- Hongcheon Institute of Medicinal Herb, Hongcheon-gun 25142, Korea
| | - Sang-Mi Jung
- Hongcheon Institute of Medicinal Herb, Hongcheon-gun 25142, Korea
| | - Jeong-Nam Yu
- Nakdonggang National Institute of Biological Resources, Sangju 37242, Korea
| | - Sung Hee Um
- Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon 16419, Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Samsung Medical Center, Sungkyunkwan University, Seoul 06351, Korea
- Biomedical Institute Convergence, Sungkyunkwan University, Suwon 16419, Korea
- Correspondence: (S.H.U.); (K.H.K.); Tel.: +82-31-299-6123 (S.H.U.); +82-31-290-7700 (K.H.K.)
| | - Ki Hyun Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea
- Correspondence: (S.H.U.); (K.H.K.); Tel.: +82-31-299-6123 (S.H.U.); +82-31-290-7700 (K.H.K.)
| |
Collapse
|
19
|
Lee J, Chung JO, Park SY, Rajamohan N, Singh A, Kim J, Lowe VJ, Lee S. Natural COA water inhibits mitochondrial ROS-mediated apoptosis through Plk3 downregulation under STZ diabetic stress in pancreatic β-cell lines. Biochem Biophys Rep 2022; 30:101247. [PMID: 35300109 PMCID: PMC8921297 DOI: 10.1016/j.bbrep.2022.101247] [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: 11/09/2021] [Revised: 03/05/2022] [Accepted: 03/07/2022] [Indexed: 01/04/2023] Open
Abstract
Diabetes from pancreatic β cell death and insulin resistance is a serious metabolic disease in the world. Although the overproduction of mitochondrial reactive oxygen species (ROS) plays an important role in the pathogenesis of diabetes, its specific molecular mechanism remains unclear. Here, we show that the natural Charisma of Aqua (COA) water plays a role in Streptozotocin (STZ) diabetic stress-induced cell death inhibition. STZ induces mitochondrial ROS by increasing Polo-like kinase 3 (Plk3), a major mitotic regulator, in both Beta TC-6 and Beta TC-tet mouse islet cells and leads to apoptosis. Overexpression of Plk3 regulates an increase in mitochondrial ROS as well as cell death, also these events were inhibited by Plk3 gene knockdown in STZ diabetic stimulated-Beta TC-6 cells. Interestingly, we found that natural COA water blocks mitochondrial ROS generation through the reduction of Plk3 and prevents apoptosis in STZ-treated beta cells. Furthermore, using the 3D organoid (ex vivo) system, we confirmed that the insulin secretion of the supernatant medium under STZ treated pancreatic β-cells is protected by the natural COA water. These findings demonstrate that the natural water COA has a beneficial role in maintaining β cell function through the inhibition of mitochondrial ROS-mediated cell death, and it might be introduced as a potential insulin stabilizer. Pancreatic β cell is stabilized through natural COA water in STZ-induced diabetes. Mitochondrial membrane potential (ΔΨm) is controlled by natural COA water. Plk3 expression under STZ treatment is negatively regulated by natural COA water. Insulin secretion is stabilized by natural COA water under STZ treatment in ex vivo (3D organoid) model.
Collapse
Affiliation(s)
- Jeyeon Lee
- Division of Radiology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Jin Ook Chung
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, 501757, Republic of Korea
| | - Seon-Young Park
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, 501757, Republic of Korea
| | | | - Aparna Singh
- Division of Radiology, Mayo Clinic, Rochester, MN, 55905, USA
| | - JungJin Kim
- Division of Radiology, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA
- Corresponding author. Division of Radiology, Mayo Clinic, Rochester, MN, 55905, USA.
| | - Val J. Lowe
- Division of Radiology, Mayo Clinic, Rochester, MN, 55905, USA
- Corresponding author.
| | - SeungBaek Lee
- Division of Radiology, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA
- Corresponding author. Division of Radiology, Mayo Clinic, Rochester, MN, 55905, USA.
| |
Collapse
|
20
|
Roles of mTOR in the Regulation of Pancreatic β-Cell Mass and Insulin Secretion. Biomolecules 2022; 12:biom12050614. [PMID: 35625542 PMCID: PMC9138643 DOI: 10.3390/biom12050614] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 12/07/2022] Open
Abstract
Pancreatic β-cells are the only type of cells that can control glycemic levels via insulin secretion. Thus, to explore the mechanisms underlying pancreatic β-cell failure, many reports have clarified the roles of important molecules, such as the mechanistic target of rapamycin (mTOR), which is a central regulator of metabolic and nutrient cues. Studies have uncovered the roles of mTOR in the function of β-cells and the progression of diabetes, and they suggest that mTOR has both positive and negative effects on pancreatic β-cells in the development of diabetes.
Collapse
|
21
|
The Roles of Carbohydrate Response Element Binding Protein in the Relationship between Carbohydrate Intake and Diseases. Int J Mol Sci 2021; 22:ijms222112058. [PMID: 34769488 PMCID: PMC8584459 DOI: 10.3390/ijms222112058] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 10/29/2021] [Accepted: 11/05/2021] [Indexed: 12/12/2022] Open
Abstract
Carbohydrates are macronutrients that serve as energy sources. Many studies have shown that carbohydrate intake is nonlinearly associated with mortality. Moreover, high-fructose corn syrup (HFCS) consumption is positively associated with obesity, cardiovascular disease, and type 2 diabetes mellitus (T2DM). Accordingly, products with equal amounts of glucose and fructose have the worst effects on caloric intake, body weight gain, and glucose intolerance, suggesting that carbohydrate amount, kind, and form determine mortality. Understanding the role of carbohydrate response element binding protein (ChREBP) in glucose and lipid metabolism will be beneficial for elucidating the harmful effects of high-fructose corn syrup (HFCS), as this glucose-activated transcription factor regulates glycolytic and lipogenic gene expression. Glucose and fructose coordinately supply the metabolites necessary for ChREBP activation and de novo lipogenesis. Chrebp overexpression causes fatty liver and lower plasma glucose levels, and ChREBP deletion prevents obesity and fatty liver. Intestinal ChREBP regulates fructose absorption and catabolism, and adipose-specific Chrebp-knockout mice show insulin resistance. ChREBP also regulates the appetite for sweets by controlling fibroblast growth factor 21, which promotes energy expenditure. Thus, ChREBP partly mimics the effects of carbohydrate, especially HFCS. The relationship between carbohydrate intake and diseases partly resembles those between ChREBP activity and diseases.
Collapse
|
22
|
Li L, Sakiyama H, Eguchi H, Yoshihara D, Fujiwara N, Suzuki K. Activation of the mitogen-activated protein kinase ERK1/2 signaling pathway suppresses the expression of ChREBPα and β in HepG2 cells. FEBS Open Bio 2021; 11:2008-2018. [PMID: 34051057 PMCID: PMC8255832 DOI: 10.1002/2211-5463.13208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 05/07/2021] [Accepted: 05/26/2021] [Indexed: 11/25/2022] Open
Abstract
The carbohydrate response element‐binding protein (ChREBP), a glucose‐responsive transcription factor that plays a critical role in the glucose‐mediated induction of genes involved in hepatic glycolysis and lipogenesis, exists as two isoforms: ChREBPα and ChREBPβ. However, the mechanism responsible for regulating the expression of both ChREBPα and β, as well as the mechanism that determines which specific isoform is more responsive to different stimuli, remains unclear. To address this issue, we compared the effects of several stimuli, including oxidative stress, on the mRNA and protein expression levels of ChREBPα and β in the hepatocyte cell line, HepG2. We found that H2O2 stimulation suppressed the expression of both mRNA and protein in HepG2 cells, but the mRNA expression level of ChREBPβ was < 1% of that for ChREBPα levels. In addition, the reduction in both ChREBPα and β mRNA levels was reversed by PD98059, a selective and cell permeable inhibitor of the MEK/ERK pathway. Additionally, the administration of 12‐O‐tetradecanoylphorbol 13‐acetate (TPA) and staurosporine (STS), activators of extracellular‐signal‐regulated kinase (ERK) signaling, also resulted in a decrease in the levels of both ChREBPα and β mRNA in HepG2 cells through ERK signaling. These collective data suggest that oxidative stress, including STS treatment, suppresses the expression of ChREBPα and β via the activation of ERK signaling in HepG2 cells. Such a decrease in the levels of expression of ChREBPα and β could result in the suppression of hepatic glycolysis and lipogenesis, and this would be expected to prevent further oxidative stress.
Collapse
Affiliation(s)
- Lan Li
- Department of Biochemistry, Hyogo College of Medicine, Nishinomiya, Japan
| | - Haruhiko Sakiyama
- Department of Biochemistry, Hyogo College of Medicine, Nishinomiya, Japan
| | - Hironobu Eguchi
- Department of Biochemistry, Hyogo College of Medicine, Nishinomiya, Japan
| | - Daisaku Yoshihara
- Department of Biochemistry, Hyogo College of Medicine, Nishinomiya, Japan
| | - Noriko Fujiwara
- Department of Biochemistry, Hyogo College of Medicine, Nishinomiya, Japan
| | - Keiichiro Suzuki
- Department of Biochemistry, Hyogo College of Medicine, Nishinomiya, Japan
| |
Collapse
|
23
|
MicroRNAs and Oxidative Stress: An Intriguing Crosstalk to Be Exploited in the Management of Type 2 Diabetes. Antioxidants (Basel) 2021; 10:antiox10050802. [PMID: 34069422 PMCID: PMC8159096 DOI: 10.3390/antiox10050802] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/17/2021] [Accepted: 05/17/2021] [Indexed: 11/16/2022] Open
Abstract
Type 2 diabetes is a chronic disease widespread throughout the world, with significant human, social, and economic costs. Its multifactorial etiology leads to persistent hyperglycemia, impaired carbohydrate and fat metabolism, chronic inflammation, and defects in insulin secretion or insulin action, or both. Emerging evidence reveals that oxidative stress has a critical role in the development of type 2 diabetes. Overproduction of reactive oxygen species can promote an imbalance between the production and neutralization of antioxidant defence systems, thus favoring lipid accumulation, cellular stress, and the activation of cytosolic signaling pathways, and inducing β-cell dysfunction, insulin resistance, and tissue inflammation. Over the last few years, microRNAs (miRNAs) have attracted growing attention as important mediators of diverse aspects of oxidative stress. These small endogenous non-coding RNAs of 19-24 nucleotides act as negative regulators of gene expression, including the modulation of redox signaling pathways. The present review aims to provide an overview of the current knowledge concerning the molecular crosstalk that takes place between oxidative stress and microRNAs in the physiopathology of type 2 diabetes, with a special emphasis on its potential as a therapeutic target.
Collapse
|
24
|
Bravo-Ruiz I, Medina MÁ, Martínez-Poveda B. From Food to Genes: Transcriptional Regulation of Metabolism by Lipids and Carbohydrates. Nutrients 2021; 13:nu13051513. [PMID: 33946267 PMCID: PMC8145205 DOI: 10.3390/nu13051513] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 04/28/2021] [Indexed: 12/31/2022] Open
Abstract
Lipids and carbohydrates regulate gene expression by means of molecules that sense these macronutrients and act as transcription factors. The peroxisome proliferator-activated receptor (PPAR), activated by some fatty acids or their derivatives, and the carbohydrate response element binding protein (ChREBP), activated by glucose-derived metabolites, play a key role in metabolic homeostasis, especially in glucose and lipid metabolism. Furthermore, the action of both factors in obesity, diabetes and fatty liver, as well as the pharmacological development in the treatment of these pathologies are indeed of high relevance. In this review we present an overview of the discovery, mechanism of activation and metabolic functions of these nutrient-dependent transcription factors in different tissues contexts, from the nutritional genomics perspective. The possibility of targeting these factors in pharmacological approaches is also discussed. Lipid and carbohydrate-dependent transcription factors are key players in the complex metabolic homeostasis, but these factors also drive an adaptive response to non-physiological situations, such as overeating. Possibly the decisive role of ChREBP and PPAR in metabolic regulation points to them as ideal therapeutic targets, but their pleiotropic functions in different tissues makes it difficult to "hit the mark".
Collapse
Affiliation(s)
- Inés Bravo-Ruiz
- Andalucía Tech, Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, E-29071 Málaga, Spain; (I.B.-R.); (M.Á.M.)
| | - Miguel Ángel Medina
- Andalucía Tech, Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, E-29071 Málaga, Spain; (I.B.-R.); (M.Á.M.)
- Instituto de Investigación Biomédica de Málaga (IBIMA), E-29071 Málaga, Spain
- CIBER de Enfermedades Raras (CIBERER), E-29071 Málaga, Spain
| | - Beatriz Martínez-Poveda
- Andalucía Tech, Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, E-29071 Málaga, Spain; (I.B.-R.); (M.Á.M.)
- Instituto de Investigación Biomédica de Málaga (IBIMA), E-29071 Málaga, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), E-28029 Madrid, Spain
- Correspondence:
| |
Collapse
|
25
|
Losada-Barragán M. Physiological effects of nutrients on insulin release by pancreatic beta cells. Mol Cell Biochem 2021; 476:3127-3139. [PMID: 33844157 DOI: 10.1007/s11010-021-04146-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 03/31/2021] [Indexed: 10/21/2022]
Abstract
Obesity and type 2 diabetes (T2D) are growing health problems associated with a loss of insulin sensitivity. Both conditions arise from a long-term energy imbalance, and frequently, lifestyle measures can be useful in its prevention, including physical activity and a healthy diet. Pancreatic β-cells are determinant nutrient sensors that participate in energetic homeostasis needs. However, when pancreatic β-cells are incapable of secreting enough insulin to counteract the reduced sensitivity, the pathology evolves to an insulin resistance condition. The primary nutrient that stimulates insulin secretion is glucose, but also, there are multiple dietary and hormonal factors influencing that response. Many studies of the physiology of β-cells have highlighted the importance of glucose, fructose, amino acids, and free fatty acids on insulin secretion. The present review summarizes recent research on how β-cells respond to the most abundant nutrients that influence insulin secretion. Taken together, understand the subjacent mechanisms of each nutrient on β-cells can help to unravel the effects of mixed variables and complexity in the context of β-cell pathology.
Collapse
Affiliation(s)
- Monica Losada-Barragán
- Grupo de investigación en Biología celular y funcional e ingeniería de biomoléculas, Universidad Antonio Nariño-Sede Circunvalar. Cra, 3 Este # 47A - 15, Bl 5, Bogotá, Colombia.
| |
Collapse
|
26
|
Eguchi N, Vaziri ND, Dafoe DC, Ichii H. The Role of Oxidative Stress in Pancreatic β Cell Dysfunction in Diabetes. Int J Mol Sci 2021; 22:ijms22041509. [PMID: 33546200 PMCID: PMC7913369 DOI: 10.3390/ijms22041509] [Citation(s) in RCA: 160] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/29/2021] [Accepted: 01/30/2021] [Indexed: 02/07/2023] Open
Abstract
Diabetes is a chronic metabolic disorder characterized by inappropriately elevated glucose levels as a result of impaired pancreatic β cell function and insulin resistance. Extensive studies have been conducted to elucidate the mechanism involved in the development of β cell failure and death under diabetic conditions such as hyperglycemia, hyperlipidemia, and inflammation. Of the plethora of proposed mechanisms, endoplasmic reticulum (ER) stress, mitochondrial dysfunction, and oxidative stress have been shown to play a central role in promoting β cell dysfunction. It has become more evident in recent years that these 3 factors are closely interrelated and importantly aggravate each other. Oxidative stress in particular is of great interest to β cell health and survival as it has been shown that β cells exhibit lower antioxidative capacity. Therefore, this review will focus on discussing factors that contribute to the development of oxidative stress in pancreatic β cells and explore the downstream effects of oxidative stress on β cell function and health. Furthermore, antioxidative capacity of β cells to counteract these effects will be discussed along with new approaches focused on preserving β cells under oxidative conditions.
Collapse
Affiliation(s)
- Natsuki Eguchi
- Department of Surgery, University of California, Irvine, CA 92697, USA; (N.E.); (D.C.D.)
| | | | - Donald C. Dafoe
- Department of Surgery, University of California, Irvine, CA 92697, USA; (N.E.); (D.C.D.)
| | - Hirohito Ichii
- Department of Surgery, University of California, Irvine, CA 92697, USA; (N.E.); (D.C.D.)
- Correspondence: ; Tel.: +1-714-456-8590
| |
Collapse
|
27
|
Vasiljević J, Torkko JM, Knoch KP, Solimena M. The making of insulin in health and disease. Diabetologia 2020; 63:1981-1989. [PMID: 32894308 PMCID: PMC7476993 DOI: 10.1007/s00125-020-05192-7] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 04/28/2020] [Indexed: 12/16/2022]
Abstract
The discovery of insulin in 1921 has been one of greatest scientific achievements of the 20th century. Since then, the availability of insulin has shifted the focus of diabetes treatment from trying to keep patients alive to saving and improving the life of millions. Throughout this time, basic and clinical research has advanced our understanding of insulin synthesis and action, both in healthy and pathological conditions. Yet, multiple aspects of insulin production remain unknown. In this review, we focus on the most recent findings on insulin synthesis, highlighting their relevance in diabetes. Graphical abstract.
Collapse
Affiliation(s)
- Jovana Vasiljević
- Molecular Diabetology, University Hospital and Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
- Paul Langerhans Institute Dresden (PLID), Helmholtz Center Munich, University Hospital and Faculty of Medicine Carl Gustav Carus, TU Dresden, Tatzberg 47/49, 01307, Dresden, Germany
| | - Juha M Torkko
- Molecular Diabetology, University Hospital and Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
- Paul Langerhans Institute Dresden (PLID), Helmholtz Center Munich, University Hospital and Faculty of Medicine Carl Gustav Carus, TU Dresden, Tatzberg 47/49, 01307, Dresden, Germany
| | - Klaus-Peter Knoch
- Molecular Diabetology, University Hospital and Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
- Paul Langerhans Institute Dresden (PLID), Helmholtz Center Munich, University Hospital and Faculty of Medicine Carl Gustav Carus, TU Dresden, Tatzberg 47/49, 01307, Dresden, Germany
| | - Michele Solimena
- Molecular Diabetology, University Hospital and Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany.
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany.
- Paul Langerhans Institute Dresden (PLID), Helmholtz Center Munich, University Hospital and Faculty of Medicine Carl Gustav Carus, TU Dresden, Tatzberg 47/49, 01307, Dresden, Germany.
- Max Planck Institute of Molecular Cell Biology and Genetics (MPI-CBG), Dresden, Germany.
| |
Collapse
|
28
|
Nagy L, Béke F, Juhász L, Kovács T, Juhász-Tóth É, Docsa T, Tóth A, Gergely P, Somsák L, Bai P. Glycogen phosphorylase inhibitor, 2,3-bis[(2E)-3-(4-hydroxyphenyl)prop-2-enamido] butanedioic acid (BF142), improves baseline insulin secretion of MIN6 insulinoma cells. PLoS One 2020; 15:e0236081. [PMID: 32960890 PMCID: PMC7508380 DOI: 10.1371/journal.pone.0236081] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 06/30/2020] [Indexed: 12/17/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM), one of the most common metabolic diseases, is characterized by insulin resistance and inadequate insulin secretion of β cells. Glycogen phosphorylase (GP) is the key enzyme in glycogen breakdown, and contributes to hepatic glucose production during fasting or during insulin resistance. Pharmacological GP inhibitors are potential glucose lowering agents, which may be used in T2DM therapy. A natural product isolated from the cultured broth of the fungal strain No. 138354, called 2,3-bis(4-hydroxycinnamoyloxy)glutaric acid (FR258900), was discovered a decade ago. In vivo studies showed that FR258900 significantly reduced blood glucose levels in diabetic mice. We previously showed that GP inhibitors can potently enhance the function of β cells. The purpose of this study was to assess whether an analogue of FR258900 can influence β cell function. BF142 (Meso-Dimethyl 2,3-bis[(E)-3-(4-acetoxyphenyl)prop-2-enamido]butanedioate) treatment activated the glucose-stimulated insulin secretion pathway, as indicated by enhanced glycolysis, increased mitochondrial oxidation, significantly increased ATP production, and elevated calcium influx in MIN6 cells. Furthermore, BF142 induced mTORC1-specific phosphorylation of S6K, increased levels of PDX1 and insulin protein, and increased insulin secretion. Our data suggest that BF142 can influence β cell function and can support the insulin producing ability of β cells.
Collapse
Affiliation(s)
- Lilla Nagy
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Ferenc Béke
- Department of Organic Chemistry, Faculty of Science and Technology, University of Debrecen, Debrecen, Hungary
| | - László Juhász
- Department of Organic Chemistry, Faculty of Science and Technology, University of Debrecen, Debrecen, Hungary
| | - Tünde Kovács
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Éva Juhász-Tóth
- Department of Organic Chemistry, Faculty of Science and Technology, University of Debrecen, Debrecen, Hungary
| | - Tibor Docsa
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Attila Tóth
- Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Pál Gergely
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - László Somsák
- Department of Organic Chemistry, Faculty of Science and Technology, University of Debrecen, Debrecen, Hungary
| | - Péter Bai
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- MTA-DE Lendület Laboratory of Cellular Metabolism, Debrecen, Hungary
- Research Center for Molecular Medicine, University of Debrecen, Debrecen, Hungary
- * E-mail:
| |
Collapse
|
29
|
Tu G, Dai C, Qu H, Wang Y, Liao B. Role of exercise and rapamycin on the expression of energy metabolism genes in liver tissues of rats fed a high‑fat diet. Mol Med Rep 2020; 22:2932-2940. [PMID: 32945385 PMCID: PMC7453655 DOI: 10.3892/mmr.2020.11362] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Accepted: 06/26/2020] [Indexed: 12/19/2022] Open
Abstract
The mTOR pathway serves an important role in the development of insulin resistance induced by obesity. Exercise improves obesity-associated insulin resistance and hepatic energy metabolism; however, the precise mechanism of this process remains unknown. Therefore, the present study investigated the role of rapamycin, an inhibitor of mTOR, on exercise-induced expression of hepatic energy metabolism genes in rats fed a high-fat diet (HFD). A total of 30 male rats were divided into the following groups: Normal group (n=6) fed chow diets and HFD group (n=24) fed an HFD for 6 weeks. The HFD rats performed exercise adaptation for 1 week and were randomly divided into the four following groups (each containing six rats): i) Group of HFD rats with sedentary (H group); ii) group of HFD rats with exercise (HE group); iii) group of HFD rats with rapamycin (HR group); and iv) group of HFD rats with exercise and rapamycin (HER group). Both HE and HER rats were placed on incremental treadmill training for 4 weeks (from week 8–11). Both HR and HER rats were injected with rapamycin intraperitoneally at the dose of 2 mg/kg once a day for 2 weeks (from week 10–11). All rats were sacrificed following a 12–16 h fasting period at the end of week 11. The levels of mitochondrial and oxidative enzyme activities, as well as of the expression of genes involved in energy metabolism were assessed in liver tissues. Biochemical assays and oil red staining were used to assess the content of hepatic triglycerides (TGs). The results indicated that exercise, but not rapamycin, reduced TG content in the liver of HFD rats. Further analysis indicated that rapamycin reduced the activity of cytochrome c oxidase, but not the activities of succinate dehydrogenase and β-hydroxyacyl-CoA dehydrogenase in the liver of HFD rats. Exercise significantly upregulated the mRNA expression of peroxisome proliferator-activated receptor γ coactivator 1 β, while rapamycin exhibited no effect on the mRNA expression levels of hepatic transcription factors associated with energy metabolism enzymes in the liver of HFD rats. Collectively, the results indicated that exercise reduced TG content and upregulated mitochondrial metabolic gene expression in the liver of HFD rats. Moreover, this mechanism may not involve the mTOR pathway.
Collapse
Affiliation(s)
- Genghong Tu
- Department of Sports Medicine, Guangzhou Sport University, Guangzhou, Guangdong 510150, P.R. China
| | - Chunyan Dai
- Department of Sports Medicine, Guangzhou Sport University, Guangzhou, Guangdong 510150, P.R. China
| | - Haofei Qu
- Department of Sports Medicine, Guangzhou Sport University, Guangzhou, Guangdong 510150, P.R. China
| | - Yunzhen Wang
- Department of Sports Medicine, Guangzhou Sport University, Guangzhou, Guangdong 510150, P.R. China
| | - Bagen Liao
- Department of Sports Medicine, Guangzhou Sport University, Guangzhou, Guangdong 510150, P.R. China
| |
Collapse
|
30
|
Helman A, Cangelosi AL, Davis JC, Pham Q, Rothman A, Faust AL, Straubhaar JR, Sabatini DM, Melton DA. A Nutrient-Sensing Transition at Birth Triggers Glucose-Responsive Insulin Secretion. Cell Metab 2020; 31:1004-1016.e5. [PMID: 32375022 PMCID: PMC7480404 DOI: 10.1016/j.cmet.2020.04.004] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 01/14/2020] [Accepted: 03/31/2020] [Indexed: 12/31/2022]
Abstract
A drastic transition at birth, from constant maternal nutrient supply in utero to intermittent postnatal feeding, requires changes in the metabolic system of the neonate. Despite their central role in metabolic homeostasis, little is known about how pancreatic β cells adjust to the new nutritional challenge. Here, we find that after birth β cell function shifts from amino acid- to glucose-stimulated insulin secretion in correlation with the change in the nutritional environment. This adaptation is mediated by a transition in nutrient sensitivity of the mTORC1 pathway, which leads to intermittent mTORC1 activity. Disrupting nutrient sensitivity of mTORC1 in mature β cells reverts insulin secretion to a functionally immature state. Finally, manipulating nutrient sensitivity of mTORC1 in stem cell-derived β cells in vitro strongly enhances their glucose-responsive insulin secretion. These results reveal a mechanism by which nutrients regulate β cell function, thereby enabling a metabolic adaptation for the newborn.
Collapse
Affiliation(s)
- Aharon Helman
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA.
| | - Andrew L Cangelosi
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Cambridge, MA 02139, USA; Koch Institute for Integrative Cancer Research, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jeffrey C Davis
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Quan Pham
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Arielle Rothman
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Aubrey L Faust
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Juerg R Straubhaar
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - David M Sabatini
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Cambridge, MA 02139, USA; Koch Institute for Integrative Cancer Research, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| | - Douglas A Melton
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Howard Hughes Medical Institute, Cambridge, MA 02139, USA.
| |
Collapse
|
31
|
Weir GC. Glucolipotoxicity, β-Cells, and Diabetes: The Emperor Has No Clothes. Diabetes 2020; 69:273-278. [PMID: 31519699 PMCID: PMC7034184 DOI: 10.2337/db19-0138] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 04/08/2019] [Indexed: 12/21/2022]
Abstract
Reduction of β-cell mass and function is central to the pathogenesis of type 2 diabetes. The terms glucotoxicity, lipotoxicity, and glucolipotoxicity are used to describe potentially responsible processes. The premise is that chronically elevated glucose levels are toxic to β-cells, that elevated lipid levels in the form of circulating free fatty acids (FFA) also have toxic effects, and that the combination of the two, glucolipotoxicity, is particularly harmful. Much work has shown that high concentrations of FFA can be very damaging to β-cells when used for in vitro experiments, and when infused in large amounts in humans and rodents they produce suppression of insulin secretion. The purpose of this Perspective is to raise doubts about whether the FFA levels found in real-life situations are ever high enough to cause problems. Evidence supporting the importance of glucotoxicity is strong because there is such a tight correlation between defective insulin secretion and rising glucose levels. However, there is virtually no convincing evidence that the alterations in FFA levels occurring during progression to diabetes are pathogenic. Thus, the terms lipotoxicity and glucolipotoxicity should be used with great caution, if at all, because evidence supporting their importance has not yet emerged.
Collapse
Affiliation(s)
- Gordon C Weir
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA
| |
Collapse
|
32
|
Ganesan H, Balasubramanian V, Iyer M, Venugopal A, Subramaniam MD, Cho SG, Vellingiri B. mTOR signalling pathway - A root cause for idiopathic autism? BMB Rep 2020. [PMID: 31186084 PMCID: PMC6675248 DOI: 10.5483/bmbrep.2019.52.7.137] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental monogenic disorder with a strong genetic influence. Idiopathic autism could be defined as a type of autism that does not have a specific causative agent. Among signalling cascades, mTOR signalling pathway plays a pivotal role not only in cell cycle, but also in protein synthesis and regulation of brain homeostasis in ASD patients. The present review highlights, underlying mechanism of mTOR and its role in altered signalling cascades as a triggering factor in the onset of idiopathic autism. Further, this review discusses how distorted mTOR signalling pathway stimulates truncated translation in neuronal cells and leads to downregulation of protein synthesis at dendritic spines of the brain. This review concludes by suggesting downstream regulators such as p70S6K, eIF4B, eIF4E of mTOR signalling pathway as promising therapeutic targets for idiopathic autistic individuals. [BMB Reports 2019; 52(7): 424-433].
Collapse
Affiliation(s)
- Harsha Ganesan
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore 641046, Tamil Nadu, India
| | - Venkatesh Balasubramanian
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore 641046, Tamil Nadu, India
| | - Mahalaxmi Iyer
- Department of Zoology, Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore 641043, Tamil Nadu, India
| | - Anila Venugopal
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore 641046, Tamil Nadu, India
| | - Mohana Devi Subramaniam
- Department of Genetics and Molecular Biology, Vision Research Foundation, Sankara Nethralaya, Chennai 600006, Tamil Nadu, India
| | - Ssang-Goo Cho
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 05029, Korea
| | - Balachandar Vellingiri
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore 641046, Tamil Nadu, India
| |
Collapse
|
33
|
Park S, Lee G, Lee H, Hoang T, Chae H. Glucose-lowering effect of Gryllus bimaculatus powder on streptozotocin-induced diabetes through the AKT/mTOR pathway. Food Sci Nutr 2020; 8:402-409. [PMID: 31993166 PMCID: PMC6977414 DOI: 10.1002/fsn3.1323] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 05/26/2019] [Accepted: 06/13/2019] [Indexed: 12/19/2022] Open
Abstract
This study was carried out to elucidate the antidiabetic effects of Gryllus bimaculatus powder using a streptozotocin (STZ)-induced rat model of type I diabetes. Administration of the insect powder significantly rescued representative diabetes markers (i.e., insulin and C-peptide) in STZ-treated rats. Improved glucose tolerance test (GTT) and insulin tolerance test (ITT) results were also observed, indicating that Gryllus bimaculatus powder exerts antidiabetic effects. Gryllus bimaculatus powder administration rescued STZ-induced alterations in both islet morphology and insulin staining patterns. The extract increased antiapoptotic Bcl2 expression and decreased proapoptotic Bax and active caspase 3 expressions. In addition, the Gryllus bimaculatus powder supplementation enhanced AKT/mTOR pathway, a key marker of the state of anabolic metabolism, and its downstream effector, mTOR. Collectively, our results suggest that Gryllus bimaculatus contributes to the maintenance of pancreatic β-cell function and morphology against a diabetic state through the regulations against apoptosis and anabolic metabolism.
Collapse
Affiliation(s)
- Seon‐Ah Park
- Non‐Clinical Evaluation CenterBiomedical Research InstituteChonbuk National University HospitalJeonjuChonbukSouth Korea
| | - Geum‐Hwa Lee
- Non‐Clinical Evaluation CenterBiomedical Research InstituteChonbuk National University HospitalJeonjuChonbukSouth Korea
| | - Hwa‐Young Lee
- Non‐Clinical Evaluation CenterBiomedical Research InstituteChonbuk National University HospitalJeonjuChonbukSouth Korea
- Department of Pharmacology and Institute of New Drug DevelopmentSchool of MedicineChonbuk National UniversityJeonjuChonbukSouth Korea
| | - The‐Hiep Hoang
- Non‐Clinical Evaluation CenterBiomedical Research InstituteChonbuk National University HospitalJeonjuChonbukSouth Korea
- Department of Pharmacology and Institute of New Drug DevelopmentSchool of MedicineChonbuk National UniversityJeonjuChonbukSouth Korea
| | - Han‐Jung Chae
- Non‐Clinical Evaluation CenterBiomedical Research InstituteChonbuk National University HospitalJeonjuChonbukSouth Korea
- Department of Pharmacology and Institute of New Drug DevelopmentSchool of MedicineChonbuk National UniversityJeonjuChonbukSouth Korea
| |
Collapse
|
34
|
Esch N, Jo S, Moore M, Alejandro EU. Nutrient Sensor mTOR and OGT: Orchestrators of Organelle Homeostasis in Pancreatic β-Cells. J Diabetes Res 2020; 2020:8872639. [PMID: 33457426 PMCID: PMC7787834 DOI: 10.1155/2020/8872639] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 11/06/2020] [Accepted: 11/24/2020] [Indexed: 02/08/2023] Open
Abstract
The purpose of this review is to integrate the role of nutrient-sensing pathways into β-cell organelle dysfunction prompted by nutrient excess during type 2 diabetes (T2D). T2D encompasses chronic hyperglycemia, hyperlipidemia, and inflammation, which each contribute to β-cell failure. These factors can disrupt the function of critical β-cell organelles, namely, the ER, mitochondria, lysosomes, and autophagosomes. Dysfunctional organelles cause defects in insulin synthesis and secretion and activate apoptotic pathways if homeostasis is not restored. In this review, we will focus on mTORC1 and OGT, two major anabolic nutrient sensors with important roles in β-cell physiology. Though acute stimulation of these sensors frequently improves β-cell function and promotes adaptation to cell stress, chronic and sustained activity disturbs organelle homeostasis. mTORC1 and OGT regulate organelle function by influencing the expression and activities of key proteins, enzymes, and transcription factors, as well as by modulating autophagy to influence clearance of defective organelles. In addition, mTORC1 and OGT activity influence islet inflammation during T2D, which can further disrupt organelle and β-cell function. Therapies for T2D that fine-tune the activity of these nutrient sensors have yet to be developed, but the important role of mTORC1 and OGT in organelle homeostasis makes them promising targets to improve β-cell function and survival.
Collapse
Affiliation(s)
- Nicholas Esch
- Department of Integrative Biology & Physiology, University of Minnesota Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| | - Seokwon Jo
- Department of Integrative Biology & Physiology, University of Minnesota Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| | - Mackenzie Moore
- Department of Integrative Biology & Physiology, University of Minnesota Medical School, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Surgery, University of Minnesota Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| | - Emilyn U. Alejandro
- Department of Integrative Biology & Physiology, University of Minnesota Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
35
|
Melnik BC. Milk exosomal miRNAs: potential drivers of AMPK-to-mTORC1 switching in β-cell de-differentiation of type 2 diabetes mellitus. Nutr Metab (Lond) 2019; 16:85. [PMID: 31827573 PMCID: PMC6898964 DOI: 10.1186/s12986-019-0412-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 11/22/2019] [Indexed: 12/15/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) steadily increases in prevalence since the 1950's, the period of widespread distribution of refrigerated pasteurized cow's milk. Whereas breastfeeding protects against the development of T2DM in later life, accumulating epidemiological evidence underlines the role of cow's milk consumption in T2DM. Recent studies in rodent models demonstrate that during the breastfeeding period pancreatic β-cells are metabolically immature and preferentially proliferate by activation of mechanistic target of rapamycin complex 1 (mTORC1) and suppression of AMP-activated protein kinase (AMPK). Weaning determines a metabolic switch of β-cells from a proliferating, immature phenotype with low insulin secretion to a differentiated mature phenotype with glucose-stimulated insulin secretion, less proliferation, reduced mTORC1- but increased AMPK activity. Translational evidence presented in this perspective implies for the first time that termination of milk miRNA transfer is the driver of this metabolic switch. miRNA-148a is a key inhibitor of AMPK and phosphatase and tensin homolog, crucial suppressors of mTORC1. β-Cells of diabetic patients return to the postnatal phenotype with high mTORC1 and low AMPK activity, explained by continuous transfer of bovine milk miRNAs to the human milk consumer. Bovine milk miRNA-148a apparently promotes β-cell de-differentiation to the immature mTORC1-high/AMPK-low phenotype with functional impairments in insulin secretion, increased mTORC1-driven endoplasmic reticulum stress, reduced autophagy and early β-cell apoptosis. In contrast to pasteurized cow's milk, milk's miRNAs are inactivated by bacterial fermentation, boiling and ultra-heat treatment and are missing in current infant formula. Persistent milk miRNA signaling adds a new perspective to the pathogenesis of T2DM and explains the protective role of breastfeeding but the diabetogenic effect of continued milk miRNA signaling by persistent consumption of pasteurized cow's milk.
Collapse
Affiliation(s)
- Bodo C. Melnik
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, Am Finkenhügel 7A, D-49076 Osnabrück, Germany
| |
Collapse
|
36
|
Dafre AL, Schmitz AE, Maher P. Rapid and persistent loss of TXNIP in HT22 neuronal cells under carbonyl and hyperosmotic stress. Neurochem Int 2019; 132:104585. [PMID: 31678323 DOI: 10.1016/j.neuint.2019.104585] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 10/27/2019] [Accepted: 10/28/2019] [Indexed: 12/11/2022]
Abstract
Thioredoxin interacting protein (TXNIP) binds to thioredoxin thereby limiting its activity, but it also promotes internalization of glucose transporters, participates in inflammasome activation, and controls autophagy. Published data and this work demonstrate that TXNIP responds to a number of apparently unrelated stresses, such as serum deprivation, pH change, and oxidative, osmotic and carbonyl stress. Interestingly, we noticed that hyperosmotic (NaCl) and carbonyl (methylglyoxal, MGO) stresses in HT22 neuronal cells produced a rapid loss of TXNIP (half-life ∼12 min), prompting us to search for possible mechanisms controlling this TXNIP loss, including pH change, serum deprivation, calcium metabolism and inhibition of the proteasome and other proteases, autophagy and MAPKs. None of these routes stopped the TXNIP loss induced by hyperosmotic and carbonyl stress. Besides transcriptional, translational and microRNA regulation, there is evidence indicating that mTOR and AMPK also control TXNIP expression. Indeed, AMPK-deficient mouse embryonic fibroblasts failed to respond to phenformin (AMPK activator) and compound C (AMPK inhibitor), while rapamycin induced a marked increase in TXNIP levels, confirming the known AMPK/mTOR control over TXNIP. However, the TXNIP loss induced by NaCl or MGO were observed even in AMPK deficient MEFs or after mTOR inhibition, indicating AMPK/mTOR does not participate in this rapid TXNIP loss. These results suggest that rapid TXNIP loss is a general and immediate response to stress that can improve energy availability and antioxidant protection, eventually culminating in better cell survival.
Collapse
Affiliation(s)
- Alcir Luiz Dafre
- Biochemistry Department, Federal University of Santa Catarina, 88040-900, Florianópolis, SC, Brazil.
| | - Ariana Ern Schmitz
- Biochemistry Department, Federal University of Santa Catarina, 88040-900, Florianópolis, SC, Brazil
| | - Pamela Maher
- Cellular Neurobiology Laboratory, Salk Institute for Biological Studies, CA, 92037, La Jolla, United States.
| |
Collapse
|
37
|
Song Z, Yang H, Zhou L, Yang F. Glucose-Sensing Transcription Factor MondoA/ChREBP as Targets for Type 2 Diabetes: Opportunities and Challenges. Int J Mol Sci 2019; 20:5132. [PMID: 31623194 PMCID: PMC6829382 DOI: 10.3390/ijms20205132] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/12/2019] [Accepted: 10/14/2019] [Indexed: 12/16/2022] Open
Abstract
The worldwide increase in type 2 diabetes (T2D) is becoming a major health concern, thus searching for novel preventive and therapeutic strategies has become urgent. In last decade, the paralogous transcription factors MondoA and carbohydrate response element-binding protein (ChREBP) have been revealed to be central mediators of glucose sensing in multiple metabolic organs. Under normal nutrient conditions, MondoA/ChREBP plays vital roles in maintaining glucose homeostasis. However, under chronic nutrient overload, the dysregulation of MondoA/ChREBP contributes to metabolic disorders, such as insulin resistance (IR) and T2D. In this review, we aim to provide an overview of recent advances in the understanding of MondoA/ChREBP and its roles in T2D development. Specifically, we will briefly summarize the functional similarities and differences between MondoA and ChREBP. Then, we will update the roles of MondoA/ChREBP in four T2D-associated metabolic organs (i.e., the skeletal muscle, liver, adipose tissue, and pancreas) in physiological and pathological conditions. Finally, we will discuss the opportunities and challenges of MondoA/ChREBP as drug targets for anti-diabetes. By doing so, we highlight the potential use of therapies targeting MondoA/ChREBP to counteract T2D and its complications.
Collapse
Affiliation(s)
- Ziyi Song
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China.
- Departments of Medicine and Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Hao Yang
- Division of Medical Genetics, Department of Pediatrics, Université de Montréal and CHU Sainte-Justine, Montreal, QC H3T 1C5, Canada.
| | - Lei Zhou
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China.
| | - Fajun Yang
- Departments of Medicine and Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
38
|
Son SW, Chau GC, Kim ST, Um SH. Vacuolar H +-ATPase Subunit V0C Regulates Aerobic Glycolysis of Esophageal Cancer Cells via PKM2 Signaling. Cells 2019; 8:E1137. [PMID: 31554233 PMCID: PMC6830105 DOI: 10.3390/cells8101137] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 09/16/2019] [Accepted: 09/20/2019] [Indexed: 12/16/2022] Open
Abstract
The vacuolar H+-adenosine triphosphatase (ATPase) subunit V0C (ATP6V0C), a proton-conducting, pore-forming subunit of vacuolar ATPase, maintains pH homeostasis and induces organelle acidification. The intracellular and extracellular pH of cancer cells affects their growth; however, the role of ATP6V0C in highly invasive esophageal cancer cells (ECCs) remains unclear. In this study, we examined the role of ATP6V0C in glucose metabolism in ECCs. The ATP6V0C depletion attenuated ECC proliferation, invasion, and suppressed glucose metabolism, as indicated by reduced glucose uptake and decreased lactate and adenosine triphosphate (ATP) production in cells. Consistent with this, expression of glycolytic enzyme and the extracellular acidification rate (ECAR) were also decreased by ATP6V0C knockdown. Mechanistically, ATP6V0C interacted with pyruvate kinase isoform M2 (PKM2), a key regulator of glycolysis in ECCs. The ATP6V0C depletion reduced PKM2 phosphorylation at tyrosine residue 105 (Tyr105), leading to inhibition of nuclear translocation of PKM2. In addition, ATP6V0C was recruited at hypoxia response element (HRE) sites in the lactate dehydrogenase A (LDHA) gene for glycolysis. Thus, our data suggest that ATP6V0C enhances aerobic glycolysis and motility in ECCs.
Collapse
Affiliation(s)
- Sung Wook Son
- Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, Gyeonggi-do 16419, Korea.
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Samsung Medical Center, Sungkyunkwan University, Seoul 06351, Korea.
| | - Gia Cac Chau
- Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, Gyeonggi-do 16419, Korea.
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Samsung Medical Center, Sungkyunkwan University, Seoul 06351, Korea.
| | - Seong-Tae Kim
- Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, Gyeonggi-do 16419, Korea.
| | - Sung Hee Um
- Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, Gyeonggi-do 16419, Korea.
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Samsung Medical Center, Sungkyunkwan University, Seoul 06351, Korea.
- Biomedical Institute Convergence at Sungkyunkwan University, Suwon, Gyeonggi-do 16419, Korea.
| |
Collapse
|
39
|
Jaafar R, Tran S, Shah AN, Sun G, Valdearcos M, Marchetti P, Masini M, Swisa A, Giacometti S, Bernal-Mizrachi E, Matveyenko A, Hebrok M, Dor Y, Rutter GA, Koliwad SK, Bhushan A. mTORC1 to AMPK switching underlies β-cell metabolic plasticity during maturation and diabetes. J Clin Invest 2019; 129:4124-4137. [PMID: 31265435 PMCID: PMC6763225 DOI: 10.1172/jci127021] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 06/24/2019] [Indexed: 12/28/2022] Open
Abstract
Pancreatic beta cells (β-cells) differentiate during fetal life, but only postnatally acquire the capacity for glucose-stimulated insulin secretion (GSIS). How this happens is not clear. In exploring what molecular mechanisms drive the maturation of β-cell function, we found that the control of cellular signaling in β-cells fundamentally switched from the nutrient sensor target of rapamycin (mTORC1) to the energy sensor 5'-adenosine monophosphate-activated protein kinase (AMPK), and that this was critical for functional maturation. Moreover, AMPK was activated by the dietary transition taking place during weaning, and this in turn inhibited mTORC1 activity to drive the adult β-cell phenotype. While forcing constitutive mTORC1 signaling in adult β-cells relegated them to a functionally immature phenotype with characteristic transcriptional and metabolic profiles, engineering the switch from mTORC1 to AMPK signaling was sufficient to promote β-cell mitochondrial biogenesis, a shift to oxidative metabolism, and functional maturation. We also found that type 2 diabetes, a condition marked by both mitochondrial degeneration and dysregulated GSIS, was associated with a remarkable reversion of the normal AMPK-dependent adult β-cell signature to a more neonatal one characterized by mTORC1 activation. Manipulating the way in which cellular nutrient signaling pathways regulate β-cell metabolism may thus offer new targets to improve β-cell function in diabetes.
Collapse
Affiliation(s)
- Rami Jaafar
- The Diabetes Center, UCSF, San Francisco, California, USA
| | - Stella Tran
- The Diabetes Center, UCSF, San Francisco, California, USA
| | - Ajit N. Shah
- The Diabetes Center, UCSF, San Francisco, California, USA
| | - Gao Sun
- Section of Cell Biology and Functional Genomics, Department of Medicine, Imperial College London, Hammersmith Hospital London, United Kingdom
| | | | - Piero Marchetti
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Matilde Masini
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Avital Swisa
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | | | - Ernesto Bernal-Mizrachi
- Division of Endocrinology, Diabetes and Metabolism, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | - Aleksey Matveyenko
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Yuval Dor
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Guy A. Rutter
- Section of Cell Biology and Functional Genomics, Department of Medicine, Imperial College London, Hammersmith Hospital London, United Kingdom
| | | | - Anil Bhushan
- The Diabetes Center, UCSF, San Francisco, California, USA
| |
Collapse
|
40
|
Sangüesa G, Roglans N, Baena M, Velázquez AM, Laguna JC, Alegret M. mTOR is a Key Protein Involved in the Metabolic Effects of Simple Sugars. Int J Mol Sci 2019; 20:ijms20051117. [PMID: 30841536 PMCID: PMC6429387 DOI: 10.3390/ijms20051117] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 02/18/2019] [Accepted: 02/28/2019] [Indexed: 12/14/2022] Open
Abstract
One of the most important threats to global human health is the increasing incidences of metabolic pathologies (including obesity, type 2 diabetes and non-alcoholic fatty liver disease), which is paralleled by increasing consumptions of hypercaloric diets enriched in simple sugars. The challenge is to identify the metabolic pathways affected by the excessive consumption of these dietary components when they are consumed in excess, to unravel the molecular mechanisms leading to metabolic pathologies and identify novel therapeutic targets to manage them. Mechanistic (mammalian) target of rapamycin (mTOR) has emerged as one of the key molecular nodes that integrate extracellular signals, such as energy status and nutrient availability, to trigger cell responses that could lead to the above-mentioned diseases through the regulation of lipid and glucose metabolism. By activating mTOR signalling, excessive consumption of simple sugars (such as fructose and glucose), could modulate hepatic gluconeogenesis, lipogenesis and fatty acid uptake and catabolism and thus lipid deposition in the liver. In the present review we will discuss some of the most recent studies showing the central role of mTOR in the metabolic effects of excessive simple sugar consumption.
Collapse
Affiliation(s)
- Gemma Sangüesa
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, School of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain.
- Institute of Biomedicine, University of Barcelona, 08028 Barcelona, Spain.
| | - Núria Roglans
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, School of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain.
- Institute of Biomedicine, University of Barcelona, 08028 Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBERObn), 28029 Madrid, Spain.
| | - Miguel Baena
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, School of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain.
- Institute of Biomedicine, University of Barcelona, 08028 Barcelona, Spain.
| | - Ana Magdalena Velázquez
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, School of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain.
| | - Juan Carlos Laguna
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, School of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain.
- Institute of Biomedicine, University of Barcelona, 08028 Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBERObn), 28029 Madrid, Spain.
| | - Marta Alegret
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, School of Pharmacy and Food Science, University of Barcelona, 08028 Barcelona, Spain.
- Institute of Biomedicine, University of Barcelona, 08028 Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBERObn), 28029 Madrid, Spain.
| |
Collapse
|
41
|
Ye R, Onodera T, Scherer PE. Lipotoxicity and β Cell Maintenance in Obesity and Type 2 Diabetes. J Endocr Soc 2019; 3:617-631. [PMID: 30834357 PMCID: PMC6391718 DOI: 10.1210/js.2018-00372] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Accepted: 01/30/2019] [Indexed: 12/11/2022] Open
Abstract
Obesity and diabetes are often associated with lipotoxic conditions in multiple tissues. The insulin-producing β cells are susceptible to elevated lipid levels and the ensuing lipotoxicity. The preservation of β cell mass and function is one of the main goals of diabetes management under these metabolically stressful conditions. However, the adverse effects from the adaptive signaling pathways that β cells use to counteract lipotoxic stress have secondary negative effects in their own right. Antilipotoxic signaling cascades in β cells can contribute to their eventual failure. Such dual roles are seen for many other biological adaptive processes as well.
Collapse
Affiliation(s)
- Risheng Ye
- Department of Medical Education, Texas Tech University Health Sciences Center Paul L. Foster School of Medicine, El Paso, Texas
- Touchstone Diabetes Center, Department of Internal Medicine, the University of Texas Southwestern Medical Center, Dallas, Texas
| | - Toshiharu Onodera
- Touchstone Diabetes Center, Department of Internal Medicine, the University of Texas Southwestern Medical Center, Dallas, Texas
| | - Philipp E Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, the University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
42
|
Liang H, Nie J, Van Skike CE, Valentine JM, Orr ME. Mammalian Target of Rapamycin at the Crossroad Between Alzheimer's Disease and Diabetes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1128:185-225. [PMID: 31062331 DOI: 10.1007/978-981-13-3540-2_10] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Accumulating evidence suggests that Alzheimer's disease may manifest as a metabolic disorder with pathology and/or dysfunction in numerous tissues. Adults with Alzheimer's disease suffer with significantly more comorbidities than demographically matched Medicare beneficiaries (Zhao et al, BMC Health Serv Res 8:108, 2008b). Reciprocally, comorbid health conditions increase the risk of developing Alzheimer's disease (Haaksma et al, PLoS One 12(5):e0177044, 2017). Type 2 diabetes mellitus is especially notable as the disease shares many overlapping pathologies observed in patients with Alzheimer's disease, including hyperglycemia, hyperinsulinemia, insulin resistance, glucose intolerance, dyslipidemia, inflammation, and cognitive dysfunction, as described in Chap. 8 of this book (Yoshitake et al, Neurology 45(6):1161-1168, 1995; Leibson et al, Am J Epidemiol 145(4):301-308, 1997; Ott et al, Neurology 53(9):1937-1942, 1999; Voisin et al, Rev Med Interne 24(Suppl 3):288s-291s, 2003; Janson et al. Diabetes 53(2):474-481, 2004; Ristow M, J Mol Med (Berl) 82(8):510-529, 2004; Whitmer et al, BMJ 330(7504):1360, 2005, Curr Alzheimer Res 4(2):103-109, 2007; Ohara et al, Neurology 77(12):1126-1134, 2011). Although nondiabetic older adults also experience age-related cognitive decline, diabetes is uniquely associated with a twofold increased risk of Alzheimer's disease, as described in Chap. 2 of this book (Yoshitake et al, Neurology 45(6):1161-1168, 1995; Leibson et al, Am J Epidemiol 145(4):301-308, 1997; Ott et al. Neurology 53(9):1937-1942, 1999; Ohara et al, Neurology 77(12):1126-1134, 2011). Good glycemic control has been shown to improve cognitive status (Cukierman-et al, Diabetes Care 32(2):221-226, 2009), and the use of insulin sensitizers is correlated with a lower rate of cognitive decline in older adults (Morris JK, Burns JM, Curr Neurol Neurosci Rep 12(5):520-527, 2012). At the molecular level, the mechanistic/mammalian target of rapamycin (mTOR) plays a key role in maintaining energy homeostasis. Nutrient availability and cellular stress information, both extracellular and intracellular, are integrated and transduced through mTOR signaling pathways. Aberrant regulation of mTOR occurs in the brains of patients with Alzheimer's disease and in numerous tissues of individuals with type 2 diabetes (Mannaa et al, J Mol Med (Berl) 91(10):1167-1175, 2013). Moreover, modulating mTOR activity with a pharmacological inhibitor, rapamycin, provides wide-ranging health benefits, including healthy life span extension in numerous model organisms (Vellai et al, Nature 426(6967):620, 2003; Jia et al, Development 131(16):3897-3906, 2004; Kapahi et al, Curr Biol 14(10):885-890, 2004; Kaeberlein et al, Science 310(5751):1193-1196, 2005; Powers et al, Genes Dev 20(2):174-184, 2006; Harrison et al, Nature 460(7253):392-395, 2009; Selman et al, Science 326(5949):140-144, 2009; Sharp ZD, Strong R, J Gerontol A Biol Sci Med Sci 65(6):580-589, 2010), which underscores its importance to overall organismal health and longevity. In this chapter, we discuss the physiological role of mTOR signaling and the consequences of mTOR dysregulation in the brain and peripheral tissues, with emphasis on its relevance to the development of Alzheimer's disease and link to type 2 diabetes.
Collapse
Affiliation(s)
- Hanyu Liang
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Jia Nie
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Candice E Van Skike
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Joseph M Valentine
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Miranda E Orr
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
- San Antonio Geriatric Research, Education and Clinical Center, South Texas Veterans Health Care System, San Antonio, TX, USA.
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, San Antonio, TX, USA.
| |
Collapse
|
43
|
A single extra copy of Down syndrome critical region 1-4 results in impaired hepatic glucose homeostasis. Mol Metab 2018; 21:82-89. [PMID: 30583978 PMCID: PMC6407364 DOI: 10.1016/j.molmet.2018.12.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 11/29/2018] [Accepted: 12/03/2018] [Indexed: 01/19/2023] Open
Abstract
Objectives During fasting, hepatic gluconeogenesis is induced to maintain energy homeostasis. Moreover, abnormal dysregulation of hepatic glucose production is commonly observed in type 2 diabetes. However, the signaling components controlling hepatic glucose production to maintain normal glucose levels are not fully understood. Here, we examined the physiological role of Down syndrome critical region 1–4 (DSCR1-4), an endogenous calcineurin signaling inhibitor in the liver that mediates metabolic adaptation to fasting. Methods We assessed the effect of cyclosporine A, an inhibitor of calcineurin signaling on gluconeogenic gene expression in primary hepatocytes. DSCR1-4 expression was examined in diet- and genetically-induced mouse models of obesity. We also investigated the metabolic phenotype of a single extra copy of DSCR1-4 in transgenic mice and how DSCR1-4 regulates glucose homeostasis in the liver. Results Treatment with cyclosporin A increased hepatic glucose production and gluconeogenic gene expression. The expression of DSCR1-4 was induced by refeeding and overexpressed in obese mouse livers. Moreover, transgenic mice with a single extra copy of DSCR1-4 exhibited pyruvate intolerance and impaired glucose homeostasis. Mechanistically, DSCR1-4 overexpression increased phosphorylation of the cAMP response element-binding protein, which led to elevated expression levels of gluconeogenic genes and, thus, enhanced hepatic glucose production during fasting. Conclusion A single extra copy of DSCR1-4 results in dysregulated hepatic glucose homeostasis and pyruvate intolerance. Our findings suggest that nutrient-sensitive DSCR1-4 is a novel target for controlling hepatic gluconeogenesis in diabetes. DSCR1 mRNA and protein levels are increased in livers upon nutrient availability. DSCR1-4 is overexpressed in diet- or genetically induced obesity. DSCR1-4 trisomy mice exhibit impaired glucose homeostasis and pyruvate intolerance. Trisomy of DSCR1-4 leads to increased hepatic glucose production.
Collapse
|
44
|
Role of mTOR in Glucose and Lipid Metabolism. Int J Mol Sci 2018; 19:ijms19072043. [PMID: 30011848 PMCID: PMC6073766 DOI: 10.3390/ijms19072043] [Citation(s) in RCA: 184] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 07/10/2018] [Accepted: 07/11/2018] [Indexed: 02/06/2023] Open
Abstract
The mammalian target of rapamycin, mTOR is the master regulator of a cell’s growth and metabolic state in response to nutrients, growth factors and many extracellular cues. Its dysregulation leads to a number of metabolic pathological conditions, including obesity and type 2 diabetes. Here, we review recent findings on the role of mTOR in major metabolic organs, such as adipose tissues, liver, muscle, pancreas and brain. And their potentials as the mTOR related pharmacological targets will be also discussed.
Collapse
|
45
|
Lin HS, Shi JB, Peng CM, Zheng BC, Cheng FC, Lee MW, Lee HW, Wu PF, Liu YJ. Manipulating the Temperature of Sulfurization to Synthesize α-NiS Nanosphere Film for Long-Term Preservation of Non-enzymatic Glucose Sensors. NANOSCALE RESEARCH LETTERS 2018; 13:109. [PMID: 29675727 PMCID: PMC5908778 DOI: 10.1186/s11671-018-2511-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 04/02/2018] [Indexed: 06/08/2023]
Abstract
In this study, alpha nickel sulfide (α-NiS) nanosphere films have been successfully synthesized by electroplating the nickel nanosheet film on the indium tin oxide (ITO) glass substrate and sulfuring nickel-coated ITO glass substrate. First, we electrodeposited the nickel nanosheet films on the ITO glass substrates which were cut into a 0.5 × 1 cm2 size. Second, the nanosheet nickel films were annealed in vacuum-sealed glass ampoules with sulfur sheets at different annealing temperatures (300, 400, and 500 °C) for 4 h in vacuum-sealed glass ampoules. The α-NiS films were investigated by using X-ray diffraction (XRD), variable vacuum scanning electron microscopy (VVSEM), field emission scanning electron microscopy/energy dispersive spectrometer (FE-SEM/EDS), cyclic voltammogram (CV), electrochemical impedance spectroscopy (EIS), ultraviolet/visible/near-infrared (UV/Visible/NIR) spectra, and photoluminescence (PL) spectra. Many nanospheres were observed on the surface of the α-NiS films at the annealing temperature 400 °C for 4 h. We also used the high-resolution transmission electron microscopy (HR-TEM) for the analysis of the α-NiS nanospheres. We demonstrated that our α-NiS nanosphere film had a linear current response to different glucose concentrations. Additionally, our α-NiS nanosphere films were preserved at room temperature for five and a half years and were still useful for detecting glucose at low concentration.
Collapse
Affiliation(s)
- Hsien-Sheng Lin
- Ph.D. Program in Electrical and Communications Engineering, Feng Chia University, 100, Wen-Hwa Rd, Seatwen, Taichung, 40724 Taiwan
| | - Jen-Bin Shi
- Department of Electronic Engineering, Feng Chia University, 100, Wen-Hwa Rd., Seatwen, Taichung, 40724 Taiwan
| | - Cheng-Ming Peng
- Ph.D. Program in Electrical and Communications Engineering, Feng Chia University, 100, Wen-Hwa Rd, Seatwen, Taichung, 40724 Taiwan
- Da Vinci Minimally Invasive Surgery Center, Chung Shan Medical University Hospital, No.110, Sec.1, Chien-Kuo N. Rd., Taichung, 40201 Taiwan
| | - Bo-Chi Zheng
- Ph.D. Program in Electrical and Communications Engineering, Feng Chia University, 100, Wen-Hwa Rd, Seatwen, Taichung, 40724 Taiwan
| | - Fu-Chou Cheng
- Department of Medical Research, Taichung Veterans General Hospital, No. 160, 3rd Section, Taichung Harbor Road, Taichung, 40705 Taiwan
| | - Ming-Way Lee
- Department of Physics, Institute of Nanoscience, National Chung Hsing University, 250 Kuo Kuang Road, Taichung, 40227 Taiwan
| | - Hsuan-Wei Lee
- Ph.D. Program in Electrical and Communications Engineering, Feng Chia University, 100, Wen-Hwa Rd, Seatwen, Taichung, 40724 Taiwan
| | - Po-Feng Wu
- College of General Education, No. 1018, Sec. 6, Taiwan Boulevard, Shalu District, Taichung, 43302 Taiwan
| | - Yi-Jui Liu
- Department of Automatic Control Engineering, Feng Chia University, No.100, Wenhwa Rd., Seatwen, Taichung, 40724 Taiwan
| |
Collapse
|
46
|
Thielen L, Shalev A. Diabetes pathogenic mechanisms and potential new therapies based upon a novel target called TXNIP. Curr Opin Endocrinol Diabetes Obes 2018; 25:75-80. [PMID: 29356688 PMCID: PMC5831522 DOI: 10.1097/med.0000000000000391] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE OF REVIEW Thioredoxin-interacting protein has emerged as a major factor regulating pancreatic β-cell dysfunction and death, key processes in the pathogenesis of type 1 and type 2 diabetes. Accumulating evidence based on basic, preclinical, and retrospective epidemiological research suggests that TXNIP represents a promising therapeutic target for diabetes. The present review is aimed at providing an update regarding these developments. RECENT FINDINGS TXNIP has been shown to be induced by glucose and increased in diabetes and to promote β-cell apoptosis, whereas TXNIP deletion protected against diabetes. More recently, TXNIP inhibition has also been found to promote insulin production and glucagon-like peptide 1 signaling via regulation of a microRNA. β-Cell TXNIP expression itself was found to be regulated by hypoglycemic agents, carbohydrate-response-element-binding protein, and cytosolic calcium or the calcium channel blocker, verapamil. Retrospective studies now further suggest that verapamil use might be associated with a lower incidence of type 2 diabetes in humans. SUMMARY TXNIP has emerged as a key factor in the regulation of functional β-cell mass and TXNIP inhibition has shown beneficial effects in a variety of studies. Thus, the inhibition of TXNIP may provide a novel approach to the treatment of diabetes.
Collapse
Affiliation(s)
- Lance Thielen
- Division of Endocrinology, Diabetes, and Metabolism, Comprehensive Diabetes Center and Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | |
Collapse
|
47
|
Villamayor L, Rodríguez-Seguel E, Araujo R, Carrasco M, Bru-Tarí E, Mellado-Gil JM, Gauthier BR, Martinelli P, Quesada I, Soria B, Martín F, Cano DA, Rojas A. GATA6 Controls Insulin Biosynthesis and Secretion in Adult β-Cells. Diabetes 2018; 67:448-460. [PMID: 29263149 DOI: 10.2337/db17-0364] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 12/13/2017] [Indexed: 11/13/2022]
Abstract
GATA4 and GATA6 play essential, but redundant, roles in pancreas formation in mice, and GATA6 mutations cause pancreatic agenesis in humans. GATA6 mutations have also recently been linked to adult-onset diabetes, with subclinical or no exocrine insufficiency, suggesting an important role for GATA6 in human β-cell physiology. To investigate the role of GATA6 in the adult endocrine pancreas, we generated mice in which Gata6 is specifically inactivated in the pancreas. These mice develop glucose intolerance. Islets deficient in GATA6 activity display decreased insulin content and impaired insulin secretion. Gata6-deficient β-cells exhibit ultrastructural abnormalities, including increased immature insulin granules, swollen mitochondria, and disorganized endoplasmic reticulum. We also demonstrate that Pdx1 expression in adult β-cells depends on GATA sites in transgenic reporter mice and that loss of GATA6 greatly affects β-cell-specific gene expression. These findings demonstrate the essential role of GATA6 in β-cell function.
Collapse
Affiliation(s)
- Laura Villamayor
- Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER), Universidad Pablo de Olavide, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas (CSIC), Seville, Spain
| | - Elisa Rodríguez-Seguel
- Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER), Universidad Pablo de Olavide, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas (CSIC), Seville, Spain
| | - Raquel Araujo
- Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER), Universidad Pablo de Olavide, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas (CSIC), Seville, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Manuel Carrasco
- Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER), Universidad Pablo de Olavide, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas (CSIC), Seville, Spain
| | | | - José Manuel Mellado-Gil
- Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER), Universidad Pablo de Olavide, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas (CSIC), Seville, Spain
| | - Benoit R Gauthier
- Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER), Universidad Pablo de Olavide, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas (CSIC), Seville, Spain
| | - Paola Martinelli
- Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
- Institute for Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Iván Quesada
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
- Universidad Miguel Hernández, Elche, Spain
| | - Bernat Soria
- Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER), Universidad Pablo de Olavide, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas (CSIC), Seville, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Franz Martín
- Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER), Universidad Pablo de Olavide, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas (CSIC), Seville, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - David A Cano
- Unidad de Gestión de Endocrinología y Nutrición, Instituto de Biomedicina de Sevilla (IBiS), Consejo Superior de Investigaciones Científicas, Universidad de Sevilla, Hospital Universitario Virgen del Rocío, Seville, Spain
| | - Anabel Rojas
- Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER), Universidad Pablo de Olavide, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas (CSIC), Seville, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| |
Collapse
|
48
|
mTORC2 Signaling: A Path for Pancreatic β Cell's Growth and Function. J Mol Biol 2018; 430:904-918. [PMID: 29481838 DOI: 10.1016/j.jmb.2018.02.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 02/14/2018] [Accepted: 02/14/2018] [Indexed: 12/16/2022]
Abstract
The mechanistic target of rapamycin (mTOR) signaling pathway is an evolutionary conserved pathway that senses signals from nutrients and growth factors to regulate cell growth, metabolism and survival. mTOR acts in two biochemically and functionally distinct complexes, mTOR complex 1 (mTORC1) and 2 (mTORC2), which differ in terms of regulatory mechanisms, substrate specificity and functional outputs. While mTORC1 signaling has been extensively studied in islet/β-cell biology, recent findings demonstrate a distinct role for mTORC2 in the regulation of pancreatic β-cell function and mass. mTORC2, a key component of the growth factor receptor signaling, is declined in β cells under diabetogenic conditions and in pancreatic islets from patients with type 2 diabetes. β cell-selective mTORC2 inactivation leads to glucose intolerance and acceleration of diabetes as a result of reduced β-cell mass, proliferation and impaired glucose-stimulated insulin secretion. Thereby, many mTORC2 targets, such as AKT, PKC, FOXO1, MST1 and cell cycle regulators, play an important role in β-cell survival and function. This indicates mTORC2 as important pathway for the maintenance of β-cell homeostasis, particularly to sustain proper β-cell compensatory response in the presence of nutrient overload and metabolic demand. This review summarizes recent emerging advances on the contribution of mTORC2 and its associated signaling on the regulation of glucose metabolism and functional β-cell mass under physiological and pathophysiological conditions in type 2 diabetes.
Collapse
|
49
|
Ardestani A, Lupse B, Kido Y, Leibowitz G, Maedler K. mTORC1 Signaling: A Double-Edged Sword in Diabetic β Cells. Cell Metab 2018; 27:314-331. [PMID: 29275961 DOI: 10.1016/j.cmet.2017.11.004] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 10/11/2017] [Accepted: 11/15/2017] [Indexed: 12/21/2022]
Abstract
The mechanistic target of rapamycin complex 1 (mTORC1) is a central regulator of metabolic and nutrient cues that integrates environmental inputs into downstream signaling pathways to control cellular metabolism, growth, and survival. While numerous in vitro and in vivo studies reported the positive functions of mTORC1 in the regulation of β cell survival and proliferation under physiological conditions, more recent work demonstrates the opposite in the long term; this is exemplified by the constitutive inappropriate hyper-activation of mTORC1 in diabetic islets or β cells under conditions of increased β cell stress and metabolic demands. These recent findings uncover mTORC1's importance as an emerging significant player in the development and progression of β cell failure in type 2 diabetes and suggest that mTORC1 may act as a "double edge sword" in the regulation of β cell mass and function in response to metabolic stress such as nutrient overload and insulin resistance.
Collapse
Affiliation(s)
- Amin Ardestani
- University of Bremen, Centre for Biomolecular Interactions Bremen, Bremen 28359, Germany.
| | - Blaz Lupse
- University of Bremen, Centre for Biomolecular Interactions Bremen, Bremen 28359, Germany
| | - Yoshiaki Kido
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; Division of Metabolism and Disease, Department of Biophysics, Kobe University Graduate School of Health Sciences, Kobe 654-0142, Japan
| | - Gil Leibowitz
- Endocrinology and Metabolism Service and the Hadassah Diabetes Unit, Department of Medicine, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel
| | - Kathrin Maedler
- University of Bremen, Centre for Biomolecular Interactions Bremen, Bremen 28359, Germany.
| |
Collapse
|
50
|
Giguère V. Canonical signaling and nuclear activity of mTOR-a teamwork effort to regulate metabolism and cell growth. FEBS J 2018; 285:1572-1588. [PMID: 29337437 DOI: 10.1111/febs.14384] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 12/21/2017] [Accepted: 01/10/2018] [Indexed: 01/07/2023]
Abstract
Mechanistic (or mammalian) target of rapamycin (mTOR) is a kinase that regulates almost all functions related to cell growth and metabolism in response to extra- and intracellular stimuli, such as availability of nutrients, the presence of growth factors, or the energy status of the cell. As part of two distinct protein complexes, mTORC1 and mTORC2, the kinase has been shown to influence cell growth and proliferation by controlling ribosome biogenesis, mRNA translation, carbohydrate and lipid metabolism, protein degradation, autophagy as well as microtubule and actin dynamics. In addition to these well-characterized functions, mTOR can also influence gene transcription. While most studies focused on investigating how canonical mTOR signaling regulates the activity of transcription factors outside the nucleus, recent findings point to a more direct role for mTOR as a transcription factor operating on chromatin in the nucleus. In particular, recent genome-wide identification of mTOR targets on chromatin reveals that its activities in the nucleus and cytoplasm are functionally and biologically linked, thus uncovering a novel paradigm in mTOR function.
Collapse
Affiliation(s)
- Vincent Giguère
- Departments of Biochemistry, Medicine and Oncology, Faculty of Medicine, Goodman Cancer Research Centre, McGill University, Montréal, Canada
| |
Collapse
|