1
|
Takahashi N, Kimura AP, Yoshizaki T, Ohmura K. Imeglimin modulates mitochondria biology and facilitates mitokine secretion in 3T3-L1 adipocytes. Life Sci 2024; 349:122735. [PMID: 38768776 DOI: 10.1016/j.lfs.2024.122735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 04/22/2024] [Accepted: 05/15/2024] [Indexed: 05/22/2024]
Abstract
AIMS Imeglimin, a novel antidiabetic drug, has recently been reported to affect pancreatic β-cells and hepatocytes. Adipose tissue plays a crucial role in systemic metabolism. However, its effect on adipocytes remains unexplored. Herein, we investigated the effects of imeglimin on adipocytes, particularly in the mitochondria. MAIN METHODS The 3T3-L1 adipocytes were treated with imeglimin. Mitochondrial respiratory complex I activity and NAD+, NADH, and AMP levels were measured. Protein expression levels were determined by western blotting, mitochondrial DNA and mRNA expression levels were determined using quantitative polymerase chain reaction, and secreted adipocytokine and mitokine levels were determined using adipokine array and enzyme-linked immunosorbent assay. KEY FINDINGS Imeglimin inhibited complex I activity, decreased the NAD+/NADH ratio, and increased AMP levels, which were associated with the enhanced phosphorylation of AMP-activated protein kinase. In addition, imeglimin increased the mitochondrial DNA content and levels of mitochondrial transcription factor A and peroxisome proliferator-activated receptor-γ coactivator 1-α mRNA, which were abolished by Ly294002, a phosphoinositide 3-kinase inhibitor. Furthermore, imeglimin facilitated the expression levels of markers of the mitochondrial unfolded protein response, and the gene expression and secretion of two mitokines, fibroblast growth factor 21 and growth differentiation factor 15. The production of both mitokines was transcriptionally regulated and abolished by phosphoinositide 3-kinase and Akt inhibitors. SIGNIFICANCE Imeglimin modulates mitochondrial biology in adipocytes and may exert a mitohormetic effect through mitokine secretion.
Collapse
Affiliation(s)
- Nobuhiko Takahashi
- Division of Internal Medicine, Department of Human Biology and Pathophysiology, School of Dentistry, Health Sciences University of Hokkaido, 1757 Kanazawa, Ishikari-Tobetsu, Hokkaido 061-0023, Japan.
| | - Atsushi P Kimura
- Department of Biological Sciences, Faculty of Science, Hokkaido University, Sapporo, Hokkaido 060-0810, Japan
| | - Takayuki Yoshizaki
- Department of Biotechnology, Faculty of Life Science and Biotechnology, Fukuyama University, Hiroshima 729-0292, Japan
| | - Kazumasa Ohmura
- Division of Internal Medicine, Department of Human Biology and Pathophysiology, School of Dentistry, Health Sciences University of Hokkaido, 1757 Kanazawa, Ishikari-Tobetsu, Hokkaido 061-0023, Japan
| |
Collapse
|
2
|
Chang W, Li W, Li P. The anti-diabetic effects of metformin are mediated by regulating long non-coding RNA. Front Pharmacol 2023; 14:1256705. [PMID: 38053839 PMCID: PMC10694297 DOI: 10.3389/fphar.2023.1256705] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 11/02/2023] [Indexed: 12/07/2023] Open
Abstract
Type 2 diabetes (T2D) is a metabolic disease with complex etiology and mechanisms. Long non-coding ribonucleic acid (LncRNA) is a novel class of functional long RNA molecules that regulate multiple biological functions through various mechanisms. Studies in the past decade have shown that lncRNAs may play an important role in regulating insulin resistance and the progression of T2D. As a widely used biguanide drug, metformin has been used for glucose lowering effects in clinical practice for more than 60 years. For diabetic therapy, metformin reduces glucose absorption from the intestines, lowers hepatic gluconeogenesis, reduces inflammation, and improves insulin sensitivity. However, despite being widely used as the first-line oral antidiabetic drug, its mechanism of action remains largely elusive. Currently, an increasing number of studies have demonstrated that the anti-diabetic effects of metformin were mediated by the regulation of lncRNAs. Metformin-regulated lncRNAs have been shown to participate in the inhibition of gluconeogenesis, regulation of lipid metabolism, and be anti-inflammatory. Thus, this review focuses on the mechanisms of action of metformin in regulating lncRNAs in diabetes, including pathways altered by metformin via targeting lncRNAs, and the potential targets of metformin through modulation of lncRNAs. Knowledge of the mechanisms of lncRNA modulation by metformin in diabetes will aid the development of new therapeutic drugs for T2D in the future.
Collapse
Affiliation(s)
- Wenguang Chang
- Institute for Translational Medicine, The Affiliated Hospital, College of Medicine, Qingdao University, Qingdao, China
| | - Wei Li
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
| | - Peifeng Li
- Institute for Translational Medicine, The Affiliated Hospital, College of Medicine, Qingdao University, Qingdao, China
| |
Collapse
|
3
|
Zong Y, Wang X, Cui B, Xiong X, Wu A, Lin C, Zhang Y. Decoding the regulatory roles of non-coding RNAs in cellular metabolism and disease. Mol Ther 2023; 31:1562-1576. [PMID: 37113055 PMCID: PMC10277898 DOI: 10.1016/j.ymthe.2023.04.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 04/12/2023] [Accepted: 04/21/2023] [Indexed: 04/29/2023] Open
Abstract
Non-coding RNAs, including long non-coding RNAs (lncRNAs), microRNAs (miRNAs), and circular RNAs (circRNAs), are being studied extensively in a variety of fields. Their roles in metabolism have received increasing attention in recent years but are not yet clear. The regulation of glucose, fatty acid, and amino acid metabolism is an imperative physiological process that occurs in living organisms and takes part in cancer and cardiovascular diseases. Here, we summarize the important roles played by non-coding RNAs in glucose metabolism, fatty acid metabolism, and amino acid metabolism, as well as the mechanisms involved. We also summarize the therapeutic advances for non-coding RNAs in diseases such as obesity, cardiovascular disease, and some metabolic diseases. Overall, non-coding RNAs are indispensable factors in metabolism and have a significant role in the three major metabolisms, which may be exploited as therapeutic targets in the future.
Collapse
Affiliation(s)
- Yuru Zong
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Xuliang Wang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China
| | - Bing Cui
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Xiaowei Xiong
- Department of Cardiology and Macrovascular Disease, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China
| | - Andrew Wu
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Chunru Lin
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; The Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Yaohua Zhang
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
4
|
The role of MicroRNA networks in tissue-specific direct and indirect effects of metformin and its application. Biomed Pharmacother 2022; 151:113130. [PMID: 35598373 DOI: 10.1016/j.biopha.2022.113130] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/06/2022] [Accepted: 05/13/2022] [Indexed: 11/20/2022] Open
Abstract
Metformin is a first-line oral antidiabetic agent that results in clear benefits in relation to glucose metabolism and diabetes-related complications. The specific regulatory details and mechanisms underlying these benefits are still unclear and require further investigation. There is recent mounting evidence that metformin has pleiotropic effects on the target tissue development in metabolic organs, including adipose tissue, the gastrointestinal tract and the liver. The mechanism of actions of metformin are divided into direct effects on target tissues and indirect effects via non-targeted tissues. MicroRNAs (miRNAs) are a class of endogenous, noncoding, negative gene regulators that have emerged as important regulators of a number of diseases, including type 2 diabetes mellitus (T2DM). Metformin is involved in many aspects of miRNA regulation, and metformin treatment in T2DM should be associated with other miRNA targets. A large number of miRNAs regulation by metformin in target tissues with either direct or indirect effects has gradually been revealed in the context of numerous diseases and has gradually received increasing attention. This paper thoroughly reviews the current knowledge about the role of miRNA networks in the tissue-specific direct and indirect effects of metformin. Furthermore, this knowledge provides a novel theoretical basis and suggests therapeutic targets for the clinical treatment of metformin and miRNA regulators in the prevention and treatment of cancer, cardiovascular disorders, diabetes and its complications.
Collapse
|
5
|
Qiu C, Li C, Zheng Q, Fang S, Xu J, Wang H, Guo H. Metformin suppresses lung adenocarcinoma by downregulating long non-coding RNA (lncRNA) AFAP1-AS1 and secreted phosphoprotein 1 (SPP1) while upregulating miR-3163. Bioengineered 2022; 13:11987-12002. [PMID: 35603556 PMCID: PMC9275981 DOI: 10.1080/21655979.2021.2005981] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
AFAP1-AS1 plays a pro-tumor role in lung cancer. However, no investigation has focused on whether it is involved in the anticancer activity of metformin (Met) in the treatment of lung adenocarcinoma (LUAD). Reverse transcription quantitative polymerase chain reaction (RT-qPCR) was performed to detect the expression of long non-coding (lnc)RNA AFAP1-AS1, the microRNA (miR)-3163, and secreted phosphoprotein 1 (SPP1) in LUAD tissues, or of A549 and H3122 cells. Cell Counting Kit-8, wound scratch, and cell invasion assays were performed to evaluate the effect of the overexpression of lncRNA AFAP1-AS1, miR-3163, and SPP1 on the malignant behaviors of A549 and H3122 cells. Phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/Akt/mTOR) signaling pathway-related proteins were detected by Western blot analysis. Dual luciferase reporter or RIP assays were used to determine the interplay between AFAP1-AS1 and miR-3163, or of miR-3163 and SPP1. Met inhibits the malignant characteristics of A549 and H3122 cells in vitro. GEPIA database analysis showed that AFAP1-AS1 is a highly expressed lncRNA in LUAD tissues, which was validated by RT-qPCR. Overexpression of AFAP1-AS1 suppressed the met-mediated anti-tumor activity in A549 and H3122 cells, while AFAP1-AS1 silencing promoted it. Met inhibited AFAP1-AS1 expression, which resulted in reduced proliferation, migration, and invasion in A549 and H3122 cells. This led to AFAP1-AS1-mediated suppression of miR-3163 and, subsequently, the upregulation of SPP1. Met exerts its antitumor activities by regulating the AFAP1-AS1/miR-3163/SPP1/PI3K/Akt/mTOR axis. Our findings deepen our understanding of mechanisms underlying anti-tumor effect of Met in LUAD.
Collapse
Affiliation(s)
- Caiyu Qiu
- Department of Physical Examination Center, Wuhan Third Hospital, Wuhan, Hubei, China
| | - Chuanxiang Li
- Department of Respiratory and Critical Care Medicine, Wuhan Third Hospital, Wuhan, Hubei, China
| | - Quan Zheng
- Department of Respiratory and Critical Care Medicine, Wuhan Third Hospital, Wuhan, Hubei, China
| | - Si Fang
- Department of Respiratory and Critical Care Medicine, Wuhan Third Hospital, Wuhan, Hubei, China
| | - Jianqun Xu
- Department of Respiratory and Critical Care Medicine, Wuhan Third Hospital, Wuhan, Hubei, China
| | - Hongjuan Wang
- Department of Respiratory and Critical Care Medicine, Wuhan Third Hospital, Wuhan, Hubei, China
| | - Hongrong Guo
- Department of Respiratory and Critical Care Medicine, Wuhan Third Hospital, Wuhan, Hubei, China
| |
Collapse
|
6
|
Juni RP, ’t Hart KC, Houtkooper RH, Boon R. Long non‐coding RNAs in cardiometabolic disorders. FEBS Lett 2022; 596:1367-1387. [DOI: 10.1002/1873-3468.14370] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 03/29/2022] [Accepted: 04/07/2022] [Indexed: 11/08/2022]
Affiliation(s)
- Rio P. Juni
- Department of Physiology Amsterdam University Medical Centers Amsterdam Cardiovascular Science Frankfurt am Main Germany
| | - Kelly C. ’t Hart
- Department of Physiology Amsterdam University Medical Centers Amsterdam Cardiovascular Science Frankfurt am Main Germany
- Laboratory Genetic Metabolic Diseases Amsterdam University Medical Centers; Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam Cardiovascular Science, University of Amsterdam Frankfurt am Main Germany
| | - Riekelt H. Houtkooper
- Laboratory Genetic Metabolic Diseases Amsterdam University Medical Centers; Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam Cardiovascular Science, University of Amsterdam Frankfurt am Main Germany
| | - Reinier Boon
- Department of Physiology Amsterdam University Medical Centers Amsterdam Cardiovascular Science Frankfurt am Main Germany
- Institute for Cardiovascular Regeneration Centre for Molecular Medicine Goethe University Frankfurt am Main Frankfurt am Main Germany
- German Centre for Cardiovascular Research DZHK Partner site Frankfurt Rhein/Main Frankfurt am Main Germany
| |
Collapse
|
7
|
Metformin and Insulin Resistance: A Review of the Underlying Mechanisms behind Changes in GLUT4-Mediated Glucose Transport. Int J Mol Sci 2022; 23:ijms23031264. [PMID: 35163187 PMCID: PMC8836112 DOI: 10.3390/ijms23031264] [Citation(s) in RCA: 109] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/21/2022] [Accepted: 01/21/2022] [Indexed: 02/06/2023] Open
Abstract
Metformin is the most commonly used treatment to increase insulin sensitivity in insulin-resistant (IR) conditions such as diabetes, prediabetes, polycystic ovary syndrome, and obesity. There is a well-documented correlation between glucose transporter 4 (GLUT4) expression and the level of IR. Therefore, the observed increase in peripheral glucose utilization after metformin treatment most likely comes from the induction of GLUT4 expression and its increased translocation to the plasma membrane. However, the mechanisms behind this effect and the critical metformin targets are still largely undefined. The present review explores the evidence for the crucial role of changes in the expression and activation of insulin signaling pathway mediators, AMPK, several GLUT4 translocation mediators, and the effect of posttranscriptional modifications based on previously published preclinical and clinical models of metformin’s mode of action in animal and human studies. Our aim is to provide a comprehensive review of the studies in this field in order to shed some light on the complex interactions between metformin action, GLUT4 expression, GLUT4 translocation, and the observed increase in peripheral insulin sensitivity.
Collapse
|
8
|
Tello-Flores VA, Beltrán-Anaya FO, Ramírez-Vargas MA, Esteban-Casales BE, Navarro-Tito N, Alarcón-Romero LDC, Luciano-Villa CA, Ramírez M, del Moral-Hernández Ó, Flores-Alfaro E. Role of Long Non-Coding RNAs and the Molecular Mechanisms Involved in Insulin Resistance. Int J Mol Sci 2021; 22:7256. [PMID: 34298896 PMCID: PMC8306787 DOI: 10.3390/ijms22147256] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 06/27/2021] [Accepted: 07/02/2021] [Indexed: 12/14/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) are single-stranded RNA biomolecules with a length of >200 nt, and they are currently considered to be master regulators of many pathological processes. Recent publications have shown that lncRNAs play important roles in the pathogenesis and progression of insulin resistance (IR) and glucose homeostasis by regulating inflammatory and lipogenic processes. lncRNAs regulate gene expression by binding to other non-coding RNAs, mRNAs, proteins, and DNA. In recent years, several mechanisms have been reported to explain the key roles of lncRNAs in the development of IR, including metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), imprinted maternal-ly expressed transcript (H19), maternally expressed gene 3 (MEG3), myocardial infarction-associated transcript (MIAT), and steroid receptor RNA activator (SRA), HOX transcript antisense RNA (HOTAIR), and downregulated Expression-Related Hexose/Glucose Transport Enhancer (DREH). LncRNAs participate in the regulation of lipid and carbohydrate metabolism, the inflammatory process, and oxidative stress through different pathways, such as cyclic adenosine monophosphate/protein kinase A (cAMP/PKA), phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT), polypyrimidine tract-binding protein 1/element-binding transcription factor 1c (PTBP1/SREBP-1c), AKT/nitric oxide synthase (eNOS), AKT/forkhead box O1 (FoxO1), and tumor necrosis factor-alpha (TNF-α)/c-Jun-N-terminal kinases (JNK). On the other hand, the mechanisms linked to the molecular, cellular, and biochemical actions of lncRNAs vary according to the tissue, biological species, and the severity of IR. Therefore, it is essential to elucidate the role of lncRNAs in the insulin signaling pathway and glucose and lipid metabolism. This review analyzes the function and molecular mechanisms of lncRNAs involved in the development of IR.
Collapse
Affiliation(s)
- Vianet Argelia Tello-Flores
- Laboratorio de Epidemiología Clínica y Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39087, GRO, Mexico; (V.A.T.-F.); (F.O.B.-A.); (M.A.R.-V.); (B.E.E.-C.); (C.A.L.-V.)
| | - Fredy Omar Beltrán-Anaya
- Laboratorio de Epidemiología Clínica y Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39087, GRO, Mexico; (V.A.T.-F.); (F.O.B.-A.); (M.A.R.-V.); (B.E.E.-C.); (C.A.L.-V.)
| | - Marco Antonio Ramírez-Vargas
- Laboratorio de Epidemiología Clínica y Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39087, GRO, Mexico; (V.A.T.-F.); (F.O.B.-A.); (M.A.R.-V.); (B.E.E.-C.); (C.A.L.-V.)
| | - Brenda Ely Esteban-Casales
- Laboratorio de Epidemiología Clínica y Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39087, GRO, Mexico; (V.A.T.-F.); (F.O.B.-A.); (M.A.R.-V.); (B.E.E.-C.); (C.A.L.-V.)
| | - Napoleón Navarro-Tito
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39087, GRO, Mexico;
| | - Luz del Carmen Alarcón-Romero
- Laboratorio de Citopatología e Histoquímica, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39087, GRO, Mexico;
| | - Carlos Aldair Luciano-Villa
- Laboratorio de Epidemiología Clínica y Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39087, GRO, Mexico; (V.A.T.-F.); (F.O.B.-A.); (M.A.R.-V.); (B.E.E.-C.); (C.A.L.-V.)
| | - Mónica Ramírez
- CONACyT, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39087, GRO, Mexico;
| | - Óscar del Moral-Hernández
- Laboratorio de Virología, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39087, GRO, Mexico
| | - Eugenia Flores-Alfaro
- Laboratorio de Epidemiología Clínica y Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo 39087, GRO, Mexico; (V.A.T.-F.); (F.O.B.-A.); (M.A.R.-V.); (B.E.E.-C.); (C.A.L.-V.)
| |
Collapse
|
9
|
Takahashi N, Kimura AP, Ohmura K, Naito S, Yoshida M, Ieko M. Knockdown of long noncoding RNA dreh facilitates cell surface GLUT4 expression and glucose uptake through the involvement of vimentin in 3T3-L1 adipocytes. Gene 2020; 735:144404. [PMID: 32018013 DOI: 10.1016/j.gene.2020.144404] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 01/27/2020] [Indexed: 01/16/2023]
Abstract
Glucose uptake in adipocytes is crucial for regulating systemic metabolism. Long noncoding RNAs (lncRNAs), defined as being transcripts with lengths exceeding 200 nucleotides that are not translated, are recently identified regulators of cellular functions. Previously, we have shown that an lncRNA, "down-regulated expression by hepatitis B virus X" (dreh), is involved in glucose transport in skeletal muscle cells. Here, we aimed to examine the involvement of dreh in glucose transport in 3T3-L1 adipocytes. Expression analysis showed that dreh was expressed in 3T3-L1 fibroblasts and adipocytes. Knockdown of dreh expression using its specific siRNAs lowered the glucose concentration of the medium and facilitated [3H]-2-deoxyglucose transport in adipocytes. Additionally, dreh silencing enhanced the protein expression of glucose transporter (GLUT4) in the plasma membrane of adipocytes. Treatment with siRNA against vimentin attenuated the glucose-lowering effect of dreh depletion. These results suggest that the repression of dreh facilitates glucose transport via increased GLUT4 expression in the plasma membrane through the involvement of vimentin in 3T3-L1 adipocytes. In conclusion, dreh is the first observed lncRNA that regulates glucose transport in adipocytes and could serve as a novel therapeutic target for diabetes by modulating adipocyte function. Considering the new function of dreh, we propose that dreh be renamed "down-regulated expression-related hexose/glucose transport enhancer."
Collapse
Affiliation(s)
- Nobuhiko Takahashi
- Department of Internal Medicine, School of Dentistry, Health Sciences University of Hokkaido, 1757 Kanazawa, Ishikari-Tobetsu, Hokkaido 061-0023, Japan.
| | - Atsushi P Kimura
- Department of Biological Sciences, Faculty of Science, Hokkaido University, Sapporo, Hokkaido 060-0810, Japan
| | - Kazumasa Ohmura
- Department of Internal Medicine, School of Dentistry, Health Sciences University of Hokkaido, 1757 Kanazawa, Ishikari-Tobetsu, Hokkaido 061-0023, Japan
| | - Sumiyoshi Naito
- Division of Clinical Laboratory, Health Sciences University of Hokkaido Hospital, 2-5 Ainosato, Kita-ku, Sapporo, Hokkaido 002-8072, Japan
| | - Mika Yoshida
- Division of Clinical Laboratory, Health Sciences University of Hokkaido Hospital, 2-5 Ainosato, Kita-ku, Sapporo, Hokkaido 002-8072, Japan
| | - Masahiro Ieko
- Department of Internal Medicine, School of Dentistry, Health Sciences University of Hokkaido, 1757 Kanazawa, Ishikari-Tobetsu, Hokkaido 061-0023, Japan
| |
Collapse
|
10
|
Zhang TN, Wang W, Yang N, Huang XM, Liu CF. Regulation of Glucose and Lipid Metabolism by Long Non-coding RNAs: Facts and Research Progress. Front Endocrinol (Lausanne) 2020; 11:457. [PMID: 32765426 PMCID: PMC7381111 DOI: 10.3389/fendo.2020.00457] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 06/10/2020] [Indexed: 12/17/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) are a type of non-coding RNA with a length that exceeds 200 nucleotides. Previous studies have shown that lncRNAs play an important role in the pathogenesis of various diseases. Research in both animal models and humans has begun to unravel the profound complexity of lncRNAs and demonstrated that lncRNAs exert direct effects on glucose and lipid metabolism both in vivo and in vitro. Such research has elucidated the regulatory role of lncRNAs in glucose and lipid metabolism in human disease. lncRNAs mediate glucose and lipid metabolism under physiological and pathological conditions and contribute to various metabolism disorders. This review provides an update on our understanding of the regulatory role of lncRNAs in glucose and lipid metabolism in various diseases. As our understanding of the function of lncRNAs improves, the future is promising for the development of new diagnostic biomarkers that utilize lncRNAs and treatments that target lncRNAs to improve clinical outcomes.
Collapse
Affiliation(s)
- Tie-Ning Zhang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
- *Correspondence: Tie-Ning Zhang
| | - Wei Wang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ni Yang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xin-Mei Huang
- Department of Endocrinology, the Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
- Department of Obstetrics and Gynecology, Yale University School of Medicine, New Haven, CT, United States
- Xin-Mei Huang
| | - Chun-Feng Liu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
- Chun-Feng Liu
| |
Collapse
|