1
|
Rasmi Y, Mohamed YA, Alipour S, Ahmed S, Abdelmajed SS. The role of miR-143/miR-145 in the development, diagnosis, and treatment of diabetes. J Diabetes Metab Disord 2024; 23:39-47. [PMID: 38932869 PMCID: PMC11196424 DOI: 10.1007/s40200-023-01317-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 09/14/2023] [Indexed: 06/28/2024]
Abstract
Objectives Diabetes mellitus [DM], is a multifaceted metabolic disease, which has become a worldwide threat to human wellness. Over the past decades, an enormous amount of attention has been devoted to understanding how microRNAs [miRNAs], a class of small non-coding RNA regulators of gene expression at the post-transcriptional level, are tied to DM pathology. It has been demonstrated that miRNAs control insulin synthesis, secretion, and activity. This review aims to provide an evaluation of the use of miR-143 and miR-145 as biomarkers for the diagnosis and prognosis of diabetes. Methods The use of miR-143 and miR-145 as biomarkers for the diagnosis and prognosis of diabetes has been studied, and research that examined this link was sought after in the literature. In addition, we will discuss the cellular and molecular pathways of insulin secretion regulation by miR-143/145 expression and finally their role in diabetes. Results In the current review, we emphasize recent findings on the miR-143/145 expression profiles as novel DM biomarkers in clinical studies and animal models and highlight recent discoveries on the complex regulatory effect and functional role of miR-143/145 expression in DM. Conclusion A novel clinical treatment that alters the expression and activity of miR-143/miR-145 may be able to return cells to their natural state of glucose homeostasis, demonstrating the value of using comprehensive miRNA profiles to predict the beginning of diabetes. Supplementary Information The online version contains supplementary material available at 10.1007/s40200-023-01317-y.
Collapse
Affiliation(s)
- Yousef Rasmi
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
- Cellular and Molecular Research Center, Cellular and Molecular Research Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Yara Ahmed Mohamed
- Faculty of Biotechnology, October University for Modern Sciences and Arts University [MSA], Giza, Egypt
| | - Shahriar Alipour
- Cellular and Molecular Research Center, Cellular and Molecular Research Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Salma Ahmed
- Faculty of Biotechnology, October University for Modern Sciences and Arts University [MSA], Giza, Egypt
| | - Samar Samir Abdelmajed
- Faculty of Dentistry- Medical Biochemistry and Genetics department, October University for Modern Sciences and Arts University [MSA], Giza, Egypt
| |
Collapse
|
2
|
Pereira G, Charpigny G, Guo Y, Silva E, Silva MF, Ye T, Lopes-da-Costa L, Humblot P. Characterization of circulating microRNA profiles of postpartum dairy cows with persistent subclinical endometritis. J Dairy Sci 2023; 106:9704-9717. [PMID: 37641364 DOI: 10.3168/jds.2023-23616] [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: 04/14/2023] [Accepted: 05/30/2023] [Indexed: 08/31/2023]
Abstract
Subclinical endometritis (SCE) is an unresolved inflammation of the endometrium of postpartum dairy cows, seriously affecting fertility. Current diagnosis, which relies on uterine cytology or even more invasive biopsy sampling, would benefit from the identification of blood-based diagnostic biomarkers. Due to the known role of microRNAs (miRNAs) in other diseases, this case-control study evaluated the cell-free circulating miRNA profiles of SCE cows, and the network of transcripts predicted to interact with those miRNAs, previously identified as differentially expressed genes (DEG) in the endometrium of the same cows. Healthy (H, n = 6) and persistent SCE (n = 11) cows characterized by endometrial cytology and biopsy were blood sampled at 21 and 44 d postpartum (DPP). Following extraction of cell-free plasma miRNAs and RNA-seq analysis, differential abundance analysis of miRNAs was performed with the DESeq2 R package (adjusted p-value of 0.05), and in silico prediction of miRNA-interacting genes on a sequence complementary basis was conducted using the miRWalk database. The principal component analysis showed a clear clustering between groups of uterine health phenotypes (H vs. SCE), although the clustering between groups was less pronounced at 44 DPP than at 21 DPP. No effect of the stage (21 vs. 44 DPP) was observed. A total of 799 known circulating miRNAs were identified, from which 34 demonstrated differential abundance between H and SCE cows (12 less abundant and 22 more abundant in SCE than in H cows). These 34 miRNAs are predicted to interact with 10,104 transcripts, among which 43, 81, and 147 were previously identified as differentially expressed in, respectively, endometrial luminal epithelial, glandular epithelial, and stromal cells of the same cows. This accounts for approximately half of the DEG identified between those H and SCE cows, including genes involved in endometrial cell proliferation, angiogenesis and immune response, whose dysregulation in SCE cows may impair pregnancy establishment. From 219 miRNAs with mean normalized read counts above 100, the presence and abundance of miR-425-3p and miR-2285z had the highest discriminatory level to differentiate SCE from H cows. In conclusion, despite apparent confinement to the endometrium, SCE is associated with a distinct circulating miRNA profile, which may represent a link between the systemic changes associated with disease and the endometrial immune response. The validation of a miRNA panel consisting of circulating cell-free miR-425-3p and miR-2285z may prove a relevant advancement for the noninvasive diagnosis of persistent SCE.
Collapse
Affiliation(s)
- Gonçalo Pereira
- CIISA-Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, 1300-477 Lisbon, Portugal; Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1300-477 Lisbon, Portugal
| | - Gilles Charpigny
- Université Paris-Saclay, INRAE, ENVA, BREED, 78350, Jouy-en-Josas, France
| | - Yongzhi Guo
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, SLU, PO, 750 07 Uppsala, Sweden
| | - Elisabete Silva
- CIISA-Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, 1300-477 Lisbon, Portugal; Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1300-477 Lisbon, Portugal
| | - Marta Filipa Silva
- CIISA-Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, 1300-477 Lisbon, Portugal; Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1300-477 Lisbon, Portugal; Faculty of Veterinary Medicine, Lusófona University, 1749-024 Lisbon, Portugal
| | - Tao Ye
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Institut National de la Santé et de la Recherche Médicale (INSERM) U1258, Centre National de Recherche Scientifique (CNRS) UMR7104, Université de Strasbourg,1 rue Laurent Fries, 67404 Illkirch, France
| | - Luís Lopes-da-Costa
- CIISA-Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, 1300-477 Lisbon, Portugal; Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1300-477 Lisbon, Portugal.
| | - Patrice Humblot
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, SLU, PO, 750 07 Uppsala, Sweden
| |
Collapse
|
3
|
Kamalumpundi V, Shams E, Tucker C, Cheng L, Peterson J, Thangavel S, Ofori O, Correia M. Mechanisms and pharmacotherapy of hypertension associated with type 2 diabetes. Biochem Pharmacol 2022; 206:115304. [DOI: 10.1016/j.bcp.2022.115304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 10/09/2022] [Accepted: 10/11/2022] [Indexed: 11/28/2022]
|
4
|
Regulatory Networks, Management Approaches, and Emerging Treatments of Nonalcoholic Fatty Liver Disease. Can J Gastroenterol Hepatol 2022; 2022:6799414. [PMID: 36397950 PMCID: PMC9666027 DOI: 10.1155/2022/6799414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 09/05/2022] [Indexed: 11/09/2022] Open
Abstract
The pathogenesis of NAFLD is complex and diverse, involving multiple signaling pathways and cytokines from various organs. Hepatokines, stellakines, adipokines, and myokines secreted by hepatocytes, hepatic stellate cells, adipose tissue, and myocytes play an important role in the occurrence and development of nonalcoholic fatty liver disease (NAFLD). The nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) contributes to the progression of NAFLD by mediating liver inflammation, immune response, hepatocyte death, and later compensatory proliferation. In this review, we first discuss the crosstalk and interaction between hepatokines, stellakines, adipokines, and myokines and NF-κB in NAFLD. The characterization of the crosstalk of NF-κB with these factors will provide a better understanding of the molecular mechanisms involved in the progression of NAFLD. In addition, we examine new expert management opinions for NAFLD and explore the therapeutic potential of silymarin in NAFLD/NASH.
Collapse
|
5
|
Pandey A, Ajgaonkar S, Jadhav N, Saha P, Gurav P, Panda S, Mehta D, Nair S. Current Insights into miRNA and lncRNA Dysregulation in Diabetes: Signal Transduction, Clinical Trials and Biomarker Discovery. Pharmaceuticals (Basel) 2022; 15:1269. [PMID: 36297381 PMCID: PMC9610703 DOI: 10.3390/ph15101269] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/27/2022] [Accepted: 10/09/2022] [Indexed: 01/24/2023] Open
Abstract
Diabetes is one of the most frequently occurring metabolic disorders, affecting almost one tenth of the global population. Despite advances in antihyperglycemic therapeutics, the management of diabetes is limited due to its complexity and associated comorbidities, including diabetic neuropathy, diabetic nephropathy and diabetic retinopathy. Noncoding RNAs (ncRNAs), including microRNAs (miRNAs) and long noncoding RNAs (lncRNAs), are involved in the regulation of gene expression as well as various disease pathways in humans. Several ncRNAs are dysregulated in diabetes and are responsible for modulating the expression of various genes that contribute to the 'symptom complex' in diabetes. We review various miRNAs and lncRNAs implicated in diabetes and delineate ncRNA biological networks as well as key ncRNA targets in diabetes. Further, we discuss the spatial regulation of ncRNAs and their role(s) as prognostic markers in diabetes. We also shed light on the molecular mechanisms of signal transduction with diabetes-associated ncRNAs and ncRNA-mediated epigenetic events. Lastly, we summarize clinical trials on diabetes-associated ncRNAs and discuss the functional relevance of the dysregulated ncRNA interactome in diabetes. This knowledge will facilitate the identification of putative biomarkers for the therapeutic management of diabetes and its comorbidities. Taken together, the elucidation of the architecture of signature ncRNA regulatory networks in diabetes may enable the identification of novel biomarkers in the discovery pipeline for diabetes, which may lead to better management of this metabolic disorder.
Collapse
Affiliation(s)
| | | | | | - Praful Saha
- Viridis Biopharma Pvt. Ltd., Mumbai 400 022, India
| | - Pranay Gurav
- Viridis Biopharma Pvt. Ltd., Mumbai 400 022, India
| | | | - Dilip Mehta
- Synergia Life Sciences Pvt. Ltd., Mumbai 400 022, India
| | - Sujit Nair
- Viridis Biopharma Pvt. Ltd., Mumbai 400 022, India
| |
Collapse
|
6
|
Nguyen MT, Min KH, Lee W. MiR-183-5p Induced by Saturated Fatty Acids Hinders Insulin Signaling by Downregulating IRS-1 in Hepatocytes. Int J Mol Sci 2022; 23:ijms23062979. [PMID: 35328400 PMCID: PMC8953084 DOI: 10.3390/ijms23062979] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/07/2022] [Accepted: 03/09/2022] [Indexed: 02/06/2023] Open
Abstract
Excessive saturated fatty acids (SFA) uptake is known to be a primary cause of obesity, a widely acknowledged risk factor of insulin resistance and type 2 diabetes. Although specific microRNAs (miRNAs) targeting insulin signaling intermediates are dysregulated by SFA, their effects on insulin signaling and sensitivity are largely unknown. Here, we investigated the role of SFA-induced miR-183-5p in the regulation of proximal insulin signaling molecules and the development of hepatic insulin resistance. HepG2 hepatocytes treated with palmitate and the livers of high-fat diet (HFD)-fed mice exhibited impaired insulin signaling resulting from dramatic reductions in the protein expressions of insulin receptor (INSR) and insulin receptor substrate-1 (IRS-1). Differential expression analysis showed the level of miR-183-5p, which tentatively targets the 3'UTR of IRS-1, was significantly elevated in palmitate-treated HepG2 hepatocytes and the livers of HFD-fed mice. Dual-luciferase analysis showed miR-183-5p bound directly to the 3'UTR of IRS-1 and reduced IRS-1 expression at the post-transcriptional stage. Moreover, transfection of HepG2 hepatocytes with miR-183-5p mimic significantly inhibited IRS-1 expression and hindered insulin signaling, consequently inhibiting insulin-stimulated glycogen synthesis. Collectively, this study reveals a novel mechanism whereby miR-183-5p induction by SFA impairs insulin signaling and suggests miR-183-5p plays a crucial role in the pathogenesis of hepatic insulin resistance in the background of obesity.
Collapse
Affiliation(s)
- Mai Thi Nguyen
- Department of Biochemistry, College of Medicine, Dongguk University, 123 Dongdae-ro, Gyeongju 38066, Korea; (M.T.N.); (K.-H.M.)
| | - Kyung-Ho Min
- Department of Biochemistry, College of Medicine, Dongguk University, 123 Dongdae-ro, Gyeongju 38066, Korea; (M.T.N.); (K.-H.M.)
| | - Wan Lee
- Department of Biochemistry, College of Medicine, Dongguk University, 123 Dongdae-ro, Gyeongju 38066, Korea; (M.T.N.); (K.-H.M.)
- Channelopathy Research Center, College of Medicine, Dongguk University, 32 Dongguk-ro, Ilsan Dong-gu, Goyang 10326, Korea
- Correspondence: ; Tel.: +82-54-770-2409
| |
Collapse
|
7
|
Zhang Y, Jin D, An X, Duan L, Duan Y, Lian F. Lychee Seed as a Potential Hypoglycemic Agent, and Exploration of its Underlying Mechanisms. Front Pharmacol 2021; 12:737803. [PMID: 34690773 PMCID: PMC8531476 DOI: 10.3389/fphar.2021.737803] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 09/06/2021] [Indexed: 12/12/2022] Open
Abstract
Food is people's primal want. A reasonable diet and healthy food not only provide nutrients for human growth but also contribute to disease prevention and treatment, while following an unhealthy diet can lead to an increased risk of many diseases, especially metabolic disorders, such as diabetes. Nature is enriched with different food sources, and it seems that purely natural products are more in line with the current concept of health, which enhance the formation of the notion that "Food/Diet Supplements from Natural Sources as a Medicine." As a delicious fruit, the medicinal values such as anticancer, antibacterial, antioxidation, and antiglycating properties of lychee have been found. Lychee (Litchi in Chinese) is a subtropical fruit plant belonging to the family Sapindaceae. It has been widely cultivated in warm climates worldwide, particularly in China, for thousands of years. In recent years, various phytochemical components such as quercetin, procyanidin A2, and (2R)-naringenin-7-O-(3-O-αL-rhamnopyranosyl-β-D-glucopyranoside) have been identified in a lychee seed, which may lend a lychee seed as a relatively safe and inexpensive adjuvant treatment for diabetes and diabetic complications. In fact, accumulating evidence has shown that lychee seed, lychee seed extracts, and related compounds have promising antihyperglycemic activities, including improving insulin resistance, anti-inflammatory effect, lipid regulation, neuroprotection, antineurotoxic effect, and renoprotection effect. In this review, we summarized publications on antiglycemic effects and mechanisms of lychee seed, lychee seed extracts, and related compounds, which included their efficacies as a cure for diabetes and diabetic complications in cells, animals, and humans, attempting to obtain a robust evidence basis for the clinical application and value of lychee seed.
Collapse
Affiliation(s)
- Yuehong Zhang
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - De Jin
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xuedong An
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Liyun Duan
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yingying Duan
- Clinical department of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Fengmei Lian
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
8
|
White MF, Kahn CR. Insulin action at a molecular level - 100 years of progress. Mol Metab 2021; 52:101304. [PMID: 34274528 PMCID: PMC8551477 DOI: 10.1016/j.molmet.2021.101304] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/07/2021] [Accepted: 07/13/2021] [Indexed: 12/15/2022] Open
Abstract
The discovery of insulin 100 years ago and its application to the treatment of human disease in the years since have marked a major turning point in the history of medicine. The availability of purified insulin allowed for the establishment of its physiological role in the regulation of blood glucose and ketones, the determination of its amino acid sequence, and the solving of its structure. Over the last 50 years, the function of insulin has been applied into the discovery of the insulin receptor and its signaling cascade to reveal the role of impaired insulin signaling-or resistance-in the progression of type 2 diabetes. It has also become clear that insulin signaling can impact not only classical insulin-sensitive tissues, but all tissues of the body, and that in many of these tissues the insulin signaling cascade regulates unexpected physiological functions. Despite these remarkable advances, much remains to be learned about both insulin signaling and how to use this molecular knowledge to advance the treatment of type 2 diabetes and other insulin-resistant states.
Collapse
Affiliation(s)
- Morris F White
- Boston Children's Hospital and Harvard Medical School, Boston, MA, 02215, USA.
| | - C Ronald Kahn
- Joslin Diabetes Center, Harvard Medical School, Boston, MA, 02215, USA.
| |
Collapse
|
9
|
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: 27] [Impact Index Per Article: 6.8] [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
|
10
|
Lu J, Pang L, Zhang B, Gong Z, Song C. Silencing circANKRD36 inhibits streptozotocin-induced insulin resistance and inflammation in diabetic rats by targeting miR-145 via XBP1. Inflamm Res 2021; 70:695-704. [PMID: 33978765 DOI: 10.1007/s00011-021-01467-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 04/19/2021] [Accepted: 04/28/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Diabetes mellitus (DM) is defined as a group of metabolic diseases characterized by hyperglycemia, which results from a deficiency in insulin secretion and/or insulin action. In diabetic patients, type 2 diabetes mellitus (T2DM) is in the majority. We explored the effects of circANKRD36 on streptozotocin (STZ)-induced insulin resistance and inflammation in diabetic rats with the aim of uncovering the underlying mechanism. METHODS STZ was used to induce the in vivo T2DM rat model. After circANKRD36 interference, blood glucose, insulin and adiponectin were respectively detected. Hematoxylin and eosin (H&E), enzyme-linked immunosorbent assay (ELISA) and terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling assay (TUNEL) were conducted to examine inflammation and apoptosis in T2DM rats, and western blot was used for detecting apoptosis-related proteins. The binding relationships among circANKRD36, miR-145 and XBP1 were examined by luciferase reporter assay. RESULTS Results showed that circANKRD36 was expressed at a high level in T2DM rats, while silencing circANKRD36 led to decreased blood glucose and insulin, accompanied by increased adiponectin level, and ameliorating insulin resistance. Silencing circANKRD36 alleviated the inflammation and suppressed cell apoptosis in the pancreatic tissues of T2DM rats, which was abated by miR-145 inhibitor. The binding of miR-145 to XBP1 was then confirmed. Additionally, miR-145 inhibitor increased the level of XBP1 in T2DM rats, which was decreased in the presence of circANKRD36 silencing. CONCLUSION This study is the first to prove that silencing circANKRD36 inhibits STZ-induced insulin resistance and inflammation in diabetic rats by targeting miR- 145 via XBP1. The results warrant the importance of circRNAs as drug target and thereby pave way for the development of newer therapeutic measures for T2DM.
Collapse
MESH Headings
- Animals
- Cytokines/blood
- Diabetes Mellitus, Experimental/blood
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/immunology
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Type 2/blood
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/immunology
- Diabetes Mellitus, Type 2/pathology
- Inflammation/genetics
- Insulin Resistance/genetics
- Male
- MicroRNAs
- Pancreas/metabolism
- Pancreas/pathology
- RNA, Circular
- Rats, Sprague-Dawley
- Up-Regulation
- X-Box Binding Protein 1/genetics
- X-Box Binding Protein 1/metabolism
- Rats
Collapse
Affiliation(s)
- Jinger Lu
- Department of Endocrinology, The Affiliated People's Hospital of Ningbo University, Ningbo, 315040, Zhejiang, China
| | - Linrong Pang
- Department of Chemoradiotherapy Centre, The Affiliated People's Hospital of Ningbo University, Ningbo, 315040, Zhejiang, China
| | - Bo Zhang
- Department of Infectious Disease, The Affiliated People's Hospital of Ningbo University, Ningbo, 315040, Zhejiang, China
| | - Zhigang Gong
- College of Physical Education, Jiangxi Normal University, Nanchang, 330022, Jiangxi, China
| | - Chunhui Song
- College of Life Sciences, Jiangxi Normal University, No. 99 Ziyang Avenue, Nanchang, 330022, Jiangxi, China.
| |
Collapse
|
11
|
Foudi N, Legeay S. Effects of physical activity on cell-to-cell communication during type 2 diabetes: A focus on miRNA signaling. Fundam Clin Pharmacol 2021; 35:808-821. [PMID: 33675090 DOI: 10.1111/fcp.12665] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 02/13/2021] [Accepted: 03/01/2021] [Indexed: 12/12/2022]
Abstract
Type 2 diabetes (TD2) is a progressive disease characterized by hyperglycemia that results from alteration in insulin secretion, insulin resistance, or both. A number of alterations involving different tissues and organs have been reported to the development and the progression of T2D, and more relevantly, through cell-to-cell communication pathways. Recent studies demonstrated that miRNAs are considerably implicated to cell-to-cell communication during T2D. Physical activity (PA) is associated with decreasing risks of developing T2D and acts as insulin-like factor. Cumulative evidence suggests that this effect could be mediated in part through improving insulin sensitivity in T2D and obese patients and modulating miRNAs synthesis and release in healthy patients. Therefore, the practice of PA should ideally be established before the initiation of T2D. This review describes cell-to-cell communications involved in the pathophysiology of T2D during PA.
Collapse
Affiliation(s)
- Nabil Foudi
- Department of Pharmacy, UNIV Angers, Angers, France.,Faculty of Medicine, Department of Pharmacy, University Ferhat Abbas Setif 1, Setif, Algeria
| | - Samuel Legeay
- MINT, UNIV Angers, INSERM 1066, CNRS 6021, IRIS-IBS-CHU, Angers, France
| |
Collapse
|
12
|
Lan S, Albinsson S. Regulation of IRS-1, insulin signaling and glucose uptake by miR-143/145 in vascular smooth muscle cells. Biochem Biophys Res Commun 2020; 529:119-125. [PMID: 32560812 DOI: 10.1016/j.bbrc.2020.05.148] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 05/20/2020] [Indexed: 11/25/2022]
Abstract
Regulation of insulin signaling by microRNAs in smooth muscle cells may contribute to diabetic vascular disease. The two smooth muscle enriched miRNAs miR-143 and miR-145 have been reported to target mediators of insulin signaling in non-smooth muscle cells. In this study, we aimed to determine the importance of this regulation in vascular smooth muscle cells, where expression of miR-143/145 is much higher than in other cell types. Smooth muscle cells deficient of the miR-143/145 cluster were used, as well as smooth muscle cells transfected with mimics/inhibitors for either miR-143 or miR-145. We found that deletion of miR-143/145 in smooth muscle results in a dramatic upregulation IRS-1 expression and insulin signaling, and an increased insulin-induced glucose uptake. Furthermore, specific modulation of either miR-145 or miR-143 expression regulated specific targets (IRS-1, ORP8 and the IGF-1 receptor) in the insulin signaling pathway. Consequently, transient inhibition or overexpression of either miR-143 or miR-145 was sufficient to regulate insulin signaling in smooth muscle cells. In conclusion, the results of this study support an important role for both miR-143 and miR-145 in the regulation of insulin signaling and glucose uptake in vascular smooth muscle cells.
Collapse
MESH Headings
- Animals
- Biological Transport, Active
- Cells, Cultured
- Glucose/metabolism
- Insulin/metabolism
- Insulin Receptor Substrate Proteins/metabolism
- Mice
- Mice, Knockout
- MicroRNAs/antagonists & inhibitors
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/metabolism
- Receptor, IGF Type 1/metabolism
- Signal Transduction
Collapse
Affiliation(s)
- Susan Lan
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | |
Collapse
|
13
|
Kaur P, Kotru S, Singh S, Behera BS, Munshi A. Role of miRNAs in the pathogenesis of T2DM, insulin secretion, insulin resistance, and β cell dysfunction: the story so far. J Physiol Biochem 2020; 76:485-502. [PMID: 32749641 DOI: 10.1007/s13105-020-00760-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Accepted: 07/29/2020] [Indexed: 01/24/2023]
Abstract
Diabetes, the most common endocrine disorder, also known as a silent killer disease, is characterized by uncontrolled hyperglycemia. According to the International Diabetes Federation, there were 451 million people with diabetes mellitus worldwide in 2017. It is a multifactorial syndrome caused by genetic as well as environmental factors. Noncoding RNAs, especially the miRNAs, play a significant role in the development as well as the progression of the disease. This is on account of insulin resistance or defects in β cell function. Various miRNAs including miR-7, miR-9, miR-16, miR-27, miR-24, miR-29, miR-124a, miR-135, miR-130a, miR-144, miR-181a, and miR-375 and many more have been associated with insulin resistance and other pathogenic conditions leading to the development of the disease. These miRNAs play significant roles in various pathways underlying insulin resistance such as PI3K, AKT/GSK, and mTOR. The main target genes of these miRNAs are FOXO1, FOXA2, STAT3, and PTEN. The miRNAs carry out important functions in insulin target tissues like the adipose tissue, liver, and muscle. MiRNAs miR-9, miR-375, and miR-124a, are also associated with the secretion of insulin from pancreatic cells. There is an interplay between the miRNAs and pancreatic cell growth, especially the miRNAs affecting development and proliferation of these cells. Most of the miRNAs target more than one gene which not only justifies their use as biomarkers but also their therapeutic potential. The current review has been compiled with an aim to discuss the role of various miRNAs involved in various pathogenic mechanisms including insulin resistance, insulin secretion, and the β cell dysfunction.
Collapse
Affiliation(s)
- Prabhsimran Kaur
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, 151001, India
| | - Sushil Kotru
- Max Endocrinology, Diabetes and Obesity Care Centre, Max Superspeciality Hospital, Bathinda, 151001, India
| | - Sandeep Singh
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, 151001, India
| | - Bidwan Sekhar Behera
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, 151001, India
| | - Anjana Munshi
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, 151001, India.
| |
Collapse
|
14
|
Statin Treatment-Induced Development of Type 2 Diabetes: From Clinical Evidence to Mechanistic Insights. Int J Mol Sci 2020; 21:ijms21134725. [PMID: 32630698 PMCID: PMC7369709 DOI: 10.3390/ijms21134725] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 06/29/2020] [Accepted: 06/30/2020] [Indexed: 12/17/2022] Open
Abstract
Statins are the gold-standard treatment for the prevention of primary and secondary cardiovascular disease, which is the leading cause of mortality worldwide. Despite the safety and relative tolerability of statins, observational studies, clinical trials and meta-analyses indicate an increased risk of developing new-onset type 2 diabetes mellitus (T2DM) after long-term statin treatment. It has been shown that statins can impair insulin sensitivity and secretion by pancreatic β-cells and increase insulin resistance in peripheral tissues. The mechanisms involved in these processes include, among others, impaired Ca2+ signaling in pancreatic β-cells, down-regulation of GLUT-4 in adipocytes and compromised insulin signaling. In addition, it has also been described that statins’ impact on epigenetics may also contribute to statin-induced T2DM via differential expression of microRNAs. This review focuses on the evidence and mechanisms by which statin therapy is associated with the development of T2DM. This review describes the multifactorial combination of effects that most likely contributes to the diabetogenic effects of statins. Clinically, these findings should encourage clinicians to consider diabetes monitoring in patients receiving statin therapy in order to ensure early diagnosis and appropriate management.
Collapse
|
15
|
Li W, Luo C, Xie X, Xiao Y, Zhao F, Cai J, Zhou X, Zeng T, Fu B, Wu Y, Xiao X, Liu S. Identification of key genes and pathways in syphilis combined with diabetes: a bioinformatics study. AMB Express 2020; 10:83. [PMID: 32342229 PMCID: PMC7186291 DOI: 10.1186/s13568-020-01009-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 04/07/2020] [Indexed: 12/29/2022] Open
Abstract
We noticed that syphilis patients seem to be more susceptible to diabetes and the lesions often involve the kidneys, but the pathogenesis is not yet completely understood. In this study, microarray analysis was performed to investigate the dysregulated expressed genes (DEGs) in rabbit model of syphilis combined with diabetes. A total of 1045 genes were identified to be significantly differentially expressed, among which 571 were up-regulated and 474 were down-regulated (≥ 2.0fold, p < 0.05). Using the database visualization and integration discovery for the Kyoto Encyclopedia of Gene and Genome (KEGG) pathway enrichment analysis. The downregulated DEGs were significantly enriched for biosynthesis of antibiotics, carbon metabolism and protein digestion, while the upregulated DEGs were mainly enriched for cancer and PI3K-Akt signaling pathway. Molecular Complex Detection (MCODE) plugins were used to visualize protein–protein interaction (PPI) network of DEGs and Screening for hub genes and gene modules. ALB, FN1, CASP3, MMP9, IL8, CTGF, STAT3, IGF1, VCAM-1 and HGF were filtrated as the hub genes according to the degree of connectivity from the PPI network. To the best of our knowledge, this study is the first to comprehensively identify the expression patterns of dysregulated genes in syphilis combined with diabetes, providing a basis for revealing the underlying pathogenesis of syphilis combined with diabetes and exploring the goals of therapeutic intervention.
Collapse
|
16
|
Wen F, Xia Q, Zhang H, Shia H, Rajesh A, Wu Y, Yang Y, Yang Z. Resistin Activates p65 Pathway and Reduces Glycogen Content through Keratin 8. Int J Endocrinol 2020; 2020:9767926. [PMID: 32508919 PMCID: PMC7251471 DOI: 10.1155/2020/9767926] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 02/28/2020] [Accepted: 04/11/2020] [Indexed: 01/27/2023] Open
Abstract
Resistin is associated with metabolic syndrome and inflammatory conditions. Many studies have suggested that resistin inhibits the accumulation of glycogen; however, the exact mechanisms of resistin-induced decrease in glycogen content remain unclear. Keratin 8 is a typical epithelial intermediate filament protein, but numerous studies suggest a vital role of K8 in glucose metabolism. However, it is still not known whether K8 participates in the mediation of resistin-induced reduction of cellular glycogen accumulation. In this study, we found that resistin upregulated expression of the p65 subunit of NF-κB, which led to the promotion of K8 transcriptional expression; in turn, the expression of K8 inhibited glycogen accumulation in HepG2 cells.
Collapse
Affiliation(s)
- Fengyun Wen
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, Henan, China
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, Hubei, China
| | - Qiao Xia
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, Hubei, China
| | - Hui Zhang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, Henan, China
| | - Haipeng Shia
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, Henan, China
| | - Amin Rajesh
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, 36849-5501 Auburn, Alabama, USA
| | - Yanling Wu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, Hubei, China
| | - Yi Yang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, Hubei, China
| | - Zaiqing Yang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, Henan, China
| |
Collapse
|
17
|
Ghasemi A, Hashemy SI, Azimi-Nezhad M, Dehghani A, Saeidi J, Mohtashami M. The cross-talk between adipokines and miRNAs in health and obesity-mediated diseases. Clin Chim Acta 2019; 499:41-53. [PMID: 31476303 DOI: 10.1016/j.cca.2019.08.028] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 08/28/2019] [Accepted: 08/28/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND Multiple studies have revealed a direct correlation between obesity and the development of multiple comorbidities, including metabolic diseases, cardiovascular disorders, chronic inflammatory disease, and cancers. However, the molecular mechanism underlying the link between obesity and the progression of these diseases is not completely understood. Adipokines are factors that are secreted by adipocytes and play a key role in whole body homeostasis. Collaboratively, miRNAs are suggested to have key functions in the development of obesity and obesity-related disorders. Based on recently emerging evidence, obesity leads to the dysregulation of both adipokines and obesity-related miRNAs. In the present study, we described the correlations between obesity and its related diseases that are mediated by the mutual regulatory effects of adipokines and miRNAs. METHODS We reviewed current knowledge of the modulatory effects of adipokines on miRNAs activity and their relevant functions in pathological conditions and vice versa. RESULTS Our research reveals the ability of adipokines and miRNAs to control the expression and activity of the other class of molecules, and their effects on obesity-related diseases. CONCLUSIONS This study may help researchers develop a roadmap for future investigations and provide opportunities to develop new therapeutic and diagnostic methods for treating obesity-related diseases.
Collapse
Affiliation(s)
- Ahmad Ghasemi
- Non-communicable Disease Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran.
| | - Seyed Isaac Hashemy
- Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Mohsen Azimi-Nezhad
- Non-communicable Disease Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran; UMR INSERM U 1122, IGE-PCV, Interactions Gène-Environment en Physiopathologie Cardiovascular Université de Lorraine, France
| | - Alireza Dehghani
- Institute of Biochemistry and Molecular Biology, University of Bonn, Bonn, Germany
| | - Jafar Saeidi
- Department of Physiology, School of Basic Science, Neyshabur Branch, Islamic Azad University, Neyshabur, Iran
| | - Mahnaz Mohtashami
- Department of Biology, School of Basic Science, Neyshabur Branch, Islamic Azad University, Neyshabur, Iran
| |
Collapse
|
18
|
MicroRNA-34a and MicroRNA-181a Mediate Visfatin-Induced Apoptosis and Oxidative Stress via NF-κB Pathway in Human Osteoarthritic Chondrocytes. Cells 2019; 8:cells8080874. [PMID: 31405216 PMCID: PMC6721672 DOI: 10.3390/cells8080874] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 07/31/2019] [Accepted: 08/09/2019] [Indexed: 01/16/2023] Open
Abstract
Current evidence suggests a complex interaction between adipokines and microRNA (miRNA) in osteoarthritis (OA) pathogenesis. The present study explored the role of miR-34a and miR-181a in regulating apoptosis and oxidative stress induced by visfatin in human OA chondrocytes. Chondrocytes were transfected with miR-34a and miR-181a inhibitors and stimulated with visfatin for 24 h, in the presence of nuclear factor (NF)-κB inhibitor (BAY-11-7082, 2 h pre-incubation). Apoptosis and reactive oxygen species (ROS) production were detected by cytometry, miRNA, antioxidant enzymes, nuclear factor erythroid (NRF)2 and B-cell lymphoma (BCL)2 expressions by quantitative real time polymerase chain reaction (real time PCR) and western blot. P50 NF-κB subunit was measured by immunofluorescence. Visfatin significantly induced apoptosis and superoxide anion production, increased miR-34a, miR-181a, superoxide dismutase (SOD)-2, catalase (CAT), NRF2 and decreased BCL2 gene and protein expression in OA chondrocytes. All the visfatin-caused effects were suppressed by using miR-34a and miR-181a inhibitors. Pre-incubation with BAY-11-7082 counteracted visfatin-induced expression of miRNA, BCL2, SOD-2, CAT and NRF2. Inhibition of miR-34a and miR-181a significantly reduced the activation of p50 NF-κB. Visfatin confirms its ability to induce apoptosis and oxidative stress in human OA chondrocytes; these effects appeared mediated by miR-34a and miR-181a via NF-κB pathway. We highlight the relevance of visfatin as potential therapeutic target for OA treatment.
Collapse
|
19
|
Liu SX, Zheng F, Xie KL, Xie MR, Jiang LJ, Cai Y. Exercise Reduces Insulin Resistance in Type 2 Diabetes Mellitus via Mediating the lncRNA MALAT1/MicroRNA-382-3p/Resistin Axis. MOLECULAR THERAPY-NUCLEIC ACIDS 2019; 18:34-44. [PMID: 31479923 PMCID: PMC6726922 DOI: 10.1016/j.omtn.2019.08.002] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 07/24/2019] [Accepted: 08/01/2019] [Indexed: 12/13/2022]
Abstract
Insulin resistance (IR) is the primary pathological mechanism underlying type 2 diabetes mellitus (T2DM). Here, the study aimed to ascertain whether and how exercise mediates IR in T2DM. An in vivo mouse model of high-fat diet-induced IR and an in vitro high-glucose-induced IR model were constructed. High long non-coding RNA (lncRNA) metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) expression was detected in T2MD and was positively correlated with HOMA-IR and resistin levels. Then, short hairpin RNA targeting MALAT1 (sh-MALAT1) or pcDNA-MALAT1 was delivered into human umbilical vein endothelial cells (HUVECs) to knock down or upregulate its expression, respectively. Silencing of MALAT1 resulted in reduced levels of resistin, Ang II, tumor necrosis factor alpha (TNF-α), interleukin-6 (IL-6), soluble intercellular adhesion molecule-1 (sICAM-1), soluble vascular cell adhesion molecule-1 (sVCAM-1), endothelin-1 (ET-1), and p-insulin receptor substrate-1 (p-IRS)/ISR-1, and decreased cell migration, as well as enhanced glucose uptake and levels of nitric oxide (NO) and p-Akt/Akt. In the IR mouse model, exercise was observed to downregulate MALAT1 to reduce resistin, whereby exercise reduced homeostatic model assessment-insulin resistance (HOMA-IR). Besides, exercise also elevated microRNA-382-3p (miR-382-3p) expression in the serum of IR mice. Dual-luciferase reporter and RNA binding protein immunoprecipitation (RIP) assays identified that MALAT1 could bind to miR-382-3p to upregulate resistin. Collectively, the key observations of the study provide evidence that inhibition of MALAT1 elevates miR-382-3p to repress resistin, which consequently underlies the mechanism of exercise protecting against IR, highlighting a direction for T2DM therapy development.
Collapse
Affiliation(s)
- Sui-Xin Liu
- Cardiac Rehabilitation Center, Department of Rehabilitation, Xiangya Hospital, Central South University, Changsha 410008, P.R. China
| | - Fan Zheng
- Cardiac Rehabilitation Center, Department of Rehabilitation, Xiangya Hospital, Central South University, Changsha 410008, P.R. China
| | - Kang-Ling Xie
- Cardiac Rehabilitation Center, Department of Rehabilitation, Xiangya Hospital, Central South University, Changsha 410008, P.R. China
| | - Mu-Rong Xie
- Cardiac Rehabilitation Center, Department of Rehabilitation, Xiangya Hospital, Central South University, Changsha 410008, P.R. China
| | - Ling-Jun Jiang
- Cardiac Rehabilitation Center, Department of Rehabilitation, Xiangya Hospital, Central South University, Changsha 410008, P.R. China
| | - Ying Cai
- Cardiac Rehabilitation Center, Department of Rehabilitation, Xiangya Hospital, Central South University, Changsha 410008, P.R. China.
| |
Collapse
|
20
|
Ebrahimi R, Bahiraee A, Niazpour F, Emamgholipour S, Meshkani R. The role of microRNAs in the regulation of insulin signaling pathway with respect to metabolic and mitogenic cascades: A review. J Cell Biochem 2019; 120:19290-19309. [PMID: 31364207 DOI: 10.1002/jcb.29299] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 06/27/2019] [Indexed: 12/18/2022]
Abstract
Insulin resistance (IR) is a shared pathological condition among type 2 diabetes, obesity, cardiovascular disease, and other metabolic disorders. It is growing significantly all over the world and consequently, a substantial effort is needed for developing the potential novel diagnostics and therapeutics. An insulin signaling pathway is tightly modulated by different mechanisms including the epigenetic modifications. Today, a deal of great attention has been shifted towards the regulatory role of noncoding RNAs on target proteins of the insulin signaling pathway. Noncoding RNAs are a major area of the epigenetics which control gene expression at the posttranscriptional levels and include a large class of microRNAs (miRNAs). With this in view, many studies have implicated the mediatory effects of miRNAs on the downstream metabolic and mitogenic proteins of the insulin signaling pathway. Since providing new biomarkers for the early diagnosis of IR and related metabolic traits are very significant, we intended to review the possible role of miRNAs in the regulation of the insulin signaling pathway, with a primary focus on the downstream target proteins of the metabolic and mitogenic cascades.
Collapse
Affiliation(s)
- Reyhane Ebrahimi
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Students' Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Bahiraee
- Department of Medical Genetics, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Farshad Niazpour
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Solaleh Emamgholipour
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Meshkani
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
21
|
Gao J, Feng X, Wang F, Wang J, Wang H, Li H, Zhang W, Hao L, Shi Z. microRNA‐448 inhibits the progression of non‐small–cell lung cancer through regulating IRS2. J Cell Biochem 2019; 120:13453-13463. [PMID: 30912183 DOI: 10.1002/jcb.28619] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 01/21/2019] [Accepted: 01/24/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Jie Gao
- Department of CT Affiliated Hospital of Hebei University of Engineering Handan Hebei China
| | | | - Fang Wang
- Department of Respiration Affiliated Hospital of Hebei University of Engineering Handan Hebei China
| | - Junlan Wang
- Department of CT The Second Hospital of Handan City Handan Hebei China
| | - Hongmei Wang
- Staff Hospital of Handan Iron and Steel Group Company Handan Hebei China
| | - Hua Li
- Department of Neurology Affiliated Hospital of Hebei University of Engineering Handan Hebei China
| | - Weiwei Zhang
- Handan Maternal and Child Health Hospital Handan China
| | - Lijun Hao
- Staff Hospital of Handan Iron and Steel Group Company Handan Hebei China
| | - Zhiming Shi
- Department of Pathology Affiliated Hospital of Hebei University of Engineering Handan Hebei China
| |
Collapse
|
22
|
Kong J, Qiu Y, Li Y, Zhang H, Wang W. TGF-β1 elevates P-gp and BCRP in hepatocellular carcinoma through HOTAIR/miR-145 axis. Biopharm Drug Dispos 2019; 40:70-80. [DOI: 10.1002/bdd.2172] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 01/13/2019] [Accepted: 01/23/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Jiehong Kong
- Center for Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences; Soochow University; Suzhou 215123 China
| | - Yajing Qiu
- Center for Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences; Soochow University; Suzhou 215123 China
| | - Yuan Li
- Center for Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences; Soochow University; Suzhou 215123 China
| | - Hongjian Zhang
- Center for Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences; Soochow University; Suzhou 215123 China
| | - Weipeng Wang
- Center for Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences; Soochow University; Suzhou 215123 China
| |
Collapse
|
23
|
Nunez Lopez YO, Retnakaran R, Zinman B, Pratley RE, Seyhan AA. Predicting and understanding the response to short-term intensive insulin therapy in people with early type 2 diabetes. Mol Metab 2019; 20:63-78. [PMID: 30503831 PMCID: PMC6358589 DOI: 10.1016/j.molmet.2018.11.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 11/05/2018] [Accepted: 11/12/2018] [Indexed: 12/23/2022] Open
Abstract
OBJECTIVE Short-term intensive insulin therapy (IIT) early in the course of type 2 diabetes acutely improves beta-cell function with long-lasting effects on glycemic control. However, conventional measures cannot determine which patients are better suited for IIT, and little is known about the molecular mechanisms determining response. Therefore, this study aimed to develop a model that could accurately predict the response to IIT and provide insight into molecular mechanisms driving such response in humans. METHODS Twenty-four patients with early type 2 diabetes were assessed at baseline and four weeks after IIT, consisting of basal detemir and premeal insulin aspart. Twelve individuals had a beneficial beta-cell response to IIT (responders) and 12 did not (nonresponders). Beta-cell function was assessed by multiple methods, including Insulin Secretion-Sensitivity Index-2. MicroRNAs (miRNAs) were profiled in plasma samples before and after IIT. The response to IIT was modeled using a machine learning algorithm and potential miRNA-mediated regulatory mechanisms assessed by differential expression, correlation, and functional network analyses (FNA). RESULTS Baseline levels of circulating miR-145-5p, miR-29c-3p, and HbA1c accurately (91.7%) predicted the response to IIT (OR = 121 [95% CI: 6.7, 2188.3]). Mechanistically, a previously described regulatory loop between miR-145-5p and miR-483-3p/5p, which controls TP53-mediated apoptosis, appears to also occur in our study population of humans with early type 2 diabetes. In addition, significant (fold change > 2, P < 0.05) longitudinal changes due to IIT in the circulating levels of miR-138-5p, miR-192-5p, miR-195-5p, miR-320b, and let-7a-5p further characterized the responder group and significantly correlated (|r| > 0.4, P < 0.05) with the changes in measures of beta-cell function and insulin sensitivity. FNA identified a network of coordinately/cooperatively regulated miRNA-targeted genes that potentially drives the IIT response through negative regulation of apoptotic processes that underlie beta cell dysfunction and concomitant positive regulation of proliferation. CONCLUSIONS Responses to IIT in people with early type 2 diabetes are associated with characteristic miRNA signatures. This study represents a first step to identify potential responders to IIT (a current limitation in the field) and provides important insight into the pathophysiologic determinants of the reversibility of beta-cell dysfunction. ClinicalTrial.gov identifier: NCT01270789.
Collapse
Affiliation(s)
- Yury O Nunez Lopez
- Translational Research Institute for Metabolism and Diabetes, Florida Hospital, Orlando, FL 32804, USA
| | - Ravi Retnakaran
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Bernard Zinman
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Richard E Pratley
- Translational Research Institute for Metabolism and Diabetes, Florida Hospital, Orlando, FL 32804, USA.
| | - Attila A Seyhan
- Translational Research Institute for Metabolism and Diabetes, Florida Hospital, Orlando, FL 32804, USA; The Chemical Engineering Department, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
24
|
Jasinski-Bergner S, Kielstein H. Adipokines Regulate the Expression of Tumor-Relevant MicroRNAs. Obes Facts 2019; 12:211-225. [PMID: 30999294 PMCID: PMC6547259 DOI: 10.1159/000496625] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 01/05/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Increasing prevalence of obesity requires the investigation of respective comorbidities, including tumor diseases like colorectal, renal, post-menopausal breast, prostate cancer, and leukemia. To date, molecular mechanisms of the malignant transformation of these peripheral tissues induced by obesity remain unclear. Adipose tissue secretes factors with hormone-like functions, the adipokines, and is therefore categorized as an endocrine organ. Current research demonstrates the ability of adipose tissue to alter DNA methylation and gene expression in peripheral tissues, probably affecting microRNA (miR) expression. METHODS Literature was analyzed for adipokine-regulated miRs. Many of these adipokine upregulated or downregulated miRs exert either oncogenic or anti-tumoral potential. RESULTS The three selected and analyzed adipokines, adiponectin, leptin, and resistin, induce more strongly oncogenic miRs and simultaneously reduce anti-tumoral miRs than vice versa. This effect is not only true for the pure number of regulated miRs, it is also the case by consideration of the abundance of the respective miR expression based on actual data sets derived from next-generation sequencing. CONCLUSION The link of obesity and cancer is analyzed under the aspect of adipokine-regulated miRs. At the same time the impact of miR abundance is considered as a regulatory variable. This context offers new strategies for tumor therapy and diagnostics.
Collapse
Affiliation(s)
- Simon Jasinski-Bergner
- Department of Anatomy and Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany,
| | - Heike Kielstein
- Department of Anatomy and Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| |
Collapse
|
25
|
A New Insight into the Roles of MiRNAs in Metabolic Syndrome. BIOMED RESEARCH INTERNATIONAL 2018; 2018:7372636. [PMID: 30648107 PMCID: PMC6311798 DOI: 10.1155/2018/7372636] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 11/28/2018] [Indexed: 12/13/2022]
Abstract
Metabolic syndrome (MetS), which includes several clinical components such as abdominal obesity, insulin resistance (IR), dyslipidemia, microalbuminuria, hypertension, proinflammatory state, and oxidative stress (OS), has become a global epidemic health issue contributing to a high risk of type 2 diabetes mellitus (T2DM). In recent years, microRNAs (miRNAs), used as noninvasive biomarkers for diagnosis and therapy, have aroused global interest in complex processes in health and diseases, including MetS and its components. MiRNAs can exist stably in serum, liver, skeletal muscle (SM), heart muscle, adipose tissue (AT), and βcells, because of their ability to escape the digestion of RNase. Here we first present an overall review on recent findings of the relationship between miRNAs and several main components of MetS, such as IR, obesity, diabetes, lipid metabolism, hypertension, hyperuricemia, and stress, to illustrate the targeting proteins or relevant pathways that are involved in the progress of MetS and also help us find promising novel diagnostic and therapeutic strategies.
Collapse
|
26
|
MicroRNAs as Regulators of Insulin Signaling: Research Updates and Potential Therapeutic Perspectives in Type 2 Diabetes. Int J Mol Sci 2018; 19:ijms19123705. [PMID: 30469501 PMCID: PMC6321520 DOI: 10.3390/ijms19123705] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 11/08/2018] [Accepted: 11/17/2018] [Indexed: 12/21/2022] Open
Abstract
The insulin signaling pathway is composed of a large number of molecules that positively or negatively modulate insulin specific signal transduction following its binding to the cognate receptor. Given the importance of the final effects of insulin signal transduction, it is conceivable that many regulators are needed in order to tightly control the metabolic or proliferative functional outputs. MicroRNAs (miRNAs) are small non-coding RNA molecules that negatively modulate gene expression through their specific binding within the 3′UTR sequence of messenger RNA (mRNA), thus causing mRNA decoy or translational inhibition. In the last decade, miRNAs have been addressed as pivotal cellular rheostats which control many fundamental signaling pathways, including insulin signal transduction. Several studies demonstrated that multiple alterations of miRNAs expression or function are relevant for the development of insulin resistance in type 2 diabetes (T2D); such alterations have been highlighted in multiple insulin target organs including liver, muscles, and adipose tissue. Indirectly, miRNAs have been identified as modulators of inflammation-derived insulin resistance, by controlling/tuning the activity of innate immune cells in insulin target tissues. Here, we review main findings on miRNA functions as modulators of insulin signaling in physiologic- or in T2D insulin resistance- status. Additionally, we report the latest hypotheses of prospective therapies involving miRNAs as potential targets for future drugs in T2D.
Collapse
|
27
|
Cardoso AL, Fernandes A, Aguilar-Pimentel JA, de Angelis MH, Guedes JR, Brito MA, Ortolano S, Pani G, Athanasopoulou S, Gonos ES, Schosserer M, Grillari J, Peterson P, Tuna BG, Dogan S, Meyer A, van Os R, Trendelenburg AU. Towards frailty biomarkers: Candidates from genes and pathways regulated in aging and age-related diseases. Ageing Res Rev 2018; 47:214-277. [PMID: 30071357 DOI: 10.1016/j.arr.2018.07.004] [Citation(s) in RCA: 315] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 07/08/2018] [Accepted: 07/10/2018] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Use of the frailty index to measure an accumulation of deficits has been proven a valuable method for identifying elderly people at risk for increased vulnerability, disease, injury, and mortality. However, complementary molecular frailty biomarkers or ideally biomarker panels have not yet been identified. We conducted a systematic search to identify biomarker candidates for a frailty biomarker panel. METHODS Gene expression databases were searched (http://genomics.senescence.info/genes including GenAge, AnAge, LongevityMap, CellAge, DrugAge, Digital Aging Atlas) to identify genes regulated in aging, longevity, and age-related diseases with a focus on secreted factors or molecules detectable in body fluids as potential frailty biomarkers. Factors broadly expressed, related to several "hallmark of aging" pathways as well as used or predicted as biomarkers in other disease settings, particularly age-related pathologies, were identified. This set of biomarkers was further expanded according to the expertise and experience of the authors. In the next step, biomarkers were assigned to six "hallmark of aging" pathways, namely (1) inflammation, (2) mitochondria and apoptosis, (3) calcium homeostasis, (4) fibrosis, (5) NMJ (neuromuscular junction) and neurons, (6) cytoskeleton and hormones, or (7) other principles and an extensive literature search was performed for each candidate to explore their potential and priority as frailty biomarkers. RESULTS A total of 44 markers were evaluated in the seven categories listed above, and 19 were awarded a high priority score, 22 identified as medium priority and three were low priority. In each category high and medium priority markers were identified. CONCLUSION Biomarker panels for frailty would be of high value and better than single markers. Based on our search we would propose a core panel of frailty biomarkers consisting of (1) CXCL10 (C-X-C motif chemokine ligand 10), IL-6 (interleukin 6), CX3CL1 (C-X3-C motif chemokine ligand 1), (2) GDF15 (growth differentiation factor 15), FNDC5 (fibronectin type III domain containing 5), vimentin (VIM), (3) regucalcin (RGN/SMP30), calreticulin, (4) PLAU (plasminogen activator, urokinase), AGT (angiotensinogen), (5) BDNF (brain derived neurotrophic factor), progranulin (PGRN), (6) α-klotho (KL), FGF23 (fibroblast growth factor 23), FGF21, leptin (LEP), (7) miRNA (micro Ribonucleic acid) panel (to be further defined), AHCY (adenosylhomocysteinase) and KRT18 (keratin 18). An expanded panel would also include (1) pentraxin (PTX3), sVCAM/ICAM (soluble vascular cell adhesion molecule 1/Intercellular adhesion molecule 1), defensin α, (2) APP (amyloid beta precursor protein), LDH (lactate dehydrogenase), (3) S100B (S100 calcium binding protein B), (4) TGFβ (transforming growth factor beta), PAI-1 (plasminogen activator inhibitor 1), TGM2 (transglutaminase 2), (5) sRAGE (soluble receptor for advanced glycosylation end products), HMGB1 (high mobility group box 1), C3/C1Q (complement factor 3/1Q), ST2 (Interleukin 1 receptor like 1), agrin (AGRN), (6) IGF-1 (insulin-like growth factor 1), resistin (RETN), adiponectin (ADIPOQ), ghrelin (GHRL), growth hormone (GH), (7) microparticle panel (to be further defined), GpnmB (glycoprotein nonmetastatic melanoma protein B) and lactoferrin (LTF). We believe that these predicted panels need to be experimentally explored in animal models and frail cohorts in order to ascertain their diagnostic, prognostic and therapeutic potential.
Collapse
|
28
|
Luan B, Sun C. MiR-138-5p affects insulin resistance to regulate type 2 diabetes progression through inducing autophagy in HepG2 cells by regulating SIRT1. Nutr Res 2018; 59:90-98. [PMID: 30442237 DOI: 10.1016/j.nutres.2018.05.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 04/28/2018] [Accepted: 05/06/2018] [Indexed: 01/22/2023]
Abstract
Insulin resistance (IR) is considered as a major factor of type 2 diabetes (T2D), which is seriously detrimental to human health. In our present study, we found that the expression of miR-138-5p was increased in the insulin-resistant HepG2 cells induced by TNF-α. Therefore, we hypothesized that miR-138-5p might play a regulatory role in the IR. To examine this hypothesis, HepG2 cells were transfected with miR-138-5p inhibitor. Silencing of miR-138-5p increased glucose uptake and glycogen synthesis of TNF-α-stimulated HepG2 cells and decreased glucose concentration in medium, suggesting that downregulation of miR-138-5p suppressed IR in HepG2 cells. Besides that, we found that sirtuin 1 (SIRT1) was the target gene of the miR-138-5p. Moreover, co-transfection with SIRT1-siRNA and miR-138-5p inhibitor suppressed glucose uptake and glycogen synthesis of HepG2 cells compared with miR-138-5p inhibitor-transfected group, indicating that downregulation of SIRT1 weakened the inhibitory effect of miR-138-5p inhibitor on IR. In addition, overexpressed SIRT1 increased Beclin1, LC3 II/I level, and the number of GFP-LC3 dots and decreased p62 level, whereas downregulation of SIRT1 had the opposite effects. Our results demonstrated that overexpressed SIRT1 activated autophagy in HepG2 cells. Moreover, we observed that 3-methyladenine (an inhibitor of autophagy) treatment decreased the high glucose uptake and glycogen synthesis of miR-138-5p inhibitor-transfected HepG2 cells, suggesting that the inhibition of autophagy abolished the inhibitory effect of miR-138-5p inhibitor on IR in HepG2 cells. Taken together, this study suggested that miR-138-5p contributed to the TNF-α-induced IR, possibly through inducing autophagy in HepG2 cells by regulating SIRT1. MiR-138-5p might be a potential and promising target for the treatment of IR.
Collapse
Affiliation(s)
- Bingguo Luan
- Department of Endocrinology and metabolism, Yantaishan Hospital of Yantai, Shandong, 264000, China.
| | - Caixia Sun
- Department of Endocrinology and metabolism, Yantaishan Hospital of Yantai, Shandong, 264000, China
| |
Collapse
|
29
|
Li R, Shen Q, Wu N, He M, Liu N, Huang J, Lu B, Yao Q, Yang Y, Hu R. MiR-145 improves macrophage-mediated inflammation through targeting Arf6. Endocrine 2018; 60:73-82. [PMID: 29388044 DOI: 10.1007/s12020-018-1521-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 01/03/2018] [Indexed: 12/18/2022]
Abstract
PURPOSE To explore the relationship between miR-145 and ADP ribosylation factor 6 (Arf6) in regulating macrophage-mediated inflammation. METHODS THP-1 cells were induced by 160 nM of phorbol 12-myristate 13-acetate (PMA) for 48 h to differentiate to macrophages and then were treated with LPS (100 ng/ml) for 8 h to simulate chronic metabolic inflammation in vitro. Dual-luciferase reporter assay was performed. MiR-145 siRNA and LV-ARF6-RNAi were used to up or down regulate miR-145 and Arf6 expression in THP-1 cells, respectively. Omental adipose tissue from patients in surgical ward were collected to detect the expression of miR-145, Arf6 and production of proinflammatory cytokines. Patients were divided into three groups according to their body mass index and history of diabetes. RESULTS Dual-luciferase reporter assays showed the direct down-regulation of Arf6 by miR-145. Forty-eight-hour-transfection of miR-145 inhibitor resulted in significant increase of Arf6, IL-1beta, TNF-alpha and IL-6 as well as phosphorylation of p65 in NF-kappaB pathway in THP-1 cells, which, inversely, were reversed by overexpressing miR-145. In addition, down-regulation of Arf6 in macrophages reduced expression and secretion of cytokines. Expression of miR-145 was found to be attenuated in the omental adipose tissue of obese patients and diabetics with greater Arf6 expression, confirming the role of miR-145 in regulating macrophage-mediated inflammation targeting Arf6. CONCLUSIONS By means of reducing the expression of Arf6 and subsequent signal transduction via NF-kappaB, miR-145 plays a role in inhibiting the secretion of inflammatory factors and then improving the inflammatory status. MiR-145 might be one of the candidates for anti-inflammatory treatment for metabolic diseases.
Collapse
Affiliation(s)
- Rumei Li
- Department of Endocrinology, Huashan Hospital, Fudan University, Shanghai, 200040, China
- Institute of Endocrinology and Diabetology, Fudan University, Shanghai, 200040, China
| | - Qiwei Shen
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Nan Wu
- Department of Endocrinology, Huashan Hospital, Fudan University, Shanghai, 200040, China
- Institute of Endocrinology and Diabetology, Fudan University, Shanghai, 200040, China
| | - Min He
- Department of Endocrinology, Huashan Hospital, Fudan University, Shanghai, 200040, China
- Institute of Endocrinology and Diabetology, Fudan University, Shanghai, 200040, China
| | - Naijia Liu
- Department of Endocrinology, Huashan Hospital, Fudan University, Shanghai, 200040, China
- Institute of Endocrinology and Diabetology, Fudan University, Shanghai, 200040, China
| | - Jinya Huang
- Department of Endocrinology, Huashan Hospital, Fudan University, Shanghai, 200040, China
- Institute of Endocrinology and Diabetology, Fudan University, Shanghai, 200040, China
| | - Bin Lu
- Department of Endocrinology, Huashan Hospital, Fudan University, Shanghai, 200040, China
- Institute of Endocrinology and Diabetology, Fudan University, Shanghai, 200040, China
| | - Qiyuan Yao
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Yehong Yang
- Department of Endocrinology, Huashan Hospital, Fudan University, Shanghai, 200040, China.
- Institute of Endocrinology and Diabetology, Fudan University, Shanghai, 200040, China.
| | - Renming Hu
- Department of Endocrinology, Huashan Hospital, Fudan University, Shanghai, 200040, China.
- Institute of Endocrinology and Diabetology, Fudan University, Shanghai, 200040, China.
| |
Collapse
|
30
|
Mirra P, Nigro C, Prevenzano I, Leone A, Raciti GA, Formisano P, Beguinot F, Miele C. The Destiny of Glucose from a MicroRNA Perspective. Front Endocrinol (Lausanne) 2018; 9:46. [PMID: 29535681 PMCID: PMC5834423 DOI: 10.3389/fendo.2018.00046] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Glucose serves as a primary, and for some tissues the unique, fuel source in order to generate and maintain the biological functions. Hyperglycemia is a hallmark of type 2 diabetes and is the direct consequence of perturbations in the glucose homeostasis. Insulin resistance, referred to as a reduced response of target tissues to the hormone, contributes to the development of hyperglycemia. The molecular mechanisms responsible for the altered glucose homeostasis are numerous and not completely understood. MicroRNAs (miRNAs) are now recognized as regulators of the lipid and glucose metabolism and are involved in the onset of metabolic diseases. Indeed, these small non-coding RNA molecules operate in the RNA silencing and posttranscriptional regulation of gene expression and may modulate the levels of kinases and enzymes in the glucose metabolism. Therefore, a better characterization of the function of miRNAs and a deeper understanding of their role in disease may represent a fundamental step toward innovative treatments addressing the causes, not only the symptoms, of hyperglycemia, using approaches aimed at restoring either miRNAs or their specific targets. In this review, we outline the current understanding regarding the impact of miRNAs in the glucose metabolism and highlight the need for further research focused on altered key kinases and enzymes in metabolic diseases.
Collapse
Affiliation(s)
- Paola Mirra
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale “Gaetano Salvatore” - CNR, Naples, Italy
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Cecilia Nigro
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale “Gaetano Salvatore” - CNR, Naples, Italy
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Immacolata Prevenzano
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale “Gaetano Salvatore” - CNR, Naples, Italy
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Alessia Leone
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale “Gaetano Salvatore” - CNR, Naples, Italy
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Gregory Alexander Raciti
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale “Gaetano Salvatore” - CNR, Naples, Italy
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Pietro Formisano
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale “Gaetano Salvatore” - CNR, Naples, Italy
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Francesco Beguinot
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale “Gaetano Salvatore” - CNR, Naples, Italy
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Claudia Miele
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale “Gaetano Salvatore” - CNR, Naples, Italy
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
- *Correspondence: Claudia Miele,
| |
Collapse
|
31
|
Lupini L, Pepe F, Ferracin M, Braconi C, Callegari E, Pagotto S, Spizzo R, Zagatti B, Lanuti P, Fornari F, Ghasemi R, Mariani-Costantini R, Bolondi L, Gramantieri L, Calin GA, Sabbioni S, Visone R, Veronese A, Negrini M. Over-expression of the miR-483-3p overcomes the miR-145/TP53 pro-apoptotic loop in hepatocellular carcinoma. Oncotarget 2017; 7:31361-71. [PMID: 27120784 PMCID: PMC5058762 DOI: 10.18632/oncotarget.8913] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 04/10/2016] [Indexed: 01/01/2023] Open
Abstract
The miR-145-5p, which induces TP53-dependent apoptosis, is down-regulated in several tumors, including hepatocellular carcinomas (HCCs), but some HCCs show physiological expression of this miR. Here we demonstrate that in HCC cells carrying wild-type TP53 the steady activation of the miR-145 signaling selects clones resistant to apoptosis via up-regulation of the oncogenic miR-483-3p. Expression of the miR-145-5p and of the miR-483-3p correlated negatively in non-neoplastic liver (n=41; ρ=−0.342, P=0.028), but positively in HCCs (n=21; ρ=0.791, P<0.0001), which we hypothesized to be due to impaired glucose metabolism in HCCs versus normal liver. In fact, when liver cancer cells were grown in low glucose, miR-145-5p lowered miR-483-3p expression, allowing apoptosis, whereas when cells were grown in high glucose the levels of miR-483-3p increased, reducing the apoptotic rate. This indicates that depending on glucose availability the miR-145-5p has double effects on the miR-483-3p, either inhibitory or stimulatory. Moreover, resistance to apoptosis in clones overexpressing both miR-145-5p and miR-483-3p was abrogated by silencing the miR-483-3p. Our data highlight a novel mechanism of resistance to apoptosis in liver cancer cells harbouring wild type TP53 and suggest a potential role of miR-145-5p and miR-483-3p as druggable targets in a subset of HCCs.
Collapse
Affiliation(s)
- Laura Lupini
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Felice Pepe
- Unit of General Pathology, Aging Research Center (Ce.S.I.), G. d'Annunzio University Foundation, Chieti, Italy.,Department of Medical, Oral and Biotechnological Sciences, G. d'Annunzio University, Chieti, Italy
| | - Manuela Ferracin
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Chiara Braconi
- Division of Cancer Therapeutics, Institute of Cancer Research, London, UK
| | - Elisa Callegari
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Sara Pagotto
- Unit of General Pathology, Aging Research Center (Ce.S.I.), G. d'Annunzio University Foundation, Chieti, Italy.,Department of Medical, Oral and Biotechnological Sciences, G. d'Annunzio University, Chieti, Italy
| | | | - Barbara Zagatti
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Paola Lanuti
- Department of Medicine and Aging Science, G. d'Annunzio University, Chieti, Italy
| | | | - Reza Ghasemi
- Unit of General Pathology, Aging Research Center (Ce.S.I.), G. d'Annunzio University Foundation, Chieti, Italy
| | - Renato Mariani-Costantini
- Unit of General Pathology, Aging Research Center (Ce.S.I.), G. d'Annunzio University Foundation, Chieti, Italy.,Department of Medical, Oral and Biotechnological Sciences, G. d'Annunzio University, Chieti, Italy
| | - Luigi Bolondi
- S.Orsola-Malpighi University Hospital, Bologna, Italy
| | | | - George A Calin
- Department of Experimental Therapeutics, MD Anderson Medical Centre, Houston, TX, USA
| | - Silvia Sabbioni
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Rosa Visone
- Unit of General Pathology, Aging Research Center (Ce.S.I.), G. d'Annunzio University Foundation, Chieti, Italy.,Department of Medical, Oral and Biotechnological Sciences, G. d'Annunzio University, Chieti, Italy
| | - Angelo Veronese
- Unit of General Pathology, Aging Research Center (Ce.S.I.), G. d'Annunzio University Foundation, Chieti, Italy.,Department of Medical, Oral and Biotechnological Sciences, G. d'Annunzio University, Chieti, Italy
| | - Massimo Negrini
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| |
Collapse
|
32
|
Saeedi Borujeni MJ, Esfandiary E, Taheripak G, Codoñer‐Franch P, Alonso‐Iglesias E, Mirzaei H. Molecular aspects of diabetes mellitus: Resistin, microRNA, and exosome. J Cell Biochem 2017; 119:1257-1272. [PMID: 28688216 DOI: 10.1002/jcb.26271] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 07/07/2017] [Indexed: 12/16/2022]
Affiliation(s)
- Mohammad Javad Saeedi Borujeni
- Department of Anatomical SCIENCES and Molecular BiologySchool of MedicineIsfahan University of Medical SciencesIsfahanIran
| | - Ebrahim Esfandiary
- Department of Anatomical SCIENCES and Molecular BiologySchool of MedicineIsfahan University of Medical SciencesIsfahanIran
| | - Gholamreza Taheripak
- Faculty of MedicineDepartment of BiochemistryIran University of Medical SciencesTehranIran
| | - Pilar Codoñer‐Franch
- Department of PediatricsObstetrics and GynecologyUniversity of ValenciaValenciaSpain
| | | | - Hamed Mirzaei
- Department of Medical BiotechnologySchool of MedicineMashhad University of Medical SciencesMashhadIran
| |
Collapse
|
33
|
Zhang Y, Sun X, Icli B, Feinberg MW. Emerging Roles for MicroRNAs in Diabetic Microvascular Disease: Novel Targets for Therapy. Endocr Rev 2017. [DOI: 10.1210/er.2016-1122.2017.1.test] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
34
|
Zhang Y, Sun X, Icli B, Feinberg MW. Emerging Roles for MicroRNAs in Diabetic Microvascular Disease: Novel Targets for Therapy. Endocr Rev 2017; 38:145-168. [PMID: 28323921 PMCID: PMC5460677 DOI: 10.1210/er.2016-1122] [Citation(s) in RCA: 135] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 02/13/2017] [Indexed: 12/11/2022]
Abstract
Chronic, low-grade systemic inflammation and impaired microvascular function are critical hallmarks in the development of insulin resistance. Accordingly, insulin resistance is a major risk factor for type 2 diabetes and cardiovascular disease. Accumulating studies demonstrate that restoration of impaired function of the diabetic macro- and microvasculature may ameliorate a range of cardiovascular disease states and diabetes-associated complications. In this review, we focus on the emerging role of microRNAs (miRNAs), noncoding RNAs that fine-tune target gene expression and signaling pathways, in insulin-responsive tissues and cell types important for maintaining optimal vascular homeostasis and preventing the sequelae of diabetes-induced end organ injury. We highlight current pathophysiological paradigms of miRNAs and their targets involved in regulating the diabetic microvasculature in a range of diabetes-associated complications such as retinopathy, nephropathy, wound healing, and myocardial injury. We provide an update of the potential use of circulating miRNAs diagnostically in type I or type II diabetes. Finally, we discuss emerging delivery platforms for manipulating miRNA expression or function as the next frontier in therapeutic intervention to improve diabetes-associated microvascular dysfunction and its attendant clinical consequences.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Medicine, Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115
- Department of Pharmacology and Pharmacy, University of Hong Kong, Pokfulam, Hong Kong SAR, China, and
| | - Xinghui Sun
- Department of Medicine, Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, Nebraska 68588
| | - Basak Icli
- Department of Medicine, Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Mark W. Feinberg
- Department of Medicine, Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115
| |
Collapse
|
35
|
Ling Q, Xie H, Li J, Liu J, Cao J, Yang F, Wang C, Hu Q, Xu X, Zheng S. Donor Graft MicroRNAs: A Newly Identified Player in the Development of New-onset Diabetes After Liver Transplantation. Am J Transplant 2017; 17:255-264. [PMID: 27458792 PMCID: PMC5215980 DOI: 10.1111/ajt.13984] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 07/17/2016] [Accepted: 07/18/2016] [Indexed: 01/25/2023]
Abstract
New-onset diabetes after liver transplantation (NODALT) is a frequent complication with an unfavorable outcome. We previously demonstrated a crucial link between donor graft genetics and the risk of NODALT. We selected 15 matched pairs of NODALT and non-NODALT liver recipients using propensity score matching analysis. The donor liver tissues were tested for the expression of 10 microRNAs (miRNAs) regulating human hepatic glucose homeostasis. The biological functions of potential target genes were predicted using gene ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis. Both miR-103 and miR-181a were significantly highly expressed in the NODALT group as compared to the non-NODALT group. The predicted target genes (e.g. Irs2, Pik3r1, Akt2, and Gsk3b) were involved in glucose import and the insulin signaling pathway. We also observed dysregulation of miRNAs (e.g. let-7, miR-26b, miR-145, and miR-183) in cultured human hepatocytes treated with tacrolimus or high glucose, the two independent risk factors of NODALT identified in this cohort. The hepatic miRNA profiles altered by tacrolimus or hyperglycemia were associated with insulin resistance and glucose homeostatic imbalance as revealed by enrichment analysis. The disease susceptibility miRNA expressive pattern could be imported directly from the donor and consolidated by the transplant factors.
Collapse
Affiliation(s)
- Q. Ling
- Department of SurgeryCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesFirst Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhouChina,Key Lab of Combined Multi‐Organ TransplantationMinistry of Public HealthHangzhouChina
| | - H. Xie
- Key Lab of Combined Multi‐Organ TransplantationMinistry of Public HealthHangzhouChina
| | - J. Li
- Department of SurgeryCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesFirst Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhouChina
| | - J. Liu
- Department of SurgeryCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesFirst Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhouChina,Department of Pathology and Molecular MedicineFaculty of Health SciencesMcMaster UniversityHamiltonOntarioCanada
| | - J. Cao
- Key Lab of Combined Multi‐Organ TransplantationMinistry of Public HealthHangzhouChina
| | - F. Yang
- Department of SurgeryCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesFirst Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhouChina
| | - C. Wang
- Department of SurgeryCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesFirst Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhouChina
| | - Q. Hu
- Key Lab of Combined Multi‐Organ TransplantationMinistry of Public HealthHangzhouChina
| | - X. Xu
- Department of SurgeryCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesFirst Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhouChina,Key Lab of Combined Multi‐Organ TransplantationMinistry of Public HealthHangzhouChina
| | - S. Zheng
- Department of SurgeryCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesFirst Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhouChina,Key Lab of Combined Multi‐Organ TransplantationMinistry of Public HealthHangzhouChina
| |
Collapse
|
36
|
Tian Y, Mok MTS, Yang P, Cheng ASL. Epigenetic Activation of Wnt/β-Catenin Signaling in NAFLD-Associated Hepatocarcinogenesis. Cancers (Basel) 2016; 8:E76. [PMID: 27556491 PMCID: PMC4999785 DOI: 10.3390/cancers8080076] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 08/01/2016] [Accepted: 08/15/2016] [Indexed: 02/07/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD), characterized by fat accumulation in liver, is closely associated with central obesity, over-nutrition and other features of metabolic syndrome, which elevate the risk of developing hepatocellular carcinoma (HCC). The Wnt/β-catenin signaling pathway plays a significant role in the physiology and pathology of liver. Up to half of HCC patients have activation of Wnt/β-catenin signaling. However, the mutation frequencies of CTNNB1 (encoding β-catenin protein) or other antagonists targeting Wnt/β-catenin signaling are low in HCC patients, suggesting that genetic mutations are not the major factor driving abnormal β-catenin activities in HCC. Emerging evidence has demonstrated that obesity-induced metabolic pathways can deregulate chromatin modifiers such as histone deacetylase 8 to trigger undesired global epigenetic changes, thereby modifying gene expression program which contributes to oncogenic signaling. This review focuses on the aberrant epigenetic activation of Wnt/β-catenin in the development of NAFLD-associated HCC. A deeper understanding of the molecular mechanisms underlying such deregulation may shed light on the identification of novel druggable epigenetic targets for the prevention and/or treatment of HCC in obese and diabetic patients.
Collapse
Affiliation(s)
- Yuan Tian
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.
| | - Myth T S Mok
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China.
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen 518057, China.
| | - Pengyuan Yang
- Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.
| | - Alfred S L Cheng
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China.
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen 518057, China.
- State Key Laboratory of Digestive Disease and Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
37
|
Su CM, Huang CY, Tang CH. Characteristics of resistin in rheumatoid arthritis angiogenesis. Biomark Med 2016; 10:651-60. [PMID: 26867862 DOI: 10.2217/bmm.15.125] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Adipokines have been reported to be involved in the regulation of various physiological processes, including the immune response. Rheumatoid arthritis (RA) is an example of a systemic immune disease that causes chronic inflammation of the synovium and bone destruction in the joint. Recent therapeutic strategies based on the understanding of the role of cytokines and cellular mechanisms in RA have improved our understanding of angiogenesis. On the other hand, endogenous endothelial progenitor cells, which are a population isolated from peripheral blood monocytes have recently been identified as a homing target for pro-angiogeneic factor and vessel formation. In this review, we summarize the effects of common adipokines, such as adiponectin, leptin and resistin in RA pathogenesis and discuss other potential mechanisms of relevance for the therapeutic treatment of RA.
Collapse
Affiliation(s)
- Chen-Ming Su
- Department of Biomedical Sciences Laboratory, Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, Zhejiang, China.,Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Chun-Yin Huang
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan.,Department of Orthopedic Surgery, China Medical University Beigang Hospital, Yun-Lin County, Taiwan
| | - Chih-Hsin Tang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan.,Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan.,Department of Biotechnology, College of Health Science, Asia University, Taichung, Taiwan
| |
Collapse
|
38
|
Hu X, Chi L, Zhang W, Bai T, Zhao W, Feng Z, Tian H. Down-regulation of the miR-543 alleviates insulin resistance through targeting the SIRT1. Biochem Biophys Res Commun 2015; 468:781-7. [PMID: 26562529 DOI: 10.1016/j.bbrc.2015.11.032] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 11/05/2015] [Indexed: 11/24/2022]
Abstract
Insulin resistance plays an important role in the development of hypertension, which is seriously detrimental to human health. Recently, Sirtuin-1 (SIRT1) has been found to participate in regulation of insulin resistance. Therefore, further studies focused on the SIRT1 regulators might provide a potential approach for combating insulin resistance and hypertension. Interestingly, in this study, we found that SIRT1 was the target gene of the miR-543 by the Dual-Luciferase Reporter Assay. Moreover, the miR-543 expression notably increased in the insulin-resistant HepG2 cells induced by TNF-α. Further analysis showed that the overexpression of the miR-543 lowered the SIRT1 mRNA and protein levels, resulting in the insulin resistance in the HepG2 cells; the inhibition of miR-543, however, enhanced the mRNA and protein expression of the SIRT1, and alleviated the insulin resistance. Furthermore, the SIRT1 overexpression abrogated the effect of miR-543 on insulin resistance. In addition, the overexpression of the miR-543 by the lentivirus-mediated gene transfer markedly impaired the insulin signaling assessed by the Western blot analysis of the glycogen synthesis and the phosphorylation of Akt and GSK3β. In summary, our study suggested that the downregulation of the miR-543 could alleviate the insulin resistance via the modulation of the SIRT1 expression, which might be a potential new strategy for treating insulin resistance and a promising therapeutic method for hypertension.
Collapse
Affiliation(s)
- Xiaojing Hu
- Departments of Cardiology, Ninth Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, 710054, China
| | - Liyi Chi
- Departments of Neurology, The 451st Hospital of People's Liberation Army, Xi'an, 710054, China
| | - Wentao Zhang
- Departments of Osteology, Xi'an Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710000, China
| | - Tiao Bai
- Departments of Gereology, Ninth Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, 710054, China
| | - Wei Zhao
- Departments of Cardiology, Ninth Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, 710054, China
| | - Zhanbin Feng
- Departments of Cardiology, Ninth Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, 710054, China
| | - Hongyan Tian
- Departments of Peripheral Angiopathy, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277 Yanta West Road, Xi'an, 710061, China.
| |
Collapse
|
39
|
Das AV, Pillai RM. Implications of miR cluster 143/145 as universal anti-oncomiRs and their dysregulation during tumorigenesis. Cancer Cell Int 2015; 15:92. [PMID: 26425114 PMCID: PMC4588501 DOI: 10.1186/s12935-015-0247-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 09/22/2015] [Indexed: 02/07/2023] Open
Abstract
Tumorigenesis is a multistep process, de-regulated due to the imbalance of oncogenes as well as anti-oncogenes, resulting in disruption of tissue homeostasis. In many cases the effect of oncogenes and anti-oncogenes are mediated by various other molecules such as microRNAs. microRNAs are small non-coding RNAs established to post-transcriptionally regulate more than half of the protein coding genes. miR cluster 143/145 is one such cancer-related microRNA cluster which is down-regulated in most of the cancers and is able to hinder tumorigenesis by targeting tumor-associated genes. The fact that they could sensitize drug-resistant cancer cells by targeting multidrug resistant genes makes them potent tools to target cancer cells. Their low levels precede events which lead to cancer progression and therefore could be considered also as biomarkers to stage the disease. Interestingly, evidence suggests the existence of several in vivo mechanisms by which this cluster is differentially regulated at the molecular level to keep their levels low in cancer. In this review, we summarize the roles of miR cluster 143/145 in cancer, their potential prognostic applications and also their regulation during tumorigenesis.
Collapse
Affiliation(s)
- Ani V Das
- Cancer Research Program-9, Rajiv Gandhi Centre for Biotechnology, Thycaud.P.O., Thiruvananthapuram-14, Kerala India
| | - Radhakrishna M Pillai
- Cancer Research Program-9, Rajiv Gandhi Centre for Biotechnology, Thycaud.P.O., Thiruvananthapuram-14, Kerala India
| |
Collapse
|
40
|
MicroRNAs as regulators of metabolic disease: pathophysiologic significance and emerging role as biomarkers and therapeutics. Int J Obes (Lond) 2015; 40:88-101. [PMID: 26311337 PMCID: PMC4722234 DOI: 10.1038/ijo.2015.170] [Citation(s) in RCA: 240] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 07/01/2015] [Accepted: 07/22/2015] [Indexed: 12/13/2022]
Abstract
The prevalence of overweight and obesity in developed and developing countries has greatly increased the risk of insulin resistance and type 2 diabetes mellitus. It is evident from human and animal studies that obesity alters microRNA (miRNA) expression in metabolically important organs, and that miRNAs are involved in changes to normal physiology, acting as mediators of disease. miRNAs regulate multiple pathways including insulin signaling, immune-mediated inflammation, adipokine expression, adipogenesis, lipid metabolism, and food intake regulation. Thus, miRNA-based therapeutics represent an innovative and attractive treatment modality, with non-human primate studies showing great promise. In addition, miRNA measures in plasma or bodily fluids may be used as disease biomarkers and predictors of metabolic disease in humans. This review analyzes the role of miRNAs in obesity and insulin resistance, focusing on the miR-17/92, miR-143-145, miR-130, let-7, miR-221/222, miR-200, miR-223, miR-29 and miR-375 families, as well as miRNA changes by relevant tissue (adipose, liver and skeletal muscle). Further, the current and future applications of miRNA-based therapeutics and diagnostics in metabolic disease are discussed.
Collapse
|
41
|
Shyu KG, Cheng WP, Wang BW. Angiotensin II Downregulates MicroRNA-145 to Regulate Kruppel-like Factor 4 and Myocardin Expression in Human Coronary Arterial Smooth Muscle Cells under High Glucose Conditions. Mol Med 2015; 21:616-25. [PMID: 26181633 DOI: 10.2119/molmed.2015.00041] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 07/13/2015] [Indexed: 01/26/2023] Open
Abstract
MicroRNA (miR)-145 is the most abundant miR in vascular smooth muscle cells (VSMCs). However, the effect of hyperglycemia on the regulation of miR-145 is unknown. We hypothesized that the hyperglycemic condition activates a proinflammatory response that mediates the expression of miR-145 in VSMCs. We investigated whether miR-145 serves as a critical regulator to regulate the downstream proliferation factors (including Kruppel-like factor 4 [Klf4] and myocardin) in VSMCs under hyperglycemic conditions. Human coronary artery smooth muscle cells (HCASMCs) were cultured under high glucose conditions. Sustained high glucose at 25 mmol/L significantly decreased the expression of miR-145 in HCASMCs. High glucose significantly increased angiotensin II (Ang II) secretion from HCASMCs and Ang II suppressed miR-145 expression in HCASMCs. Ang II repression of miR145 expression resulted in increased Klf4 and decreased myocardin expression under conditions of high glucose. Overexpression of miR-145 significantly decreased Klf4 and increased myocardin expression and inhibited HCASMC proliferation and migration induced by a high glucose state. Balloon injury of the carotid artery in diabetic rats was performed to investigate miR-145, Klf and myocardin expression. The expression of miR-145 was maximally increased at 7 d after carotid injury and gradually declined thereafter. Overexpression of miR-145 and treatment with valsartan reversed Klf4 and myocardin protein expression induced by balloon injury and improved vascular injury. In conclusion, our study reveals that Ang II downregulates miR-145 to regulate Klf4 and myocardin expression in HCASMCs under high glucose conditions. Ang II plays a critical role in the regulation of miR-145 under hyperglycemic conditions.
Collapse
Affiliation(s)
- Kou-Gi Shyu
- Division of Cardiology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan.,Graduate Institute of Clinical Medicine, Taipei Medical University, Taipei, Taiwan
| | - Wen-Ping Cheng
- Department of Medical Education and Research, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Bao-Wei Wang
- Department of Medical Education and Research, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| |
Collapse
|
42
|
Midbrain dopamine neurons in Parkinson's disease exhibit a dysregulated miRNA and target-gene network. Brain Res 2015; 1618:111-21. [PMID: 26047984 DOI: 10.1016/j.brainres.2015.05.021] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2015] [Revised: 05/09/2015] [Accepted: 05/15/2015] [Indexed: 11/21/2022]
Abstract
The degeneration of substantia nigra (SN) dopamine (DA) neurons in sporadic Parkinson׳s disease (PD) is characterized by disturbed gene expression networks. Micro(mi)RNAs are post-transcriptional regulators of gene expression and we recently provided evidence that these molecules may play a functional role in the pathogenesis of PD. Here, we document a comprehensive analysis of miRNAs in SN DA neurons and PD, including sex differences. Our data show that miRNAs are dysregulated in disease-affected neurons and differentially expressed between male and female samples with a trend of more up-regulated miRNAs in males and more down-regulated miRNAs in females. Unbiased Ingenuity Pathway Analysis (IPA) revealed a network of miRNA/target-gene associations that is consistent with dysfunctional gene and signaling pathways in PD pathology. Our study provides evidence for a general association of miRNAs with the cellular function and identity of SN DA neurons, and with deregulated gene expression networks and signaling pathways related to PD pathogenesis that may be sex-specific.
Collapse
|
43
|
Su CM, Hsu CJ, Tsai CH, Huang CY, Wang SW, Tang CH. Resistin Promotes Angiogenesis in Endothelial Progenitor Cells Through Inhibition of MicroRNA206: Potential Implications for Rheumatoid Arthritis. Stem Cells 2015; 33:2243-55. [PMID: 25828083 DOI: 10.1002/stem.2024] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 03/11/2015] [Indexed: 12/15/2022]
Abstract
Endothelial progenitor cells (EPCs) promote angiogenesis and are therefore key contributors to a wide variety of angiogenesis-related autoimmune diseases such as rheumatoid arthritis (RA). However, the signaling mechanisms through which these progenitor cells influence RA pathogenesis remain unknown. The aim of this study was to examine whether resistin plays a role in the pathogenesis of and angiogenesis associated with RA by circulating EPCs. We found that levels of resistin in synovial fluid and tissue from patients with RA and from mice with collagen-induced arthritis were overexpressed and promoted the homing of EPCs into the synovium, thereby inducing angiogenesis. EPCs isolated from healthy donors were used to investigate the signal transduction pathway underlying EPC migration and tube formation after treatment with resistin. We found that resistin directly induced a significant increase in expression of vascular endothelial growth factor (VEGF) in EPCs. We also found that the expression of microRNA-206 (miR-206) was negatively correlated with the expression of resistin during EPC-mediated angiogenesis. Notably, the increased expression of VEGF was associated with decreased binding of miR-206 to the VEGF-A 3' untranslated region through protein kinase C delta-dependent AMP-activated protein kinase signaling pathway. Moreover, blockade of resistin reduced EPC homing into synovial fluid and angiogenesis in vivo. Taken together, our study is the first to demonstrate that resistin promotes EPCs homing into the synovium during RA angiogenesis via a signal transduction pathway that involves VEGF expression in primary EPCs. These findings provide support for resistin as a therapeutic target for the patients with RA. Stem Cells 2015;33:2243-2255.
Collapse
Affiliation(s)
- Chen-Ming Su
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Chin-Jung Hsu
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan.,Department of Orthopedic Surgery, China Medical University, Taichung, Taiwan
| | - Chun-Hao Tsai
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan.,Department of Orthopedic Surgery, China Medical University, Taichung, Taiwan
| | - Chun-Yin Huang
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan.,Department of Orthopedic Surgery, China Medical University Beigang Hospital, Yun-Lin County, Taiwan
| | - Shih-Wei Wang
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
| | - Chih-Hsin Tang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan.,Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan.,Department of Biotechnology, College of Health Science, Asia University, Taichung, Taiwan
| |
Collapse
|
44
|
Wen F, Zhang H, Bao C, Yang M, Wang N, Zhang J, Hu Y, Yang X, Geng J, Yang Z. Resistin Increases Ectopic Deposition of Lipids Through miR-696 in C2C12 Cells. Biochem Genet 2015; 53:63-71. [DOI: 10.1007/s10528-015-9672-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2015] [Accepted: 04/04/2015] [Indexed: 12/19/2022]
|