1
|
Vohra MS, Ahmad B, Taylor ER, Benchoula K, Fong IL, Parhar IS, Ogawa S, Serpell CJ, Wong EH. 5,7,3',4',5'-pentamethoxyflavone (PMF) exhibits anti-obesity and neuroprotective effects in an obese zebrafish model. Mol Cell Endocrinol 2025; 604:112554. [PMID: 40252912 DOI: 10.1016/j.mce.2025.112554] [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: 01/14/2025] [Revised: 04/14/2025] [Accepted: 04/17/2025] [Indexed: 04/21/2025]
Abstract
Obesity is a multi-chronic illness characterized by superfluous fat accumulation, contributing to significant metabolic and neurological complications. Current therapeutic approaches have limited efficacy and notable side effects, underscoring an urgent demand for novel, safer alternatives. This study is the first to investigate the anti-obesity potential of 5,7,3',4',5'-pentamethoxyflavone (PMF) in vivo using a zebrafish model. Our findings demonstrate that PMF administration exerts pronounced anti-obesogenic effects, evidenced by reductions in blood glucose, plasma triglycerides, total cholesterol, hepatic low-density lipoproteins (LDL), and high-density lipoproteins (HDL). Mechanistically, PMF suppressed hepatic adipogenic and lipogenic gene expression while promoting lipid catabolism through activation of peroxisome proliferator-activated receptor-alpha (PPAR-α) and its downstream enzymes, including acyl-CoA oxidase 1 (ACOX1), medium-chain acyl-CoA dehydrogenase (ACADM), and carnitine palmitoyl transferase 1B (CPT-1β). Additionally, PMF markedly mitigated oxidative stress by lowering malondialdehyde (MDA) and nitric oxide (NO) levels, accompanied by increased antioxidant enzyme activities, including superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GSH-Px), and glutathione S-transferase (GST). Notably, PMF effectively prevented obesity by suppressing food intake, downregulating orexigenic genes, and enhancing anorexigenic signals. Furthermore, PMF exhibited neuroprotective properties by elevating brain-derived neurotrophic factor (BDNF) and its receptor tropomyosin receptor kinase B2 (TrkB2), revealing a novel link between metabolic and neurological regulation. This study provides pioneering, comprehensive in vivo evidence supporting PMF as a promising therapeutic candidate with dual beneficial roles in metabolic health and neuroprotection.
Collapse
Affiliation(s)
- Muhammad Sufyan Vohra
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University Lakeside Campus, 47500, Subang Jaya, Selangor Darul Ehsan, Malaysia
| | - Bilal Ahmad
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor's University Lakeside Campus, 47500, Subang Jaya, Selangor Darul Ehsan, Malaysia
| | - Emerald R Taylor
- School of Chemistry and Forensic Science, Ingram Building, University of Kent, Kent, Canterbury, CT2 7NH, United Kingdom
| | - Khaled Benchoula
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University Lakeside Campus, 47500, Subang Jaya, Selangor Darul Ehsan, Malaysia
| | - Isabel Lim Fong
- Department of Paraclinical Sciences, Faculty of Medicine and Health Sciences, 94300, Kota Samarahan, Malaysia; Universiti Malaysia Sarawak, Malaysia
| | - Ishwar S Parhar
- School of Medicine and Health Sciences, Monash University, Sunway Campus, PJ 46150, Selangor, Malaysia
| | - Satoshi Ogawa
- School of Medicine and Health Sciences, Monash University, Sunway Campus, PJ 46150, Selangor, Malaysia
| | - Christopher J Serpell
- Department of Pharmaceutical and Biological Chemistry, School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1 1AX, United Kingdom.
| | - Eng Hwa Wong
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University Lakeside Campus, 47500, Subang Jaya, Selangor Darul Ehsan, Malaysia; Digital Health and Medical Advancement Impact Lab, Taylor's University Lakeside Campus, 1, Jalan Taylor's, Subang Jaya, Selangor, 47500, Malaysia.
| |
Collapse
|
2
|
Rezq S, Huffman AM, Basnet J, Alsemeh AE, do Carmo JM, Yanes Cardozo LL, Romero DG. MicroRNA-21 modulates brown adipose tissue adipogenesis and thermogenesis in a mouse model of polycystic ovary syndrome. Biol Sex Differ 2024; 15:53. [PMID: 38987854 PMCID: PMC11238487 DOI: 10.1186/s13293-024-00630-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 06/26/2024] [Indexed: 07/12/2024] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS), the most common endocrine disorder in premenopausal women, is associated with increased obesity, hyperandrogenism, and altered brown adipose tissue (BAT) thermogenesis. MicroRNAs play critical functions in brown adipocyte differentiation and maintenance. We aim to study the role of microRNA-21 (miR-21) in altered energy homeostasis and BAT thermogenesis in a PCOS mouse model of peripubertal androgen exposure. METHODS Three-week-old miR-21 knockout (miR21KO) or wild-type (WT) female mice were treated with dihydrotestosterone (DHT) or vehicle for 90 days. Body composition was determined by EchoMRI. Energy expenditure (EE), oxygen consumption (VO2), carbon dioxide production (VCO2), and respiratory exchange ratio (RER) were measured by indirect calorimetry. Androgen receptor (AR), and markers of adipogenesis, de novo lipogenesis, angiogenesis, extracellular matrix remodeling, and thermogenesis were quantified by RT-qPCR and/or Western-blot. RESULTS MiR-21 ablation attenuated DHT-mediated increase in body weight while having no effect on fat or BAT mass. MiR-21 ablation attenuated DHT-mediated BAT AR upregulation. MiR-21 ablation did not alter EE; however, miR21KO DHT-treated mice have reduced VO2, VCO2, and RER. MiR-21 ablation reversed DHT-mediated decrease in food intake and increase in sleep time. MiR-21 ablation decreased some adipogenesis (Adipoq, Pparγ, and Cebpβ) and extracellular matrix remodeling (Mmp-9 and Timp-1) markers expression in DHT-treated mice. MiR-21 ablation abolished DHT-mediated increases in thermogenesis markers Cpt1a and Cpt1b, while decreasing CIDE-A expression. CONCLUSIONS Our findings suggest that BAT miR-21 may play a role in regulating DHT-mediated thermogenic dysfunction in PCOS. Modulation of BAT miR-21 levels could be a novel therapeutic approach for the treatment of PCOS-associated metabolic derangements.
Collapse
Affiliation(s)
- Samar Rezq
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA.
- Women's Health Research Center, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA.
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA.
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA.
| | - Alexandra M Huffman
- Women's Health Research Center, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
| | - Jelina Basnet
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
- Women's Health Research Center, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
| | - Amira E Alsemeh
- Department of Anatomy, Histology, and Embryology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Jussara M do Carmo
- Department of Physiology and Biophysics, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
- Cardiorenal and Metabolic Diseases Research Center, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
| | - Licy L Yanes Cardozo
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
- Department of Medicine, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
- Women's Health Research Center, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA
| | - Damian G Romero
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA.
- Women's Health Research Center, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA.
- Cardiovascular-Renal Research Center, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA.
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS, 39216, USA.
| |
Collapse
|
3
|
Du J, Zhu Y, Yang X, Geng X, Xu Y, Zhang M, Zhang M. Berberine attenuates obesity-induced insulin resistance by inhibiting miR-27a secretion. Diabet Med 2024; 41:e15319. [PMID: 38711201 DOI: 10.1111/dme.15319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 03/06/2024] [Accepted: 03/11/2024] [Indexed: 05/08/2024]
Abstract
INTRODUCTION Berberine (BBR) is an alkaloid found in plants. It has neuroprotective, anti-inflammatory and lipid-lowering activity. However, the efficacy of treatment with BBR and the mechanisms through which it acts need further study. AIMS This study investigated the therapeutic effects and the mechanism of action of BBR on obesity-induced insulin resistance in peripheral tissues. METHODS High-fat-fed C57BL/6J mice and low-fat-fed C57BL/6J mice with miR-27a overexpression were given BBR intervention (100 mg/kg, po), and the oral glucose tolerance test (OGTT) and insulin tolerance test (ITT) were performed. Palmitic acid-stimulated hypertrophic adipocyte models were treated with BBR (10 μM). Related indicators and protein expression levels were examined. RESULTS The AUCs of the OGTT and the ITT in the BBR intervention group were reduced significantly (p < 0.01) (p < 0.05), and the serum biochemical parameters, including FBG, TC, TG and LDL-C were significantly reduced after BBR intervention. In the in vitro experiments, the triglyceride level and volume of lipid droplets decreased significantly after BBR intervention (p < 0.01) (p < 0.05). Likewise, BBR ameliorates skeletal muscle and pancreas insulin signalling pathways in vivo and in vitro. DISCUSSION The results showed that BBR significantly ameliorated insulin resistance, reduced body weight and percent body fat and improved serum biochemical parameters in mice. Likewise, BBR reduced triglyceride level and lipid droplet volume in hypertrophic adipocytes, BBR improved obesity effectively. Meanwhile, BBR ameliorated the histomorphology of the pancreas, and skeletal muscle and pancreas insulin related signalling pathways of islets in in vitro and in vivo experiments. The results further demonstrated that BBR inhibited miR-27a levels in serum from obese mice and supernatant of hypertrophic adipocytes. miR-27a overexpression in low-fat fed mice indicated that miR-27a caused insulin resistance, and BBR intervention significantly improved the miR-27a induced insulin resistance status. CONCLUSION This study demonstrates the important role of BBR in obesity-induced peripheral insulin resistance and suggest that the mechanism of its effect may be inhibition of miR-27a secretion.
Collapse
Affiliation(s)
- Junda Du
- Department of Pharmacology, College of Basic Medical Sciences, School of nursing, Jilin University, Changchun, Jilin, China
- School of Pharmaceutical Science, Jilin University, Changchun, Jilin, China
| | - Yu Zhu
- Department of Ophthalmology of Jilin Province FAW General Hospital, Changchun, Jilin, China
| | - Xuehan Yang
- Department of Pharmacology, College of Basic Medical Sciences, School of nursing, Jilin University, Changchun, Jilin, China
| | - Xinru Geng
- Department of Pharmacology, College of Basic Medical Sciences, School of nursing, Jilin University, Changchun, Jilin, China
| | - Yang Xu
- Department of Pharmacology, College of Basic Medical Sciences, School of nursing, Jilin University, Changchun, Jilin, China
| | - Meishuang Zhang
- Department of Pharmacology, College of Basic Medical Sciences, School of nursing, Jilin University, Changchun, Jilin, China
| | - Ming Zhang
- Department of Pharmacology, College of Basic Medical Sciences, School of nursing, Jilin University, Changchun, Jilin, China
| |
Collapse
|
4
|
Tu C, Wu Q, Wang J, Chen P, Deng Y, Yu L, Xu X, Fang X, Li W. miR-486-5p-rich extracellular vesicles derived from patients with olanzapine-induced insulin resistance negatively affect glucose-regulating function. Biochem Pharmacol 2024; 225:116308. [PMID: 38788961 DOI: 10.1016/j.bcp.2024.116308] [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: 01/20/2024] [Revised: 05/02/2024] [Accepted: 05/21/2024] [Indexed: 05/26/2024]
Abstract
A high risk of glucometabolic disorder severely disturbs compliance and limits the clinical application of olanzapine. MicroRNAs (miRNAs) in extracellular vesicles (EVs) have been reported as emerging biomarkers in glucolipid metabolic disorders. A total of 81 individuals with continuous olanzapine treatment over 3 months were recruited in this study, and plasma EVs from these individuals were isolated and injected into rats via the tail vein to investigate the glucose-regulating function in vivo. Moreover, we performed a miRNA profiling assay by high through-put sequencing to clarify the differentiated miRNA profiles between two groups of patients who were either susceptible or not susceptible to olanzapine-induced insulin resistance (IR). Finally, we administered antagomir and cocultured them with adipocytes to explore the mechanism in vitro. The results showed that individual insulin sensitivity varied in those patients and in olanzapine-administered rats. Furthermore, treatment with circulating EVs from patients with olanzapine-induced IR led to the development of metabolic abnormalities in rats and adipocytes in vitro through the AKT-GLUT4 pathway. Deep sequencing illustrated that the miRNAs of plasma EVs from patients showed a clear difference based on susceptibility to olanzapine-induced IR, and miR-486-5p was identified as a notable gene. The adipocyte data indicated that miR-486-5p silencing partially reversed the impaired cellular insulin sensitivity. Collectively, this study confirmed the function of plasma EVs in the interindividual differences in olanzapine-induced insulin sensitivity.
Collapse
Affiliation(s)
- Chuyue Tu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qian Wu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Wang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Peiru Chen
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yahui Deng
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lixiu Yu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaojin Xu
- Affiliated Wuhan Mental Health Center, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Xiangming Fang
- Department of Psychiatry, Wuhan Youfu Hospital, Wuhan, China
| | - Weiyong Li
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
5
|
Engin AB, Engin A. MicroRNAs as Epigenetic Regulators of Obesity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:595-627. [PMID: 39287866 DOI: 10.1007/978-3-031-63657-8_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
In obesity, the process of adipogenesis largely determines the number of adipocytes in body fat depots. Adipogenesis is regulated by several adipocyte-selective micro-ribonucleic acids (miRNAs) and transcription factors that modulate adipocyte proliferation and differentiation. However, some miRNAs block the expression of master regulators of adipogenesis. Since the specific miRNAs display different expressions during adipogenesis, in mature adipocytes and permanent obesity, their use as biomarkers or therapeutic targets is feasible. Upregulated miRNAs in persistent obesity are downregulated during adipogenesis. Moreover, some of the downregulated miRNAs in obese individuals are upregulated in mature adipocytes. Induction of adipocyte stress and hypertrophy leads to the release of adipocyte-derived exosomes (AdEXs) that contain the cargo molecules, miRNAs. miRNAs are important messengers for intercellular communication involved in metabolic responses and have very specific signatures that direct the metabolic activity of target cells. While each miRNA targets multiple messenger RNAs (mRNAs), which may coordinate or antagonize each other's functions, several miRNAs are dysregulated in other tissues during obesity-related comorbidities. Deletion of the miRNA-processing enzyme DICER in pro-opiomelanocortin-expressing cells results in obesity, which is characterized by hyperphagia, increased adiposity, hyperleptinemia, defective glucose metabolism, and alterations in the pituitary-adrenal axis. In recent years, RNA-based therapeutical approaches have entered clinical trials as novel therapies against overweight and its complications. Development of lipid droplets, macrophage accumulation, macrophage polarization, tumor necrosis factor receptor-associated factor 6 activity, lipolysis, lipotoxicity, and insulin resistance are effectively controlled by miRNAs. Thereby, miRNAs as epigenetic regulators are used to determine the new gene transcripts and therapeutic targets.
Collapse
Affiliation(s)
- Ayse Basak Engin
- Faculty of Pharmacy, Department of Toxicology, Gazi University, Hipodrom, Ankara, Turkey.
| | - Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey
| |
Collapse
|
6
|
Elkhawaga SY, Ismail A, Elsakka EGE, Doghish AS, Elkady MA, El-Mahdy HA. miRNAs as cornerstones in adipogenesis and obesity. Life Sci 2023; 315:121382. [PMID: 36639051 DOI: 10.1016/j.lfs.2023.121382] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/06/2023] [Accepted: 01/07/2023] [Indexed: 01/12/2023]
Abstract
In recent decades, obesity has extensively emerged to the level of pandemics. It's significantly associated with serious co-morbidities that could decrease life quality and even life expectancy. Obesity has several determinants, such as age, sex, endocrine, and genetic factors. The miRNAs have emerged as genetic factors affecting obesity. The miRNAs are small noncoding nucleic acids that can modify gene expression and hence, control biological processes. The miRNAs can greatly affect many biological processes in obesity, such as adipogenesis, lipid metabolism, and homeostasis. As a result, the entry of miRNAs in obesity therapeutic approaches has been strongly advised as miRNAs mimics, inhibitors, and stimulators. Hence, this review aims to point out a summarized and updated overview of miRNAs and their roles in obesity and its included processes, such as adipogenesis and lipid metabolism. Besides, we also review recent applications of miRNAs as a treatment approach for obesity.
Collapse
Affiliation(s)
- Samy Y Elkhawaga
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Ahmed Ismail
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Elsayed G E Elsakka
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt.
| | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt.
| | - Mohamed A Elkady
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Hesham A El-Mahdy
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt
| |
Collapse
|
7
|
Insights on Dietary Polyphenols as Agents against Metabolic Disorders: Obesity as a Target Disease. Antioxidants (Basel) 2023; 12:antiox12020416. [PMID: 36829976 PMCID: PMC9952395 DOI: 10.3390/antiox12020416] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 02/02/2023] [Accepted: 02/02/2023] [Indexed: 02/11/2023] Open
Abstract
Obesity is a condition that leads to increased health problems associated with metabolic disorders. Synthetic drugs are available for obesity treatment, but some of these compounds have demonstrated considerable side effects that limit their use. Polyphenols are vital phytonutrients of plant origin that can be incorporated as functional food ingredients. This review presents recent developments in dietary polyphenols as anti-obesity agents. Evidence supporting the potential application of food-derived polyphenols as agents against obesity has been summarized. Literature evidence supports the effectiveness of plant polyphenols against obesity. The anti-obesity mechanisms of polyphenols have been explained by their potential to inhibit obesity-related digestive enzymes, modulate neurohormones/peptides involved in food intake, and their ability to improve the growth of beneficial gut microbes while inhibiting the proliferation of pathogenic ones. Metabolism of polyphenols by gut microbes produces different metabolites with enhanced biological properties. Thus, research demonstrates that dietary polyphenols can offer a novel path to developing functional foods for treating obesity. Upcoming investigations need to explore novel techniques, such as nanocarriers, to improve the content of polyphenols in foods and their delivery and bioavailability at the target sites in the body.
Collapse
|
8
|
Ataei S, Ganjali S, Banach M, Karimi E, Sahebkar A. The effect of PCSK9 immunization on the hepatic level of microRNAs associated with the PCSK9/LDLR pathway. Arch Med Sci 2023; 19:203-208. [PMID: 36817686 PMCID: PMC9897094 DOI: 10.5114/aoms/152000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 07/09/2022] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION MicroRNAs (miRNAs) are a class of gene expression epigenetic regulators that play roles in regulating genes involved in cholesterol homeostasis, including low-density lipoprotein receptor (LDLR) and PCSK9; therefore, miRNAs have been suggested as potential therapeutic targets for treating cardiometabolic disorders. Thus, the present study aimed to assess the effect of immunotherapy with the PCSK9 peptide vaccine on the hepatic expression levels of microRNAs associated with the LDLR pathway, including miRNA-27a, miRNA-30c, and miRNA-191, in normal vaccinated mice. MATERIAL AND METHODS PCSK9 immunogenic peptide and 0.4% alum adjuvant were mixed at a 1 : 1 ratio and used as a vaccine formulation. Male albino mice were randomly assigned to the vaccine or control group. Mice in the vaccine group were injected four times at two-week intervals with a PCSK9 peptide vaccine, and mice in the control group were injected with phosphate-buffered saline (PBS). Animal livers were sampled 2 weeks after the last injection to assess miRNA expression levels. The hepatic expression levels of miRNA-27a, miRNA-30c, and miRNA-191 were evaluated by SYBR Green real-time PCR, quantified by a comparative (2- Δ Δ CT) method (fold change (FC)) and normalized to U6 small nuclear RNA (U6snRNA) expression as an internal control. RESULTS The hepatic expression level of miRNA-27a was significantly lower in mice following immunotherapy with the PCSK9 peptide vaccine compared to the control group (FC: 0.731 ±0.1, p = 0.027). Also, there was a borderline significantly lower hepatic expression level of miRNA-30c in the vaccinated group compared to the control (FC: 0.569 ±0.1, p = 0.078). However, no significant differences were found in the hepatic expression level of miRNA-191 between the two studied groups (FC: 0.852 ±0.1, p = 0.343). CONCLUSIONS According to the findings, the PCSK9 peptide vaccine could effectively reduce the hepatic expression level of miRNA-27a and may be helpful in the management of LDL-C level and atherosclerosis, which may be mediated through the LDLR pathway.
Collapse
Affiliation(s)
- Sarina Ataei
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Shiva Ganjali
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maciej Banach
- Department of Preventive Cardiology and Lipidology, Medical University of Lodz (MUL), Lodz, Poland
- Cardiovascular Research Centre, University of Zielona Gora, Zielona Gora, Poland
| | - Ehsan Karimi
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Medicine, The University of Western Australia, Perth, Australia
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
9
|
Aladel A, Khatoon F, Khan MI, Alsheweir A, Almutairi MG, Almutairi SO, Almutairi FK, Osmonaliev K, Beg MMA. Evaluation of miRNA-143 and miRNA-145 Expression and Their Association with Vitamin-D Status Among Obese and Non-Obese Type-2 Diabetic Patients. J Multidiscip Healthc 2022; 15:2979-2990. [PMID: 36597468 PMCID: PMC9805745 DOI: 10.2147/jmdh.s391996] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 12/13/2022] [Indexed: 12/29/2022] Open
Abstract
OBJECTIVE Growing epidemics of type-2 diabetes mellitus (T2DM) and obesity have become a serious health concern. Since miRNAs and vitamin levels affect the development and progression of numerous pathogenic diseases, including diabetes, the present study aimed to evaluate miRNA-143 and miRNA-145 expression and vitamin-D status among obese and non-obese T2DM patients. METHODS The study included 100 clinically confirmed newly diagnosed obese and non-obese T2DM cases and 100 healthy subjects. Total RNA was extracted from collected blood samples and 100 ng of RNA was used for cDNA synthesis, then TaqMan assay was performed to evaluate the miRNA-143 and miRNA-145 relative expression. RESULTS T2DM cases with hypertension (4.08 fold, p=0.01; 5.36 fold, p=0.009), fatigue (5.07 fold, p=0.04; 5.32 fold, p=0.03) and blurred vision (5.15 fold, p=0.01) showed higher miRNA-143 and miRNA-145 relative expression compared with their counterparts, respectively. A positive correlation was observed between miRNA-143 and miRNA-145 expression and decreased vitamin-D status in T2DM had significant association with impaired blood glucose fasting (p=0.001) and HDL level (p<0.0001). Obese T2DM cases showed higher miRNA-143 and miRNA-145 expression compared with their counterparts. Vitamin-D deficient T2DM cases had higher miRNA-143 and miRNA-145 expression (5.69 fold, 5.91 fold) compared with insufficient (4.27 fold, p=0.03; 4.61 fold, p=0.03) and sufficient (4.08 fold, p=0.002; 4.29 fold, p=0.003). ROC curve for miRNA-143 and miRNA-145 between obese T2DM vs non-obese T2DM cases, at best possible cutoff value of 4.39 fold, 4.0 fold showed increased miRNA-143 and miRNA-145 expression, the sensitivity and specificity were 85%, 88% and 61%, 53% respectively (AUC=0.83, p<0.0001; AUC=0.81, p<0.0001). CONCLUSION Higher miRNA-143 and miRNA-145 expression could be a predictive indicator for obese T2DM cases, decreased status of vitamin-D was also significantly associated with impaired fasting blood sugar and HDL level, therefore it is important to evaluate the vitamin-D status among T2DM cases for better clinical outcome during the intervention.
Collapse
Affiliation(s)
- Alanoud Aladel
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Fahmida Khatoon
- Biochemistry Department, College of Medicine, University of Ha’il, Ha’il, Saudi Arabia
| | - Mohammad Idreesh Khan
- Department of Clinical Nutrition, College of Applied Health Sciences in Ar Rass, Qassim University, Ar Rass, 51921, Saudi Arabia
| | - Azzah Alsheweir
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Malak Ghazi Almutairi
- Department of Clinical Nutrition, Almethnab General Hospital, Qassim Health Cluster, Ministry of Health, Al Mithnab, Saudi Arabia
| | - Sami Owaidh Almutairi
- Department of Clinical Nutrition, Almethnab General Hospital, Qassim Health Cluster, Ministry of Health, Al Mithnab, Saudi Arabia
| | - Faisal Khalid Almutairi
- Laboratory Department, Armed Forces Hospital in Qassim, Medical Services, Ministry of Defense Qassim Buraydah Al-Rass, Buraydah, Saudi Arabia
| | | | - Mirza Masroor Ali Beg
- Faculty of Medicine, Alatoo International University, Bishkek, Kyrgyzstan
- Centre for Promotion of Medical Research, Bishkek, Kyrgyzstan
| |
Collapse
|
10
|
Saghazadeh A, Rezaei N. MicroRNA expression profiles of peripheral blood and mononuclear cells in myasthenia gravis: A systematic review. Int Immunopharmacol 2022; 112:109205. [PMID: 36087508 DOI: 10.1016/j.intimp.2022.109205] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 07/21/2022] [Accepted: 08/26/2022] [Indexed: 11/05/2022]
Abstract
BACKGROUND Studies have described the role of microRNAs (miRNAs) in thymic function, along with directly observing the altered expression of miRNAs in thymuses of myasthenia gravis (MG) patients; so, miRNAs became a core component in the pathophysiology of MG. However, because the miRNA analysis results are contradictory, the identification of MG-related miRNAs is daunting. OBJECTIVE We did a systematic review of studies analyzing the miRNA expression profile of peripheral blood and mononuclear cells for patients with MG. METHODS We ran a database search in PubMed, Scopus, and Web of Science on August 17, 2021. Original articles that analyzed miRNA profiles in peripheral blood (serum, plasma, and whole blood) and peripheral blood mononuclear cells (PBMCs) for patients with MG in comparison with a non-MG or healthy control (HC) group were eligible. The quality of studies was assessed using the Quality Assessment of Diagnostic Accuracy Studies 2 (QUADAS-2). RESULTS 26 studies were included. The quality of studies was fair (median score, 5). Among 226 different miRNAs that were deregulated in at least one study (range, 1-87), ten miRNAs were significantly deregulated in three or more studies. Five miRNAs (50%) showed the same deregulation: miR-106b-3p and miR-21-5p were consistently upregulated, and miR-20b, miR-15b, and miR-16 were consistently downregulated. Also, there were five miRNAs that were mostly upregulated, miR-150-5p, miR-146a, miR-30e-5p, and miR-338-3p, or downregulated, miR-324-3p, across studies. CONCLUSION These miRNAs contribute to different pathways, importantly neural apoptosis and autophagy, inflammation, T regulatory cell development, and T helper cell balance. Prior to being used for diagnostic and therapeutic purposes, it is required to pursue molecular mechanisms these consistently and mostly dysregulated miRNAs specifically use in the context of MG.
Collapse
Affiliation(s)
- Amene Saghazadeh
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Systematic Review and Meta-analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Network of Immunity in Infection, Malignancy, and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| |
Collapse
|
11
|
Li X, Zheng L, Zhang B, Deng ZY, Luo T. The Structure Basis of Phytochemicals as Metabolic Signals for Combating Obesity. Front Nutr 2022; 9:913883. [PMID: 35769384 PMCID: PMC9234462 DOI: 10.3389/fnut.2022.913883] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 05/23/2022] [Indexed: 12/12/2022] Open
Abstract
The consumption of phytochemicals, bioactive compounds in fruits and vegetables, has been demonstrated to ameliorate obesity and related metabolic symptoms by regulating specific metabolic pathways. This review summarizes the progress made in our understanding of the potential of phytochemicals as metabolic signals: we discuss herein selected molecular mechanisms which are involved in the occurrence of obesity that may be regulated by phytochemicals. The focus of our review highlights the regulation of transcription factors toll like receptor 4 (TLR4), nuclear factor (erythroid-derived 2)-like 2 (Nrf2), the peroxisome proliferator-activated receptors (PPARs), fat mass and obesity-associated protein (FTO) and regulation of microRNAs (miRNA). In this review, the effect of phytochemicals on signaling pathways involved in obesity were discussed on the basis of their chemical structure, suggesting molecular mechanisms for how phytochemicals may impact these signaling pathways. For example, compounds with an isothiocyanate group or an α, β-unsaturated carbonyl group may interact with the TLR4 signaling pathway. Regarding Nrf2, we examine compounds possessing an α, β-unsaturated carbonyl group which binds covalently with the cysteine thiols of Keap1. Additionally, phytochemical activation of PPARs, FTO and miRNAs were summarized. This information may be of value to better understand how specific phytochemicals interact with specific signaling pathways and help guide the development of new drugs to combat obesity and related metabolic diseases.
Collapse
|
12
|
Pérez-García A, Torrecilla-Parra M, Fernández-de Frutos M, Martín-Martín Y, Pardo-Marqués V, Ramírez CM. Posttranscriptional Regulation of Insulin Resistance: Implications for Metabolic Diseases. Biomolecules 2022; 12:biom12020208. [PMID: 35204710 PMCID: PMC8961590 DOI: 10.3390/biom12020208] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 01/14/2022] [Accepted: 01/17/2022] [Indexed: 12/14/2022] Open
Abstract
Insulin resistance defines an impairment in the biologic response to insulin action in target tissues, primarily the liver, muscle, adipose tissue, and brain. Insulin resistance affects physiology in many ways, causing hyperglycemia, hypertension, dyslipidemia, visceral adiposity, hyperinsulinemia, elevated inflammatory markers, and endothelial dysfunction, and its persistence leads to the development metabolic disease, including diabetes, obesity, cardiovascular disease, or nonalcoholic fatty liver disease (NAFLD), as well as neurological disorders such as Alzheimer’s disease. In addition to classical transcriptional factors, posttranscriptional control of gene expression exerted by microRNAs and RNA-binding proteins constitutes a new level of regulation with important implications in metabolic homeostasis. In this review, we describe miRNAs and RBPs that control key genes involved in the insulin signaling pathway and related regulatory networks, and their impact on human metabolic diseases at the molecular level, as well as their potential use for diagnosis and future therapeutics.
Collapse
|
13
|
Thibonnier M, Ghosh S, Blanchard A. Effects of a short-term cold exposure on circulating microRNAs and metabolic parameters in healthy adult subjects. J Cell Mol Med 2021; 26:548-562. [PMID: 34921497 PMCID: PMC8743656 DOI: 10.1111/jcmm.17121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 11/19/2021] [Accepted: 11/29/2021] [Indexed: 11/28/2022] Open
Abstract
This discovery study investigated in healthy subjects whether a short‐term cold exposure may alter circulating microRNAs and metabolic parameters and if co‐expression networks between these factors could be identified. This open randomized crossover (cold vs no cold exposure) study with blind end‐ point evaluation was conducted at 1 center with 10 healthy adult male volunteers. Wearing a cooling vest perfused at 14°C for 2 h reduced the local skin temperature without triggering shivering, increased norepinephrine and blood pressure while decreasing copeptin, C‐peptide and heart rate. Circulating microRNAs measured before and after wearing the cooling vest twice (4 time points) identified 196 mature microRNAs with excellent reproducibility over 72 h. Significant correlations of microRNA expression with copeptin, norepinephrine and C‐peptide were found. A co‐expression‐based microRNA‐microRNA network, as well as microRNA pairs displaying differential correlation as a function of temperature were also detected. This study demonstrates that circulating miRNAs are differentially expressed and coregulated upon cold exposure in humans, supporting their use as predictive and dynamic biomarkers of cardio‐metabolic disorders.
Collapse
Affiliation(s)
| | - Sujoy Ghosh
- Duke-NUS Medical School, Singapore City, Singapore.,Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - Anne Blanchard
- Clinical Investigation Center, Hôpital Européen Georges Pompidou, Paris, France
| |
Collapse
|
14
|
miR-21 mimic blocks obesity in mice: A novel therapeutic option. MOLECULAR THERAPY-NUCLEIC ACIDS 2021; 26:401-416. [PMID: 34552821 PMCID: PMC8426473 DOI: 10.1016/j.omtn.2021.06.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 06/25/2021] [Indexed: 12/14/2022]
Abstract
MicroRNAs (miRNAs) are promising drug targets for obesity and metabolic disorders. Recently, miRNA mimics are providing a unique mechanism of action that guides the process for drug development and sets out the context of their therapeutic application. miRNA (miR)-21 expression in white adipose tissue (WAT) has been associated with obesity. We aimed to analyze miR-21 expression levels in relation to diabetes and obesity to determine the effect that miR-21 mimic has on processes involved in WAT functionality, to dissect the underlying molecular mechanisms, and to study the potential therapeutic application of the miR-21 mimic against obesity. We found higher miR-21 levels in WAT from non-diabetic obese compared to normoweight humans and mice. Moreover, in 3T3-L1 adipocytes, miR-21 mimic affect genes involved in WAT functionality regulation and significantly increase the expression of genes involved in browning and thermogenesis. Interestingly, in vivo treatment with the miR-21 mimic blocked weight gain induced by a high-fat diet in obese mice, without modifying food intake or physical activity. This was associated with metabolic enhancement, WAT browning, and brown adipose tissue (AT) thermogenic programming through vascular endothelial growth factor A (VEGF-A), p53, and transforming growth factor β1 (TGF-β1) signaling pathways. Our findings suggest that miR-21 mimic-based therapy may provide a new opportunity to therapeutically manage obesity and consequently, its associated alterations.
Collapse
|
15
|
McLaughlin T, Schnittger I, Nagy A, Zanley E, Xu Y, Song Y, Nieman K, Tremmel JA, Dey D, Boyd J, Sacks H. Relationship Between Coronary Atheroma, Epicardial Adipose Tissue Inflammation, and Adipocyte Differentiation Across the Human Myocardial Bridge. J Am Heart Assoc 2021; 10:e021003. [PMID: 34726081 PMCID: PMC8751937 DOI: 10.1161/jaha.121.021003] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Background Inflammation in epicardial adipose tissue (EAT) may contribute to coronary atherosclerosis. Myocardial bridge is a congenital anomaly in which the left anterior descending coronary artery takes a "tunneled" course under a bridge of myocardium: while atherosclerosis develops in the proximal left anterior descending coronary artery, the bridged portion is spared, highlighting the possibility that geographic separation from inflamed EAT is protective. We tested the hypothesis that inflammation in EAT was related to atherosclerosis by comparing EAT from proximal and bridge depots in individuals with myocardial bridge and varying degrees of atherosclerotic plaque. Methods and Results Maximal plaque burden was quantified by intravascular ultrasound, and inflammation was quantified by pericoronary EAT signal attenuation (pericoronary adipose tissue attenuation) from cardiac computed tomography scans. EAT overlying the proximal left anterior descending coronary artery and myocardial bridge was harvested for measurement of mRNA and microRNA (miRNA) using custom chips by Nanostring; inflammatory cytokines were measured in tissue culture supernatants. Pericoronary adipose tissue attenuation was increased, indicating inflammation, in proximal versus bridge EAT, in proportion to atherosclerotic plaque. Individuals with moderate-high versus low plaque burden exhibited greater expression of inflammation and hypoxia genes, and lower expression of adipogenesis genes. Comparison of gene expression in proximal versus bridge depots revealed differences only in participants with moderate-high plaque: inflammation was higher in proximal and adipogenesis lower in bridge EAT. Secreted inflammatory cytokines tended to be higher in proximal EAT. Hypoxia-inducible factor 1a was highly associated with inflammatory gene expression. Seven miRNAs were differentially expressed by depot: 3192-5P, 518D-3P, and 532-5P were upregulated in proximal EAT, whereas miR 630, 575, 16-5P, and 320E were upregulated in bridge EAT. miR 630 correlated directly with plaque burden and inversely with adipogenesis genes. miR 3192-5P, 518D-3P, and 532-5P correlated inversely with hypoxia/oxidative stress, peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PCG1a), adipogenesis, and angiogenesis genes. Conclusions Inflammation is specifically elevated in EAT overlying atherosclerotic plaque, suggesting that EAT inflammation is caused by atherogenic molecular signals, including hypoxia-inducible factor 1a and/or miRNAs in an "inside-to-out" relationship. Adipogenesis was suppressed in the bridge EAT, but only in the presence of atherosclerotic plaque, supporting cross talk between the vasculature and EAT. miR 630 in EAT, expressed differentially according to burden of atherosclerotic plaque, and 3 other miRNAs appear to inhibit key genes related to adipogenesis, angiogenesis, hypoxia/oxidative stress, and thermogenesis in EAT, highlighting a role for miRNA in mediating cross talk between the coronary vasculature and EAT.
Collapse
Affiliation(s)
- Tracey McLaughlin
- Division of Endocrinology Stanford University School of Medicine Stanford CA
| | - Ingela Schnittger
- Division of Cardiovascular Medicine Stanford University School of Medicine Stanford CA
| | - Anna Nagy
- Division of Endocrinology Stanford University School of Medicine Stanford CA
| | - Elizabeth Zanley
- Division of Endocrinology Stanford University School of Medicine Stanford CA
| | - Yue Xu
- Division of Endocrinology Stanford University School of Medicine Stanford CA
| | - Yanqiu Song
- Cardiovascular Institute Tianjin Chest Hospital Tianjin China
| | - Koen Nieman
- Division of Cardiovascular Medicine Stanford University School of Medicine Stanford CA
| | - Jennifer A Tremmel
- Division of Cardiovascular Medicine Stanford University School of Medicine Stanford CA
| | - Damini Dey
- Department of Biomedical Sciences and Medicine Cedars-Sinai Medical Center Biomedical Imaging Research Institute Los Angeles CA
| | - Jack Boyd
- Department of Cardiothoracic Surgery Stanford University School of Medicine Stanford CA
| | - Harold Sacks
- Division of Endocrinology Department of Medicine David Geffen School of Medicine at UCLA Los Angeles CA
| |
Collapse
|
16
|
Bta-miR-2400 Targets SUMO1 to Affect Yak Preadipocytes Proliferation and Differentiation. BIOLOGY 2021; 10:biology10100949. [PMID: 34681048 PMCID: PMC8533534 DOI: 10.3390/biology10100949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/15/2021] [Accepted: 09/16/2021] [Indexed: 11/17/2022]
Abstract
Yak adipose tissue may have evolved a unique energy metabolism manner to accommodate the organism's seasonal growth rhythms. MiRNAs regulate multiple biological processes including systemic metabolism and energy homeostasis through post-transcriptional regulations. Rare reports have shown that miRNAs regulate lipid metabolism in domestic yaks. Therefore, we investigated the regulatory mechanisms of bta-miR-2400 in modulating yak preadipocytes proliferation and differentiation. We found that bta-miR-2400 was highly expressed in adipose tissue. Overexpression of bta-miR-2400 in yak preadipocytes significantly enhanced cell proliferation, increased the number of EdU fluorescence-stained cells, and promoted the expression of proliferation marker genes (CDK2, CDK4 and PCNA). Besides, overexpression of bta-miR-2400 repressed the expression of adipogenesis-related marker genes, and the content of cellular triglyceride was substantially reduced. Conversely, inhibition of bta-miR-2400 showed opposite effects compared to those of bta-miR-2400 overexpression in yak preadipocytes. Further, luciferase reporter assays revealed that SUMO1 is a target gene of bta-miR-2400, with bta-miR-2400 being able to down-regulate SUMO1 mRNA and protein expression. In conclusion, bta-miR-2400 regulates lipid metabolism and energy homeostasis in yak preadipocytes by directly targeting SUMO1 to promote cell proliferation and inhibit differentiation.
Collapse
|
17
|
Heyn GS, Corrêa LH, Magalhães KG. The Impact of Adipose Tissue-Derived miRNAs in Metabolic Syndrome, Obesity, and Cancer. Front Endocrinol (Lausanne) 2020; 11:563816. [PMID: 33123088 PMCID: PMC7573351 DOI: 10.3389/fendo.2020.563816] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 09/18/2020] [Indexed: 12/11/2022] Open
Abstract
Obesity is a multifactorial and complex condition that is characterized by abnormal and excessive white adipose tissue accumulation, which can lead to the development of metabolic diseases, such as type 2 diabetes mellitus, nonalcoholic fatty liver disease, cardiovascular diseases, and several types of cancer. Obesity is characterized by excessive adipose tissue accumulation and associated with alterations in immunity, displaying a chronic low-grade inflammation profile. Adipose tissue is a dynamic and complex endocrine organ composed not only by adipocytes, but several immunological cells, which can secrete hormones, cytokines and many other factors capable of regulating metabolic homeostasis and several critical biological pathways. Remarkably, adipose tissue is a major source of circulating microRNAs (miRNAs), recently described as a novel form of adipokines. Several adipose tissue-derived miRNAs are deeply associated with adipocytes differentiation and have been identified with an essential role in obesity-associated inflammation, insulin resistance, and tumor microenvironment. During obesity, adipose tissue can completely change the profile of the secreted miRNAs, influencing circulating miRNAs and impacting the development of different pathological conditions, such as obesity, metabolic syndrome, and cancer. In this review, we discuss how miRNAs can act as epigenetic regulators affecting adipogenesis, adipocyte differentiation, lipid metabolism, browning of the white adipose tissue, glucose homeostasis, and insulin resistance, impacting deeply obesity and metabolic diseases. Moreover, we characterize how miRNAs can often act as oncogenic and tumor suppressor molecules, significantly modulating cancer establishment and progression. Furthermore, we highlight in this manuscript how adipose tissue-derived miRNAs can function as important new therapeutic targets.
Collapse
Affiliation(s)
| | | | - Kelly Grace Magalhães
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasilia, Brasilia, Brazil
| |
Collapse
|
18
|
Thibonnier M, Esau C, Ghosh S, Wargent E, Stocker C. Metabolic and energetic benefits of microRNA-22 inhibition. BMJ Open Diabetes Res Care 2020; 8:8/1/e001478. [PMID: 33004402 PMCID: PMC7534675 DOI: 10.1136/bmjdrc-2020-001478] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 07/02/2020] [Accepted: 08/13/2020] [Indexed: 12/14/2022] Open
Abstract
INTRODUCTION We previously demonstrated in primary cultures of human subcutaneous adipocytes and in a mouse model of diet-induced obesity that specific microRNA-22-3p antagomirs produce a significant reduction of fat mass and an improvement of several metabolic parameters. These effects are related to the activation of target genes such as KDM3A, KDM6B, PPARA, PPARGC1B and SIRT1 involved in lipid catabolism, thermogenesis, insulin sensitivity and glucose homeostasis. RESEARCH DESIGN AND METHODS We now report a dedicated study exploring over the course of 3 months the metabolic and energetic effects of subcutaneous administration of our first miR-22-3p antagomir drug candidate (APT-110) in adult C57BL/6 male mice. Body composition, various blood parameters and energy expenditure were measured at several timepoints between week 12 and week 27 of age. RESULTS Weekly subcutaneous injections of APT-110 for 12 weeks produced a sustained increase of energy expenditure as early as day 11 of treatment, a significant fat mass reduction, but no change of appetite nor physical activity. Insulin sensitivity as well as circulating glucose, cholesterol and leptin were improved. There was a dramatic reduction of liver steatosis after 3 months of active treatment. RNA sequencing revealed an activation of lipid metabolism pathways in a tissue-specific manner. CONCLUSIONS These original findings suggest that microRNA-22-3p inhibition could lead to a potent treatment of fat accumulation, insulin resistance, and related complex metabolic disorders such as obesity, type 2 diabetes mellitus and non-alcoholic fatty liver disease.
Collapse
Affiliation(s)
| | | | - Sujoy Ghosh
- Centre for Computational Biology and Program in Cardiovascular & Metabolic Disorders, Duke-NUS Medical School, Singapore
| | - Edward Wargent
- Clore Laboratory, University of Buckingham, Buckingham, UK
| | - Claire Stocker
- Clore Laboratory, University of Buckingham, Buckingham, UK
| |
Collapse
|
19
|
Ashrafizadeh M, Zarrabi A, Hashemipour M, Vosough M, Najafi M, Shahinozzaman M, Hushmandi K, Khan H, Mirzaei H. Sensing the scent of death: Modulation of microRNAs by Curcumin in gastrointestinal cancers. Pharmacol Res 2020; 160:105199. [DOI: 10.1016/j.phrs.2020.105199] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/06/2020] [Accepted: 09/07/2020] [Indexed: 02/06/2023]
|
20
|
Youssef EM, Elfiky AM, BanglySoliman, Abu-Shahba N, Elhefnawi MM. Expression profiling and analysis of some miRNAs in subcutaneous white adipose tissue during development of obesity. GENES AND NUTRITION 2020; 15:8. [PMID: 32366215 PMCID: PMC7197174 DOI: 10.1186/s12263-020-00666-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 03/18/2020] [Indexed: 12/24/2022]
Abstract
Background MicroRNAs are emerging as new mediators in the regulation of adipocyte physiology and have been approved to play a role in obesity. Despite several studies have focused on microRNA expression profiles and functions in different metabolic tissues, little is known about their response to nutritional interventions in white adipose tissue during obesity stages, and whether they differ in this response to weight-reduction strategy is poorly understood. Our objectives were to study the dysregulation of some miRNAs in subcutaneous inguinal white adipose tissue during weight change, expansion/reduction; in response to both a high-fat diet and switching to a normal diet feeding, and to evaluate them as potential biomarkers and therapeutic targets for early obesity management Method A hundred 6-week-old male Wister rats were randomly divided into a normal diet group (N.D), a high-fat diet group (H.F.D), and a switched to a normal diet group (H.F.D/N.D). At the beginning and at intervals 2 weeks, serum lipid, hormone levels, total body fat mass, and inguinal subcutaneous white adipose tissue mass (WAT) measurements were recorded using dual-energy X-ray absorptiometry (DEXA). The expression levels of microRNAs were evaluated using real-time PCR. Results Significant alterations were observed in serum glucose, lipid profile, and adipokine hormones during the early stages of obesity development. Alteration in rno-mir 30a-5p, rno-mir 133a-5p, and rno-mir 107-5p expression levels were observed at more than one time point. While rno-let-7a-5p, rno-mir 193a-5p, and rno-mir125a-5p were downregulated and rno-mir130a-5p was upregulated at all time points within 2 to 4 weeks in response to H.F.D feeding for 10 weeks. The impact of switching to normal diet has a reversed effect on lipid profile, adipokine hormone levels, and some miRNAs. The bioinformatics results have identified a novel and important pathway related to inflammatory signalling. Conclusion Our research demonstrated significant alterations in some adipocyte-expressed miRNAs after a short time of high caloric diet consumption. This provides further evidence of the significant role of nutrition as an epigenetic factor in regulation of lipid and glucose metabolism genes by modulating of related key miRNAs. Therefore, we suggest that miRNAs could be used as biomarkers for adiposity during diet-induced obesity. Perhaps limitation in calories intake is a way to manipulate obesity and associated metabolic disorders. Further studies are needed to fully elucidate the role of microRNAs in the development of obesity
Collapse
Affiliation(s)
- Elham M Youssef
- Biochemistry Department, National Research Centre, Cairo, Egypt
| | - Asmaa M Elfiky
- Environmental and Occupational Medicine Department, Environmental Research Division, National Research Centre, Cairo, Egypt
| | - BanglySoliman
- Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Nourhan Abu-Shahba
- Stem Cell Research Group, Centre of Excellence for Advanced Sciences, Department of Medical Molecular Genetics, National Research Centre, Cairo, Egypt
| | - Mahmoud M Elhefnawi
- Informatics and Systems Department, Engineering Research Division, National Research Centre, Cairo, Egypt. .,Biomedical Informatics and Chemoinformatics Group, Center of Excellence for Advanced Sciences, Informatics and Systems Department, National Research Centre, Cairo, Egypt.
| |
Collapse
|
21
|
Lin T, Guo H, Chen X. Pentraxin 3 Regulates miR-21 Expression and Secretion in Brown Adipocytes During Lipopolysaccharide-Induced Inflammation. Obesity (Silver Spring) 2020; 28:323-332. [PMID: 31898407 DOI: 10.1002/oby.22701] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 10/08/2019] [Indexed: 12/12/2022]
Abstract
OBJECTIVE This study investigates the role of pentraxin 3 (PTX3) in the regulation of inflammatory homeostasis involving anti-inflammatory miR-21 during lipopolysaccharide (LPS) stimulation in adipocytes. METHODS Using PTX3 knockout (PTX3 KO) mouse and primary stromal vascular cell models, this study determined the effect of PTX3 deficiency on the expression and secretion of miR-21 in brown adipose tissue (BAT) and brown adipocytes as well as the rescue effect of recombinant PTX3 on miR-21 during LPS-induced inflammation. RESULTS Among three fat depots, BAT was the major tissue and fully differentiated brown adipocytes were the major cells that expressed miRNA-processing enzymes and produced miRNA. Moreover, brown adipocytes, but not stromal vascular cells, were the LPS-responsive cells in miR-21 production. PTX3 deficiency attenuated LPS-stimulated upregulation of miR-21 in sera and BAT. In wild-type brown adipocytes, LPS stimulation significantly upregulated cellular miR-21. Interestingly, this stimulatory effect of LPS was attenuated, and cellular and secreted miR-21 levels were reduced in PTX3 KO cells upon LPS stimulation. Treatment of recombinant PTX3 reversed the cellular and secreted levels of miR-21 and attenuated an LPS-stimulated increase in the expression of Tnf-α and Mcp1 genes in PTX3 KO adipocytes. CONCLUSIONS PTX3 plays an anti-inflammatory role, in part through regulating miR-21 expression and secretion.
Collapse
Affiliation(s)
- Teyueh Lin
- Department of Food Science and Nutrition, University of Minnesota, Twin Cities, Saint Paul, Minnesota, USA
| | - Hong Guo
- Department of Food Science and Nutrition, University of Minnesota, Twin Cities, Saint Paul, Minnesota, USA
| | - Xiaoli Chen
- Department of Food Science and Nutrition, University of Minnesota, Twin Cities, Saint Paul, Minnesota, USA
| |
Collapse
|
22
|
Thibonnier M, Esau C. Metabolic Benefits of MicroRNA-22 Inhibition. Nucleic Acid Ther 2019; 30:104-116. [PMID: 31873061 DOI: 10.1089/nat.2019.0820] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Diabesity is a growing pandemic with substantial health and financial consequences. We are developing microRNA (miRNA)-based drug candidates that transform fat storing adipocytes into fat burning adipocytes (browning effect) to treat metabolic diseases characterized by lipotoxicity. Through phenotypic screening in primary cultures of human subcutaneous adipocytes, we discovered that inhibition of miRNA-22-3p by several complementary antagomirs resulted in increased lipid oxidation, mitochondrial activity, and energy expenditure (EE). These effects may be mediated through activation of target genes like KDM3A, KDM6B, PPARA, PPARGC1B, and SIRT1 involved in lipid catabolism, thermogenesis, and glucose homeostasis. In the model of Diet-Induced Obesity in mice of various ages, weekly subcutaneous injections of various miRNA-22-3p antagomirs produced a significant fat mass reduction, but no change of appetite or body temperature. Insulin sensitivity, as well as circulating glucose and cholesterol levels, was also improved. These original findings suggest that miRNA-22-3p inhibition could become a potent treatment of human obesity and type 2 diabetes mellitus, the so-called diabesity characterized by lipotoxicity and insulin resistance.
Collapse
|
23
|
Lv L, Zhang J, Tian F, Li X, Li D, Yu X. Arbutin protects HK-2 cells against high glucose-induced apoptosis and autophagy by up-regulating microRNA-27a. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:2940-2947. [PMID: 31319730 DOI: 10.1080/21691401.2019.1640231] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- Lina Lv
- Department of Nephrology, Jining No.1 People's Hospital, Jining, China
- Affiliated Jining No.1 People's Hospital of Jining Medical University, Jining Medical University, Jining, China
| | - Jing Zhang
- Department of Endocrinology, Jining No.1 People's Hospital, Jining, China
| | - Fengqun Tian
- Department of Nephrology, Jiaxiang County Medicine Hospital, Jiaxiang County, Jining, China
| | - Xia Li
- Department of Nephrology, Jining No.1 People's Hospital, Jining, China
| | - Dandan Li
- Department of Endocrinology, Jining No.1 People's Hospital, Jining, China
| | - Xiulian Yu
- Department of Nephrology, Jining No.1 People's Hospital, Jining, China
| |
Collapse
|
24
|
MicroRNAs and other non-coding RNAs in adipose tissue and obesity: emerging roles as biomarkers and therapeutic targets. Clin Sci (Lond) 2019; 133:23-40. [PMID: 30606812 DOI: 10.1042/cs20180890] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 11/29/2018] [Accepted: 12/05/2018] [Indexed: 02/07/2023]
Abstract
Obesity is a metabolic condition usually accompanied by insulin resistance (IR), type 2 diabetes (T2D), and dyslipidaemia, which is characterised by excessive fat accumulation and related to white adipose tissue (WAT) dysfunction. Enlargement of WAT is associated with a transcriptional alteration of coding and non-coding RNAs (ncRNAs). For many years, big efforts have focused on understanding protein-coding RNAs and their involvement in the regulation of adipocyte physiology and subsequent role in obesity. However, diverse findings have suggested that a dysfunctional adipocyte phenotype in obesity might be also dependent on specific alterations in the expression pattern of ncRNAs, such as miRNAs. The aim of this review is to update current knowledge on the physiological roles of miRNAs and other ncRNAs in adipose tissue function and their potential impact on obesity. Therefore, we examined their regulatory role on specific WAT features: adipogenesis, adipokine secretion, inflammation, glucose metabolism, lipolysis, lipogenesis, hypoxia and WAT browning. MiRNAs can be released to body fluids and can be transported (free or inside microvesicles) to other organs, where they might trigger metabolic effects in distant tissues, thus opening new possibilities to a potential use of miRNAs as biomarkers for diagnosis, prognosis, and personalisation of obesity treatment. Understanding the role of miRNAs also opens the possibility of using these molecules on individualised dietary strategies for precision weight management. MiRNAs should be envisaged as a future therapeutic approach given that miRNA levels could be modulated by synthetic molecules (f.i. miRNA mimics and inhibitors) and/or specific nutrients or bioactive compounds.
Collapse
|
25
|
Belarbi Y, Mejhert N, Gao H, Arner P, Rydén M, Kulyté A. MicroRNAs-361-5p and miR-574-5p associate with human adipose morphology and regulate EBF1 expression in white adipose tissue. Mol Cell Endocrinol 2018; 472:50-56. [PMID: 29191698 DOI: 10.1016/j.mce.2017.11.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 09/24/2017] [Accepted: 11/23/2017] [Indexed: 12/21/2022]
Abstract
Reduced adipose expression of the transcription factor Early B cell factor 1 (EBF1) is linked to white adipose tissue (WAT) hypertrophy. We aimed to identify microRNAs (miRNAs) associated with WAT hypertrophy and EBF1 regulation. We mapped WAT miRNA expression from 26 non-obese women discordant in WAT morphology and determined EBF1 activity in the non-obese and 30 obese women. Expression of 15 miRNAs was higher in hypertrophy and 10 were predicted to target EBF1. Binding of miR-365-5p/miR-574-5p were validated with 3'-UTR assay. Overexpression of miR-365-5p or miR-574-5p reduced EBF1 while inhibition of miR-574 increased EBF1 expression in human adipocytes in vitro. Additive effects on EBF1 were observed when concomitantly overexpressing both miRNAs. EBF1 targets were affected by over expression/inhibition of either miRNAs. Finally, miR-365-5p/miR-574-5p expression in 56 individuals correlated significantly with EBF1 activity. Our results suggest that miR-365-5p and miR-574-5p may be linked to WAT hypertrophy via effects on EBF1 expression.
Collapse
Affiliation(s)
- Yasmina Belarbi
- Lipid Laboratory, Department of Medicine Huddinge, Karolinska Institutet, SE-14186 Stockholm, Sweden
| | - Niklas Mejhert
- Lipid Laboratory, Department of Medicine Huddinge, Karolinska Institutet, SE-14186 Stockholm, Sweden
| | - Hui Gao
- Lipid Laboratory, Department of Medicine Huddinge, Karolinska Institutet, SE-14186 Stockholm, Sweden; Department of Biosciences & Nutrition, Karolinska Institutet, SE-141 Stockholm, Sweden
| | - Peter Arner
- Lipid Laboratory, Department of Medicine Huddinge, Karolinska Institutet, SE-14186 Stockholm, Sweden
| | - Mikael Rydén
- Lipid Laboratory, Department of Medicine Huddinge, Karolinska Institutet, SE-14186 Stockholm, Sweden
| | - Agné Kulyté
- Lipid Laboratory, Department of Medicine Huddinge, Karolinska Institutet, SE-14186 Stockholm, Sweden.
| |
Collapse
|
26
|
Maurizi G, Babini L, Della Guardia L. Potential role of microRNAs in the regulation of adipocytes liposecretion and adipose tissue physiology. J Cell Physiol 2018; 233:9077-9086. [PMID: 29932216 DOI: 10.1002/jcp.26523] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 01/31/2018] [Indexed: 12/19/2022]
Abstract
Adipose tissue is a dynamic endocrine organ playing a pivotal role in metabolism modulation. Adipocytes differentiation requires a highly orchestrated series of changes of gene expression in precursor cells. At the same time, white mature adipocytes are plastic cells able to reversibly transdifferentiate toward fibroblast-like cells via the liposecretion process, returning back to a non-committed status of the cells. In particular, adipose tissue microenvironment along with external signaling molecules such as adipokines, cytokines and growth factors can regulate adipocytes physiology through complex molecular networks. MicroRNAs (miRNAs), a type of non-coding RNA, acting as fine regulators of biological processes and their expression is sensible to the environment and cellular status changes. MiRNAs are thought to play a pivotal role in regulating the physiology of adipose tissue as well as in the development of obesity and associated metabolic disturbances, although the underlying mechanisms have not been identified so far. Elucidating the molecular mechanisms orchestrating adipose tissue biology is required to better characterize obesity and its associated diseases. In this respect, the review aims to analyze the microRNAs potentially involved in adipogenesis highlighting their role in the process of liposecretion, adipocyte proliferation, and adipokines secretion. The role of microRNAs in the development of obesity and obesity-associated disorders is also discussed.
Collapse
Affiliation(s)
| | - Lucia Babini
- Università Politecnica delle Marche, Ancona, Italy
| | - Lucio Della Guardia
- Dipartimento di Sanità Pubblica, Medicina Sperimentale e Forense, Unità di Scienza dell'Alimentazione, Università degli studi di Pavia, Pavia, Italy
| |
Collapse
|
27
|
Zhao W, Yang H, Li J, Chen Y, Cao J, Zhong T, Wang L, Guo J, Li L, Zhang H. MiR-183 promotes preadipocyte differentiation by suppressing Smad4 in goats. Gene 2018; 666:158-164. [PMID: 29751096 DOI: 10.1016/j.gene.2018.05.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Revised: 04/23/2018] [Accepted: 05/07/2018] [Indexed: 12/30/2022]
Abstract
As a well-conserved microRNA, miR-183 is ubiquitously expressed in many tissues and cells including backfat and the 3T3-L1 adipocytes; however, the mechanisms regulating miR-183 in adipogenesis remain poorly understood. Here, we explored the expression pattern and role of miR-183 in adipogenesis using hircine preadipocytes. The results showed that miR-183 was up-regulated during preadipocyte differentiation, and overexpression of miR-183 enhanced lipid accumulation and dramatically increased the mRNA expression of the adipogenic genes PPARγ, C/EBPα, SREBP-1c, FAS, and ACC. Using bioinformatics tools, we predicted Smad4 to be a target of miR-183. This was subsequently validated with a luciferase reporter assay. Overexpression of miR-183 suppressed the mRNA and protein levels of Smad4 significantly, whereas inhibiting miR-183 had the opposite effect. However, inhibition of Smad4 greatly accelerated lipid deposition and increased the expression of adipogenic genes which consists with the results of miR-183 overexpression. In conclusion, these results indicate that miR-183 promotes hircine preadipocyte differentiation by targeting Smad4.
Collapse
Affiliation(s)
- Wei Zhao
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Hailong Yang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Juntao Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Yuan Chen
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Jiaxue Cao
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Tao Zhong
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Linjie Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Jiazhong Guo
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Li Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China.
| | - Hongping Zhang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China.
| |
Collapse
|
28
|
MicroRNA-27a promotes renal tubulointerstitial fibrosis via suppressing PPARγ pathway in diabetic nephropathy. Oncotarget 2018; 7:47760-47776. [PMID: 27351287 PMCID: PMC5216977 DOI: 10.18632/oncotarget.10283] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 06/12/2016] [Indexed: 12/24/2022] Open
Abstract
MicroRNA-27a (miR-27a) upregulation has been identified in diabetes, but the pathogenesis of miR-27a in renal tubulointerstitial fibrosis (TIF) in diabetic nephropathy (DN) has not been elucidated. Herein, we found that high glucose stimulated miR-27a expression, which directly inhibited PPARγ and promoted fibrosis in NRK-52E cells. The functional relevance of miR-27a-dependent PPARγ decrease was proven by inhibition or overexpression of miR-27a both in vitro and in streptozotocin-induced diabetic rats. MiR-27a, via repression of PPARγ, activates the TGF-β/Smad3 signaling and contributes to the expressional changes of connective tissue growth factor (CTGF), Fibronectin and Collagen I, key mediators of fibrosis. Furthermore, we provide evidences that plasma miR-27a upregulation contributed to unfavorable renal function and increased TIF in renal tissues of diabetic rats and DN patients. Notably, miR-27a exhibited clinical and biological relevance as it was linked to elevated serum creatinine, proteinuria, urinary N-acetyl-β-D-glucosaminidase (NAG), and reduced estimated glomerular filtration rate (eGFR). Thus, we propose a novel role of the miR-27a-PPARγ axis in fostering the progression toward more deteriorated renal TIF in DN. Monitoring plasma miR-27a level and its association with PPARγ can be used to reflect the severity of renal TIF. Targeting miR-27a could be evaluated as a potential therapeutic approach for DN.
Collapse
|
29
|
Chronic hyperinsulinemia induced miR-27b is linked to adipocyte insulin resistance by targeting insulin receptor. J Mol Med (Berl) 2018; 96:315-331. [PMID: 29455245 DOI: 10.1007/s00109-018-1623-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 02/01/2018] [Accepted: 02/05/2018] [Indexed: 12/31/2022]
Abstract
Defect in insulin signaling leads to the development of insulin resistance followed by type 2 diabetes. Exploiting our previously developed physiological chronic hyperinsulinemia (CI)-mediated insulin resistance (IR) model, we wanted to understand how miRNAs contribute to the development of IR. Amongst the identified and validate miRNAs, the expression of miR-27b was found to be highly upregulated during CI-induced IR in 3T3-L1 adipocytes. We also validated the expression of miR-27b in CI-induced IR in human mesenchymal stem cell (hMSC)-derived adipocytes and in vivo high fat diet (HFD)-induced IR mice model. Bioinformatics target prediction softwares and luciferase reporter assay identified insulin receptor (INSR) as one of a prime target of miR-27b. Lentiviral mediated overexpression of miR-27b impairs insulin signaling by modulating INSR expression that in turn led to decreased glucose uptake in both 3T3-L1 and hMSC-derived adipocytes. Conversely, inhibition of miR-27b reversed CI-mediated suppression of target protein INSR and improved phosphorylation of Akt, a nodal protein of insulin signaling that is impaired by CI treatment. Lentiviral mediated overexpression of miR-27b in in vivo C57BL/6 mice impaired whole body glucose tolerance and adipose tissue insulin sensitivity. Furthermore, inhibition of miR-27b in HFD-induced insulin resistance mice model improved glucose tolerance and adipose tissue insulin sensitivity by increasing the expression of its target gene INSR in eWAT. Thus, our results indicate that miR-27b functions as a prime modulator of CI-induced IR via regulating the expression of INSR. KEY MESSAGES: miR-27b is upregulated in different in vitro and in vivo models of insulin resistance. miR-27b directly suppresses the expression of INSR by targeting 3'UTR of INSR. Modulation of miR-27b expression regulates insulin sensitivity by targeting INSR.
Collapse
|
30
|
Zaiou M, El Amri H, Bakillah A. The clinical potential of adipogenesis and obesity-related microRNAs. Nutr Metab Cardiovasc Dis 2018; 28:91-111. [PMID: 29170059 DOI: 10.1016/j.numecd.2017.10.015] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 10/12/2017] [Accepted: 10/15/2017] [Indexed: 02/07/2023]
Abstract
Obesity is a growing health problem commonly associated with numerous metabolic disorders including type 2 diabetes, hypertension, cardiovascular disease, and some forms of cancer. The burden of obesity and associated cardiometabolic diseases are believed to arise through complex interplay between genetics and epigenetics predisposition, nutrition, environment, and lifestyle. However, the molecular basis and the repertoire of obesity-affecting factors are still unknown. Emerging evidence is connecting microRNAs (miRNAs) dysregulation with adipogenesis and obesity. Alteration in miRNAs expression could result in changes in the pattern of genes controlling a range of biological processes including inflammation, lipid metabolism, insulin resistance and adipogenesis. Hence, understanding exact roles of miRNAs as well as the degree of their contribution to the regulation of adipogenesis and fat cell development in obesity would provide new therapeutic targets for the development of novel and effective anti-obesity drugs. The objective of the current review is to: (i) discuss some of the latest development on relevant miRNAs dysregulation mainly in human adipogenesis and obesity, (ii) emphasize the role of circulating miRNAs as new promising therapeutics and attractive potential biomarkers for treating obesity and associated risk factor diseases, (iii) describe how dietary factors may influence obesity through modulation of miRNAs expression, (iv) highlight some of the actual limitations to the promise of miRNAs as novel therapeutics as well as to their translation for the benefit of patients, and finally (v) provide recommendations for future research on miRNA-based therapeutics that could lead to a breakthrough in the treatment of obesity and its associated pathologies.
Collapse
Affiliation(s)
- M Zaiou
- Université de Lorraine, Faculté de Pharmacie, 5 rue Albert Lebrun, 54000, Nancy, France.
| | - H El Amri
- Laboratoire de Génétique de la Gendarmerie Royale, Avenue Ibn Sina, Agdal, Rabat, Morocco
| | - A Bakillah
- State University of New York, Downstate Medical Center, Department of Medicine, 450 Clarkson Ave., Brooklyn, NY, 11203, USA
| |
Collapse
|
31
|
Giardina S, Hernández-Alonso P, Salas-Salvadó J, Rabassa-Soler A, Bulló M. Modulation of Human Subcutaneous Adipose Tissue MicroRNA Profile Associated with Changes in Adiposity-Related Parameters. Mol Nutr Food Res 2017; 62. [PMID: 29024341 DOI: 10.1002/mnfr.201700594] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 09/22/2017] [Indexed: 12/23/2022]
Abstract
SCOPE To analyze the effect of three calorie-restricted diets with different amount and quality of carbohydrates on subcutaneous adipose tissue (SAT) microRNA (miRNA) profile. METHODS AND RESULTS 6-month parallel, randomized trial conducted on overweight and obese subjects randomized to: 1) low glycemic index diet (LGI), 2) high glycemic index diet (HGI), and 3) low-fat (LF). The genome-wide SAT miRNA profile was assessed in eight randomly selected participants and the most relevant changing miRNAs (n = 13) were validated in 48 subjects. None of the miRNAs showed significant changes between the intervention groups. However, changes in some of them correlated with changes in biochemical and anthropometric variables. Stratifying our population according to tertiles of percentage change in body weight (BW), we observed a significant down-regulation of miR-210 in those subjects in Tertile 1 as compared to Tertile 3. When our population was stratified by tertiles of waist circumference, miR-132, miR-29a, miR-34a, and miR-378 were found to be significantly down-regulated, in T2 compared to T3. Furthermore, when stratified by tertiles of fat mass, we also observed the significant down-regulation of miR-132 in T1. CONCLUSION The macronutrient composition of a calorie-restricted diet does not affect the expression of the miRNAs analyzed, while changes in adiposity play a primary regulatory role.
Collapse
Affiliation(s)
- Simona Giardina
- Human Nutrition Unit, Faculty of Medicine and Health Sciences, Institute of Health Pere Virgili, Universitat Rovira i Virgili, Reus, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Pablo Hernández-Alonso
- Human Nutrition Unit, Faculty of Medicine and Health Sciences, Institute of Health Pere Virgili, Universitat Rovira i Virgili, Reus, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Jordi Salas-Salvadó
- Human Nutrition Unit, Faculty of Medicine and Health Sciences, Institute of Health Pere Virgili, Universitat Rovira i Virgili, Reus, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Antoni Rabassa-Soler
- Human Nutrition Unit, Faculty of Medicine and Health Sciences, Institute of Health Pere Virgili, Universitat Rovira i Virgili, Reus, Spain
| | - Mònica Bulló
- Human Nutrition Unit, Faculty of Medicine and Health Sciences, Institute of Health Pere Virgili, Universitat Rovira i Virgili, Reus, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
32
|
Nazari M, Saberi A, Karandish M, Neisi N, Jalali MT, Makvandi M. Influence of L-carnitine on the Expression Level of Adipose Tissue miRNAs Related to Weight Changes in Obese Rats. Pak J Biol Sci 2017; 19:227-232. [PMID: 29023027 DOI: 10.3923/pjbs.2016.227.232] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND AND OBJECTIVE Molecular mechanisms of most anti-obesity drugs are remained to be clear. MicroRNAs that are noncoding RNA molecules supposed to regulate biological processes concomitant to obesity and have attracted a lot of attention to themselves. The miR-27a and miR-143 expression levels in obese and non-obese rats during weight changes and L-carnitine (LC) effects on them was investigated in this study. MATERIALS AND METHODS In the present study 12 male Wistar rats were randomly divided into normal fat diet and high fat diet groups to develop obesity. After 8 weeks rats were weighted and half of diet induced obese rats were randomly selected to receive 200 mg LC kg -1 b.wt. for 4 weeks. At the end epididymal fat was isolated to investigate expression level of microRNAs by real-time PCR. RESULTS After 12 weeks, high fat diet in comparison with normal fat diet mediated significant decrease and increase in expression levels of miR-27a and miR-143 , respectively. These changes were modified in groups, which had received LC in a 4 weeks period. Furthermore, rats in this group gained less weight. CONCLUSION Findings of this study suggest that the changes of microRNAs expression probably play a role in pathogenesis of obesity, might be modulated by means of dietary agents and supplements and modify weight gain trend.
Collapse
Affiliation(s)
- Maryam Nazari
- Nutrition and Metabolic Diseases Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Alihossein Saberi
- Department of Medical Genetics, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Golestan Highway, Ahvaz, Iran
| | - Majid Karandish
- Nutrition and Metabolic Diseases Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Niloofar Neisi
- Research Center for Infectious Diseases of Digestive System and Department of Virology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Taha Jalali
- Hyperlipidemia Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Manoochehr Makvandi
- Health Research Institute, Infectious and Tropical Diseases Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
33
|
Amri EZ, Scheideler M. Small non coding RNAs in adipocyte biology and obesity. Mol Cell Endocrinol 2017; 456:87-94. [PMID: 28412522 DOI: 10.1016/j.mce.2017.04.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 04/10/2017] [Accepted: 04/10/2017] [Indexed: 12/12/2022]
Abstract
Obesity has reached epidemic proportions world-wide and constitutes a substantial risk factor for hypertension, type 2 diabetes, cardiovascular diseases and certain cancers. So far, regulation of energy intake by dietary and pharmacological treatments has met limited success. The main interest of current research is focused on understanding the role of different pathways involved in adipose tissue function and modulation of its mass. Whole-genome sequencing studies revealed that the majority of the human genome is transcribed, with thousands of non-protein-coding RNAs (ncRNA), which comprise small and long ncRNAs. ncRNAs regulate gene expression at the transcriptional and post-transcriptional level. Numerous studies described the involvement of ncRNAs in the pathogenesis of many diseases including obesity and associated metabolic disorders. ncRNAs represent potential diagnostic biomarkers and promising therapeutic targets. In this review, we focused on small ncRNAs involved in the formation and function of adipocytes and obesity.
Collapse
Affiliation(s)
| | - Marcel Scheideler
- Institute for Diabetes and Cancer (IDC), Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany; Joint Heidelberg-IDC Translational Diabetes Program, University Hospital Heidelberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany.
| |
Collapse
|
34
|
Pahlavani M, Ramalho T, Koboziev I, LeMieux MJ, Jayarathne S, Ramalingam L, Filgueiras LR, Moustaid-Moussa N. Adipose tissue inflammation in insulin resistance: review of mechanisms mediating anti-inflammatory effects of omega-3 polyunsaturated fatty acids. J Investig Med 2017; 65:1021-1027. [PMID: 28954844 DOI: 10.1136/jim-2017-000535] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2017] [Indexed: 12/12/2022]
Abstract
Obesity is an increasingly costly and widespread epidemic, effecting 1 in 10 adults worldwide. It has been causally linked with both the metabolic syndrome and insulin resistance, both of which are associated with increased chronic inflammation. The exact mechanisms through which inflammation may contribute to both MetS and IR are numerous and their details are still largely unknown. Recently, micro-RNAs (miRNAs) have emerged as potential interventional targets due to their potential preventive roles in the pathogenesis of several diseases, including MetS and obesity. The purpose of this review paper is to discuss some of the known roles of miRNAs as mediators of inflammation-associated obesity and IR and how omega-3 polyunsaturated fatty acids may be used as a nutritional intervention for these disorders.
Collapse
Affiliation(s)
- Mandana Pahlavani
- Department of Nutritional Sciences and Obesity Research Cluster, Texas Tech University, Lubbock, Texas, USA
| | - Theresa Ramalho
- Department of Nutritional Sciences and Obesity Research Cluster, Texas Tech University, Lubbock, Texas, USA.,Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Iurii Koboziev
- Department of Nutritional Sciences and Obesity Research Cluster, Texas Tech University, Lubbock, Texas, USA
| | - Monique J LeMieux
- Department of Nutrition and Food Science, Texas Woman's University, Denton, Texas, USA
| | - Shasika Jayarathne
- Department of Nutritional Sciences and Obesity Research Cluster, Texas Tech University, Lubbock, Texas, USA
| | - Latha Ramalingam
- Department of Nutritional Sciences and Obesity Research Cluster, Texas Tech University, Lubbock, Texas, USA
| | - Luciano R Filgueiras
- Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Naima Moustaid-Moussa
- Department of Nutritional Sciences and Obesity Research Cluster, Texas Tech University, Lubbock, Texas, USA
| |
Collapse
|
35
|
Divoux A, Xie H, Li JL, Karastergiou K, Perera RJ, Chang RJ, Fried SK, Smith SR. MicroRNA-196 Regulates HOX Gene Expression in Human Gluteal Adipose Tissue. Obesity (Silver Spring) 2017; 25. [PMID: 28649807 PMCID: PMC5551414 DOI: 10.1002/oby.21896] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
OBJECTIVE Lower body fat is associated with diminishing cardiometabolic risk. Physiological differences between gluteofemoral and abdominal subcutaneous adipocyte functions are known, but the molecular basis for depot differences in adipocyte function is poorly understood. The objective of this study was to identify depot differences in microRNA (miRNA) expression in human abdominal and gluteofemoral subcutaneous adipose tissues and their implication in gene regulation. METHODS Abdominal and gluteofemoral adipose tissue aspirates obtained from 18 participants (9 male and 9 female, age 30 ± 1.5 y, BMI 27.3 ± 1.23 kg/m2 ) were analyzed for miRNA expression profiles by next-generation DNA sequencing. The raw reads were mapped to miRBase 17, and differentially expressed miRNAs were confirmed by qRT-PCR. The hsa-mimic-miR196a was transfected into cultured abdominal preadipocytes isolated from five women with obesity. Target gene expression was evaluated by RT-qPCR. RESULTS Among the 640 miRNAs detected in adipose tissue, miR196a2, miR196a1, miR196b, and miR204 showed a higher expression in the gluteofemoral depot (fold change = 2.7, 2.3, 1.7, and 2.3, respectively) independent of sex. Bioinformatic analyses and human primary preadipocyte transfection with miR196 suggested that the differentially expressed miRNAs could directly or indirectly modulate homeobox (HOX) gene expression. CONCLUSIONS The miR196 gene family could play an important role in the regulation of HOX gene expression in subcutaneous adipose tissue and in fat distribution variation.
Collapse
Affiliation(s)
- Adeline Divoux
- Translational Research Institute for Metabolism and Diabetes, Florida Hospital, Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida, USA
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida, USA
| | - Hui Xie
- Translational Research Institute for Metabolism and Diabetes, Florida Hospital, Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida, USA
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida, USA
| | - Jian-Liang Li
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida, USA
| | - Kalypso Karastergiou
- Obesity Center, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Ranjan J Perera
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida, USA
| | - R Jeffrey Chang
- Division of Reproductive Endocrinology, Department of Reproductive Medicine, University of California, San Diego, La Jolla, California, USA
| | - Susan K Fried
- Obesity Center, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Steven R Smith
- Translational Research Institute for Metabolism and Diabetes, Florida Hospital, Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida, USA
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida, USA
| |
Collapse
|
36
|
Del Carmen Martínez-Jiménez V, Méndez-Mancilla A, Patricia Portales-Pérez D. miRNAs in nutrition, obesity, and cancer: The biology of miRNAs in metabolic disorders and its relationship with cancer development. Mol Nutr Food Res 2017; 62. [PMID: 28594107 DOI: 10.1002/mnfr.201600994] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 04/13/2017] [Accepted: 05/08/2017] [Indexed: 12/17/2022]
Abstract
SCOPE The scope of this review is to explain how metabolic disorders originated by a deficient nutrition can develop into a neoplastic process by the alteration of epigenetic mechanisms like miRNAs. Obesity is a proinflammatory state with a wide impact on health around the world that is associated with neoplastic diseases. Epigenetic mechanisms have a central role in the obesogenic environment, which participates on the development of comorbidities such as cancer. METHODS AND RESULTS We made an exhaustive review of the most recent reports about metabolic disorders with nutrition and their relationship with miRNAs, and their risk of developing into oncogenic processes. MicroRNAs (miRNAs) act as one of the major epigenetic mechanisms that can affect the metabolic reprogramming of cellular metabolism that plays an important role in the oncogenic process. There is evidence that some foods may contribute to diminishing the risk of cancer as well as epidemiological studies that support the notion that diets high in animal protein and fat promote cancer risk. Therefore, diets high in fruit and vegetables reduce the risk of cancer. One of the principal explanations is that these foods contain bioactive compounds that increase the efficacy of epigenetic mechanisms, which in turn decrease the risk of obesity and its comorbidities. CONCLUSION In this review, we show how miRNAs are implicated in several signaling pathways as well as illustrating some bioactive compounds that impact inflammation and cancer development.
Collapse
Affiliation(s)
| | - Alejandro Méndez-Mancilla
- Laboratorio de Inmunología y Biología Celular y Molecular, Facultad de Ciencias Químicas, UASLP San Luis Potosí, SLP México
| | - Diana Patricia Portales-Pérez
- Laboratorio de Inmunología y Biología Celular y Molecular, Facultad de Ciencias Químicas, UASLP San Luis Potosí, SLP México
| |
Collapse
|
37
|
|
38
|
Dahlman I, Belarbi Y, Laurencikiene J, Pettersson AM, Arner P, Kulyté A. Comprehensive functional screening of miRNAs involved in fat cell insulin sensitivity among women. Am J Physiol Endocrinol Metab 2017; 312:E482-E494. [PMID: 28270439 DOI: 10.1152/ajpendo.00251.2016] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 01/18/2017] [Accepted: 02/22/2017] [Indexed: 01/12/2023]
Abstract
The key pathological link between obesity and type 2 diabetes is insulin resistance, but the molecular mechanisms are not entirely identified. micro-RNAs (miRNA) are dysregulated in obesity and may contribute to insulin resistance. Our objective was to detect and functionally investigate miRNAs linked to insulin sensitivity in human subcutaneous white adipose tissue (scWAT). Subjects were selected based on the insulin-stimulated lipogenesis response of subcutaneous adipocytes. Global miRNA profiling was performed in abdominal scWAT of 18 obese insulin-resistance (OIR), 21 obese insulin-sensitive (OIS), and 9 lean women. miRNAs demonstrating differential expression between OIR and OIS women were overexpressed in human in vitro-differentiated adipocytes followed by assessment of lipogenesis and identification of miRNA targets by measuring mRNA/protein expression and 3'-untranslated region analysis. Eleven miRNAs displayed differential expression between OIR and OIS states. Overexpression of miR-143-3p and miR-652-3p increased insulin-stimulated lipogenesis in human in vitro differentiated adipocytes and directly or indirectly affected several genes/proteins involved in insulin signaling at transcriptional or posttranscriptional levels. Adipose expression of miR-143-3p and miR-652-3p was positively associated with insulin-stimulated lipogenesis in scWAT independent of body mass index. In conclusion, miR-143-3p and miR-652-3p are linked to scWAT insulin resistance independent of obesity and influence insulin-stimulated lipogenesis by interacting at different steps with insulin-signaling pathways.
Collapse
Affiliation(s)
- Ingrid Dahlman
- Lipid Laboratory, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Yasmina Belarbi
- Lipid Laboratory, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Jurga Laurencikiene
- Lipid Laboratory, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Annie M Pettersson
- Lipid Laboratory, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Peter Arner
- Lipid Laboratory, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Agné Kulyté
- Lipid Laboratory, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
39
|
Wilson RA, Deasy W, Hayes A, Cooke MB. High fat diet and associated changes in the expression of micro-RNAs in tissue: Lessons learned from animal studies. Mol Nutr Food Res 2017; 61. [PMID: 28233461 DOI: 10.1002/mnfr.201600943] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 01/15/2017] [Accepted: 02/13/2017] [Indexed: 12/13/2022]
Abstract
Environment and genetic factors play an important role in the development of obesity, and diet is one of the main contributing factors to this disease. High fat intake is associated with body weight gain, leading to obesity and other metabolic diseases. MicroRNAs (miRNAs) are a group of small, noncoding RNAs that are important regulators of gene expression at posttranscriptional level. Studies have shown that high fat intake, independent of body weight status, can significantly impact both negatively and positively the expression of miRNAs and thus the biological function of tissues such as adipose, skeletal, and cardiac muscle, liver, neuronal, and endothelial. This review will summarize the effects of high calorie diet in the form of high fat intake on miRNA expression in various tissues of animal models and of high fat fed offspring. We will also briefly review the impact of different dietary lipids on miRNA expression. Given changes in miRNA expression have been associated with the development of many diseases including obesity, understanding their biological role could have important clinical implications and offer tangible therapeutic targets for the prevention, management, and/or treatment of obesity and other lifestyle-related disorders.
Collapse
Affiliation(s)
- Robin A Wilson
- College of Health and Biomedicine, Victoria University, Melbourne, VIC, Australia.,Australian Institute for Musculoskeletal Science (AIMSS), Western Health, Melbourne, VIC, Australia
| | - William Deasy
- College of Health and Biomedicine, Victoria University, Melbourne, VIC, Australia.,Australian Institute for Musculoskeletal Science (AIMSS), Western Health, Melbourne, VIC, Australia
| | - Alan Hayes
- College of Health and Biomedicine, Victoria University, Melbourne, VIC, Australia.,Australian Institute for Musculoskeletal Science (AIMSS), Western Health, Melbourne, VIC, Australia
| | - Matthew B Cooke
- College of Health and Biomedicine, Victoria University, Melbourne, VIC, Australia.,Australian Institute for Musculoskeletal Science (AIMSS), Western Health, Melbourne, VIC, Australia
| |
Collapse
|
40
|
Trohatou O, Zagoura D, Orfanos NK, Pappa KI, Marinos E, Anagnou NP, Roubelakis MG. miR-26a Mediates Adipogenesis of Amniotic Fluid Mesenchymal Stem/Stromal Cells via PTEN, Cyclin E1, and CDK6. Stem Cells Dev 2017; 26:482-494. [PMID: 28068868 DOI: 10.1089/scd.2016.0203] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Recent findings indicate that microRNAs (miRNAs) are critical for the regulatory network of adipogenesis in human mesenchymal stem/stromal cells (MSCs). Fetal MSCs derived from amniotic fluid (AF-MSCs) represent a population of multipotent stem cells characterized by a wide range of differentiation properties that can be applied in cell-based therapies. In this study, miRNA microarray analysis was performed to assess miRNA expression in terminal differentiated AF-MSCs into adipocyte-like cells (AL cells). MiR-26a was identified in high expression levels in AL cells indicating a critical role in the process of adipogenesis. Overexpression of miR-26a in AF-MSCs led to significant induction of their adipogenic differentiation properties that were altered after miR-26a inhibition. We have demonstrated that miR-26a regulates adipogenesis through direct inhibition of PTEN, which in turn promotes activation of Akt pathway. Also, miR-26a modulates cell cycle during adipogenesis by interacting with Cyclin E1 and CDK6. These results point to the regulatory role of miR-26a and its target genes PTEN, Cyclin E1, and CDK6 in adipogenic differentiation of AF-MSCs, providing a basis for understanding the mechanisms of fat cell development and obesity.
Collapse
Affiliation(s)
- Ourania Trohatou
- 1 Laboratory of Biology, School of Medicine, National and Kapodistrian University of Athens , Athens, Greece .,2 Cell and Gene Therapy Laboratory, Centre of Basic Research II, Biomedical Research Foundation of the Academy of Athens (BRFAA) , Athens, Greece
| | - Dimitra Zagoura
- 1 Laboratory of Biology, School of Medicine, National and Kapodistrian University of Athens , Athens, Greece .,2 Cell and Gene Therapy Laboratory, Centre of Basic Research II, Biomedical Research Foundation of the Academy of Athens (BRFAA) , Athens, Greece
| | - Nikos K Orfanos
- 2 Cell and Gene Therapy Laboratory, Centre of Basic Research II, Biomedical Research Foundation of the Academy of Athens (BRFAA) , Athens, Greece
| | - Kalliopi I Pappa
- 3 First Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens , Athens, Greece
| | - Evangelos Marinos
- 1 Laboratory of Biology, School of Medicine, National and Kapodistrian University of Athens , Athens, Greece
| | - Nicholas P Anagnou
- 1 Laboratory of Biology, School of Medicine, National and Kapodistrian University of Athens , Athens, Greece .,2 Cell and Gene Therapy Laboratory, Centre of Basic Research II, Biomedical Research Foundation of the Academy of Athens (BRFAA) , Athens, Greece
| | - Maria G Roubelakis
- 1 Laboratory of Biology, School of Medicine, National and Kapodistrian University of Athens , Athens, Greece .,2 Cell and Gene Therapy Laboratory, Centre of Basic Research II, Biomedical Research Foundation of the Academy of Athens (BRFAA) , Athens, Greece
| |
Collapse
|
41
|
Flavonoid derivative (Fla-CN) inhibited adipocyte differentiation via activating AMPK and up-regulating microRNA-27 in 3T3-L1 cells. Eur J Pharmacol 2017; 797:45-52. [DOI: 10.1016/j.ejphar.2017.01.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 01/03/2017] [Accepted: 01/11/2017] [Indexed: 12/25/2022]
|
42
|
Tucci A, Ciaccio C, Scuvera G, Esposito S, Milani D. MIR137 is the key gene mediator of the syndromic obesity phenotype of patients with 1p21.3 microdeletions. Mol Cytogenet 2016; 9:80. [PMID: 27822311 PMCID: PMC5093957 DOI: 10.1186/s13039-016-0289-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 10/25/2016] [Indexed: 01/22/2023] Open
Abstract
Background Deletions in the long arm of chromosome 1 have been described in patients with a phenotype consisting primarily of obesity, intellectual disability and autism-spectrum disorder. The minimal region of overlap comprises two genes: DPYD and MIR137. Case presentation We describe a 10-year-old boy with syndromic obesity who carries a novel 1p21.3 deletion overlapping the critical region with the MIR137 gene only. Conclusions This study suggests that MIR137 is the mediator of the obesity phenotype of patients carrying 1p21.3 microdeletions.
Collapse
Affiliation(s)
- Arianna Tucci
- Pediatric Highly Intensive Care Unit, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Commenda 9, 20122 Milano, Italy
| | - Claudia Ciaccio
- Pediatric Highly Intensive Care Unit, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Commenda 9, 20122 Milano, Italy
| | - Giulietta Scuvera
- Pediatric Highly Intensive Care Unit, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Commenda 9, 20122 Milano, Italy
| | - Susanna Esposito
- Pediatric Highly Intensive Care Unit, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Commenda 9, 20122 Milano, Italy
| | - Donatella Milani
- Pediatric Highly Intensive Care Unit, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Commenda 9, 20122 Milano, Italy
| |
Collapse
|
43
|
Jones Buie JN, Goodwin AJ, Cook JA, Halushka PV, Fan H. The role of miRNAs in cardiovascular disease risk factors. Atherosclerosis 2016; 254:271-281. [PMID: 27693002 PMCID: PMC5125538 DOI: 10.1016/j.atherosclerosis.2016.09.067] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 08/31/2016] [Accepted: 09/22/2016] [Indexed: 12/12/2022]
Abstract
Coronary artery disease and atherosclerosis are complex pathologies that develop over time due to genetic and environmental factors. Differential expression of miRNAs has been identified in patients with coronary artery disease and atherosclerosis, however, their association with cardiovascular disease risk factors, including hyperlipidemia, hypertension, obesity, diabetes, lack of physical activity and smoking, remains unclear. This review examines the role of miRNAs as either biomarkers or potential contributors to the pathophysiology of these aforementioned risk factors. It is intended to provide an overview of the published literature which describes alterations in miRNA levels in both human and animal studies of cardiovascular risk factors and when known, the possible mechanism by which these miRNAs may exert either beneficial or deleterious effects. The intent of this review is engage clinical, translational, and basic scientists to design future collaborative studies to further elucidate the potential role of miRNAs in cardiovascular diseases.
Collapse
Affiliation(s)
- Joy N Jones Buie
- Medical University of South Carolina, Department of Pathology and Laboratory Medicine, 173 Ashley Avenue, Suite CRI 605B, Charleston, United States.
| | - Andrew J Goodwin
- Medical University of South Carolina, Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Charleston, United States
| | - James A Cook
- Medical University of South Carolina, Department of Neurosciences, Charleston, United States
| | - Perry V Halushka
- Medical University of South Carolina, Department of Pharmacology, Charleston, United States
| | - Hongkuan Fan
- Medical University of South Carolina, Department of Pathology and Laboratory Medicine, 173 Ashley Avenue, Suite CRI 605B, Charleston, United States
| |
Collapse
|
44
|
miR33a/miR33b* and miR122 as Possible Contributors to Hepatic Lipid Metabolism in Obese Women with Nonalcoholic Fatty Liver Disease. Int J Mol Sci 2016; 17:ijms17101620. [PMID: 27669236 PMCID: PMC5085653 DOI: 10.3390/ijms17101620] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 09/13/2016] [Accepted: 09/13/2016] [Indexed: 02/06/2023] Open
Abstract
Specific miRNA expression profiles have been shown to be associated with nonalcoholic fatty liver disease (NAFLD). We examined the correlation between the circulating levels and hepatic expression of miR122 and miR33a/b*, the key lipid metabolism-related gene expression and the clinicopathological factors of obese women with NAFLD. We measured miR122 and miR33a/b* expression in liver samples from 62 morbidly obese (MO), 30 moderately obese (ModO), and eight normal-weight controls. MiR122 and miR33a/b* expression was analyzed by qRT-PCR. Additionally, miR122 and miR33b* circulating levels were analyzed in 122 women. Hepatic miR33b* expression was increased in MO compared to ModO and controls, whereas miR122 expression was decreased in the MO group compared to ModO. In obese cohorts, miR33b* expression was increased in nonalcoholic steatohepatitis (NASH). Regarding circulating levels, MO patients with NASH showed higher miR122 levels than MO with simple steatosis (SS). These circulating levels are good predictors of histological features associated with disease severity. MO is associated with altered hepatic miRNA expression. In obese women, higher miR33b* liver expression is associated with NASH. Moreover, multiple correlations between miRNAs and the expression of genes related to lipid metabolism were found, that would suggest a miRNA-host gene circuit. Finally, miR122 circulating levels could be included in a panel of different biomarkers to improve accuracy in the non-invasive diagnosis of NASH.
Collapse
|
45
|
Let-7i-5p represses brite adipocyte function in mice and humans. Sci Rep 2016; 6:28613. [PMID: 27345691 PMCID: PMC4921928 DOI: 10.1038/srep28613] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 06/06/2016] [Indexed: 02/04/2023] Open
Abstract
In response to cold or β3-adrenoreceptor stimulation brown adipose tissue (BAT) promotes non-shivering thermogenesis, leading to energy dissipation. BAT has long been thought to be absent or scarce in adult humans. The recent discovery of thermogenic brite/beige adipocytes has opened the way to development of novel innovative strategies to combat overweight/obesity and associated diseases. Thus it is of great interest to identify regulatory factors that govern the brite adipogenic program. Here, we carried out global microRNA (miRNA) expression profiling on human adipocytes to identify miRNAs that are regulated upon the conversion from white to brite adipocytes. Among the miRNAs that were differentially expressed, we found that Let-7i-5p was down regulated in brite adipocytes. A detailed analysis of the Let-7i-5p levels showed an inverse expression of UCP1 in murine and human brite adipocytes both in vivo and in vitro. Functional studies with Let-7i-5p mimic in human brite adipocytes in vitro revealed a decrease in the expression of UCP1 and in the oxygen consumption rate. Moreover, the Let-7i-5p mimic when injected into murine sub-cutaneous white adipose tissue inhibited partially β3-adrenergic activation of the browning process. These results suggest that the miRNAs Let-7i-5p participates in the recruitment and the function of brite adipocytes.
Collapse
|
46
|
Giroud M, Pisani DF, Karbiener M, Barquissau V, Ghandour RA, Tews D, Fischer-Posovszky P, Chambard JC, Knippschild U, Niemi T, Taittonen M, Nuutila P, Wabitsch M, Herzig S, Virtanen KA, Langin D, Scheideler M, Amri EZ. miR-125b affects mitochondrial biogenesis and impairs brite adipocyte formation and function. Mol Metab 2016; 5:615-625. [PMID: 27656399 PMCID: PMC5021678 DOI: 10.1016/j.molmet.2016.06.005] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 06/06/2016] [Accepted: 06/08/2016] [Indexed: 12/17/2022] Open
Abstract
Objective In rodents and humans, besides brown adipose tissue (BAT), islands of thermogenic adipocytes, termed “brite” (brown-in-white) or beige adipocytes, emerge within white adipose tissue (WAT) after cold exposure or β3-adrenoceptor stimulation, which may protect from obesity and associated diseases. microRNAs are novel modulators of adipose tissue development and function. The purpose of this work was to characterize the role of microRNAs in the control of brite adipocyte formation. Methods/Results Using human multipotent adipose derived stem cells, we identified miR-125b-5p as downregulated upon brite adipocyte formation. In humans and rodents, miR-125b-5p expression was lower in BAT than in WAT. In vitro, overexpression and knockdown of miR-125b-5p decreased and increased mitochondrial biogenesis, respectively. In vivo, miR-125b-5p levels were downregulated in subcutaneous WAT and interscapular BAT upon β3-adrenergic receptor stimulation. Injections of an miR-125b-5p mimic and LNA inhibitor directly into WAT inhibited and increased β3-adrenoceptor-mediated induction of UCP1, respectively, and mitochondrial brite adipocyte marker expression and mitochondriogenesis. Conclusion Collectively, our results demonstrate that miR-125b-5p plays an important role in the repression of brite adipocyte function by modulating oxygen consumption and mitochondrial gene expression. miR-125b-5p levels negatively correlate with UCP1 expression in rodent and human. miR125b levels in white adipose tissue are positively correlated with BMI. miR-125b-5p modulates oxygen consumption. Mitochondriogenesis is controlled by miR-125b-5p. In vivo modulation of miR-125b-5p controls brown and brite adipocyte formation.
Collapse
Affiliation(s)
- Maude Giroud
- Univ. Nice Sophia Antipolis, CNRS, Inserm, iBV, 06100 Nice, France
| | - Didier F Pisani
- Univ. Nice Sophia Antipolis, CNRS, Inserm, iBV, 06100 Nice, France
| | - Michael Karbiener
- Department of Phoniatrics, ENT University Hospital, Medical University Graz, Graz, Austria
| | - Valentin Barquissau
- Inserm, UMR1048, Obesity Research Laboratory, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France; University of Toulouse, UMR1048, Paul Sabatier University, Toulouse, France
| | | | - Daniel Tews
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, D-89075 Ulm, Germany
| | - Pamela Fischer-Posovszky
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, D-89075 Ulm, Germany
| | | | - Uwe Knippschild
- Department of General and Visceral Surgery, Ulm University Surgery Center, D-89075 Ulm, Germany
| | - Tarja Niemi
- Department of Endocrinology, Turku University Hospital, Turku, 20521, Finland
| | - Markku Taittonen
- Department of Endocrinology, Turku University Hospital, Turku, 20521, Finland
| | - Pirjo Nuutila
- Department of Endocrinology, Turku University Hospital, Turku, 20521, Finland; Turku University Hospital, Turku, Finland
| | - Martin Wabitsch
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, D-89075 Ulm, Germany
| | - Stephan Herzig
- Institute for Diabetes and Cancer (IDC), Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany; Joint Heidelberg-IDC Translational Diabetes Program, Heidelberg University Hospital, Heidelberg, Germany; Molecular Metabolic Control, Medical Faculty, Technical University Munich, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Kirsi A Virtanen
- Department of Endocrinology, Turku University Hospital, Turku, 20521, Finland; Turku PET Centre, University of Turku, Turku, Finland
| | - Dominique Langin
- Inserm, UMR1048, Obesity Research Laboratory, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France; University of Toulouse, UMR1048, Paul Sabatier University, Toulouse, France; Toulouse University Hospitals, Department of Clinical Biochemistry, Toulouse, France
| | - Marcel Scheideler
- Institute for Diabetes and Cancer (IDC), Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany; Joint Heidelberg-IDC Translational Diabetes Program, Heidelberg University Hospital, Heidelberg, Germany; Molecular Metabolic Control, Medical Faculty, Technical University Munich, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Ez-Zoubir Amri
- Univ. Nice Sophia Antipolis, CNRS, Inserm, iBV, 06100 Nice, France.
| |
Collapse
|
47
|
Shi L, Kim AJ, Chang RCA, Chang JYA, Ying W, Ko ML, Zhou B, Ko GYP. Deletion of miR-150 Exacerbates Retinal Vascular Overgrowth in High-Fat-Diet Induced Diabetic Mice. PLoS One 2016; 11:e0157543. [PMID: 27304911 PMCID: PMC4909316 DOI: 10.1371/journal.pone.0157543] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 06/01/2016] [Indexed: 02/07/2023] Open
Abstract
Diabetic retinopathy (DR) is the leading cause of blindness among American adults above 40 years old. The vascular complication in DR is a major cause of visual impairment, making finding therapeutic targets to block pathological angiogenesis a primary goal for developing DR treatments. MicroRNAs (miRs) have been proposed as diagnostic biomarkers and potential therapeutic targets for various ocular diseases including DR. In diabetic animals, the expression levels of several miRs, including miR-150, are altered. The expression of miR-150 is significantly suppressed in pathological neovascularization in mice with hyperoxia-induced retinopathy. The purpose of this study was to investigate the functional role of miR-150 in the development of retinal microvasculature complications in high-fat-diet (HFD) induced type 2 diabetic mice. Wild type (WT) and miR-150 null mutant (miR-150-/-) male mice were given a HFD (59% fat calories) or normal chow diet. Chronic HFD caused a decrease of serum miR-150 in WT mice. Mice on HFD for 7 months (both WT and miR-150-/-) had significant decreases in retinal light responses measured by electroretinograms (ERGs). The retinal neovascularization in miR-150-/--HFD mice was significantly higher compared to their age matched WT-HFD mice, which indicates that miR-150 null mutation exacerbates chronic HFD-induced neovascularization in the retina. Overexpression of miR-150 in cultured endothelial cells caused a significant reduction of vascular endothelial growth factor receptor 2 (VEGFR2) protein levels. Hence, deletion of miR-150 significantly increased the retinal pathological angiogenesis in HFD induced type 2 diabetic mice, which was in part through VEGFR2.
Collapse
Affiliation(s)
- Liheng Shi
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, United States of America
| | - Andy Jeesu Kim
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, United States of America
| | - Richard Cheng-An Chang
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, United States of America
| | - Janet Ya-An Chang
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, United States of America
| | - Wei Ying
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, United States of America
| | - Michael L. Ko
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, United States of America
| | - Beiyan Zhou
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, United States of America
- Department of Immunology, University of Connecticut Health Center School of Medicine, Farmington, Connecticut, United States of America
| | - Gladys Yi-Ping Ko
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, United States of America
- Texas A&M Institute of Neuroscience, Texas A&M University, College Station, Texas 77843–4458, United States of America
| |
Collapse
|
48
|
Brandon-Warner E, Feilen NA, Culberson CR, Field CO, deLemos AS, Russo MW, Schrum LW. Processing of miR17-92 Cluster in Hepatic Stellate Cells Promotes Hepatic Fibrogenesis During Alcohol-Induced Injury. Alcohol Clin Exp Res 2016; 40:1430-42. [PMID: 27291156 DOI: 10.1111/acer.13116] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 04/27/2016] [Indexed: 12/13/2022]
Abstract
BACKGROUND Exposure to alcohol and its metabolites can initiate hepatic injury and fibrogenesis. Fibrosis is mediated through hepatic stellate cell (HSC) activation, leading to global changes in mRNA and microRNA (miR) expression. miRs are expressed in cells or shuttled to exosomes which can be detected in tissue culture media (TCM) and biological fluids. The mechanisms and function underlying the differential expression and processing of miRs and their downstream effects during hepatic injury remain poorly understood. METHODS Expression of primary (pri)-miR17-92. and individual members of this cluster, miR17a, 18a, 19a, 20a, 19b, and 92, were examined in primary HSCs and human LX2 cells exposed to alcohol-conditioned media (CM), liver tissue from a rodent model of alcoholic injury, and in exosomes from TCM and plasma of rodent models and patients with alcoholic liver disease (ALD). miR expression was examined in HSCs transduced with an AAV2 vector carrying GFP-miR19b or GFP-control transgene under the collagen promoter. RESULTS Profibrotic markers were enhanced in primary HSCs and LX2 cells exposed to alcohol-CM, concomitant with decreased miR19b expression and a significant increase in pri-miR17-92. Increased pri-miR17-92 was confirmed in a rodent model of alcohol-induced liver injury. Individual members of the cluster were inversely proportionate in cells and exosomes. AAV2-mediated miR19b overexpression inhibited miR17-92 and altered expression of individual cluster members in cells and exosomes. Expression of individual miR17-92 cluster members in plasma exosomes isolated from patients with ALD was similar to that seen in a rodent model of alcoholic injury and in vitro. CONCLUSIONS Reintroduction of miR19b inhibits HSC activation and modulates expression of pri-miR17-92 and the inverse expression of individual cluster members in cells and exosomes. Better understanding of miR17-92 processing may provide mechanistic insights into the role of individual miRs and exosomes during hepatic injury, revealing new therapeutic targets.
Collapse
Affiliation(s)
- Elizabeth Brandon-Warner
- Liver Pathobiology Laboratory, Department of Internal Medicine, Carolinas Medical Center, Charlotte, North Carolina
| | - Nicole A Feilen
- Liver Pathobiology Laboratory, Department of Internal Medicine, Carolinas Medical Center, Charlotte, North Carolina
| | - Catherine R Culberson
- Liver Pathobiology Laboratory, Department of Internal Medicine, Carolinas Medical Center, Charlotte, North Carolina
| | - Conroy O Field
- Liver Pathobiology Laboratory, Department of Internal Medicine, Carolinas Medical Center, Charlotte, North Carolina
| | - Andrew S deLemos
- Center for Liver Diseases and Liver Transplant, Carolinas Medical Center, Charlotte, North Carolina
| | - Mark W Russo
- Center for Liver Diseases and Liver Transplant, Carolinas Medical Center, Charlotte, North Carolina
| | - Laura W Schrum
- Liver Pathobiology Laboratory, Department of Internal Medicine, Carolinas Medical Center, Charlotte, North Carolina
| |
Collapse
|
49
|
Tabet F, Cuesta Torres LF, Ong KL, Shrestha S, Choteau SA, Barter PJ, Clifton P, Rye KA. High-Density Lipoprotein-Associated miR-223 Is Altered after Diet-Induced Weight Loss in Overweight and Obese Males. PLoS One 2016; 11:e0151061. [PMID: 26962854 PMCID: PMC4786204 DOI: 10.1371/journal.pone.0151061] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 02/09/2016] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND AND AIMS microRNAs (miRNAs) are small, endogenous non-coding RNAs that regulate metabolic processes, including obesity. The levels of circulating miRNAs are affected by metabolic changes in obesity, as well as in diet-induced weight loss. Circulating miRNAs are transported by high-density lipoproteins (HDL) but the regulation of HDL-associated miRNAs after diet-induced weight loss has not been studied. We aim to determine if HDL-associated miR-16, miR-17, miR-126, miR-222 and miR-223 levels are altered by diet-induced weight loss in overweight and obese males. METHODS HDL were isolated from 47 subjects following 12 weeks weight loss comparing a high protein diet (HP, 30% of energy) with a normal protein diet (NP, 20% of energy). HDL-associated miRNAs (miR-16, miR-17, miR-126, miR-222 and miR-223) at baseline and after 12 weeks of weight loss were quantified by TaqMan miRNA assays. HDL particle sizes were determined by non-denaturing polyacrylamide gradient gel electrophoresis. Serum concentrations of human HDL constituents were measured immunoturbidometrically or enzymatically. RESULTS miR-16, miR-17, miR-126, miR-222 and miR-223 were present on HDL from overweight and obese subjects at baseline and after 12 weeks of the HP and NP weight loss diets. The HP diet induced a significant decrease in HDL-associated miR-223 levels (p = 0.015), which positively correlated with changes in body weight (r = 0.488, p = 0.032). Changes in miR-223 levels were not associated to changes in HDL composition or size. CONCLUSION HDL-associated miR-223 levels are significantly decreased after HP diet-induced weight loss in overweight and obese males. This is the first study reporting changes in HDL-associated miRNA levels with diet-induced weight loss.
Collapse
Affiliation(s)
- Fatiha Tabet
- Lipid Research Group, School of Medical Sciences, University of New South Wales Australia, Randwick, New South Wales, Australia
- * E-mail: ;
| | - Luisa F. Cuesta Torres
- Lipid Research Group, School of Medical Sciences, University of New South Wales Australia, Randwick, New South Wales, Australia
| | - Kwok Leung Ong
- Lipid Research Group, School of Medical Sciences, University of New South Wales Australia, Randwick, New South Wales, Australia
| | - Sudichhya Shrestha
- Lipid Research Group, School of Medical Sciences, University of New South Wales Australia, Randwick, New South Wales, Australia
| | - Sébastien A. Choteau
- Lipid Research Group, School of Medical Sciences, University of New South Wales Australia, Randwick, New South Wales, Australia
| | - Philip J. Barter
- Lipid Research Group, School of Medical Sciences, University of New South Wales Australia, Randwick, New South Wales, Australia
| | - Peter Clifton
- School of Pharmacy and Medical Sciences, Division of Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Kerry-Anne Rye
- Lipid Research Group, School of Medical Sciences, University of New South Wales Australia, Randwick, New South Wales, Australia
| |
Collapse
|
50
|
Gracia A, Miranda J, Fernández-Quintela A, Eseberri I, Garcia-Lacarte M, Milagro FI, Martínez JA, Aguirre L, Portillo MP. Involvement of miR-539-5p in the inhibition of de novo lipogenesis induced by resveratrol in white adipose tissue. Food Funct 2016; 7:1680-8. [DOI: 10.1039/c5fo01090j] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Proposed miRNA mechanisms of action of resveratrol in triacylglycerol metabolism changes in adipose tissue.
Collapse
Affiliation(s)
- Ana Gracia
- Nutrition and Obesity Group
- Department of Nutrition and Food Science
- University of the Basque Country (UPV/EHU) and Lucio Lacaray Research Institute
- Vitoria
- Spain
| | - Jonatan Miranda
- Nutrition and Obesity Group
- Department of Nutrition and Food Science
- University of the Basque Country (UPV/EHU) and Lucio Lacaray Research Institute
- Vitoria
- Spain
| | - Alfredo Fernández-Quintela
- Nutrition and Obesity Group
- Department of Nutrition and Food Science
- University of the Basque Country (UPV/EHU) and Lucio Lacaray Research Institute
- Vitoria
- Spain
| | - Itziar Eseberri
- Nutrition and Obesity Group
- Department of Nutrition and Food Science
- University of the Basque Country (UPV/EHU) and Lucio Lacaray Research Institute
- Vitoria
- Spain
| | - Marcos Garcia-Lacarte
- Department of Nutrition
- Food Sciences and Physiology
- Centre for Nutrition Research
- University of Navarra
- Pamplona
| | - Fermín I. Milagro
- Department of Nutrition
- Food Sciences and Physiology
- Centre for Nutrition Research
- University of Navarra
- Pamplona
| | - J. Alfredo Martínez
- Department of Nutrition
- Food Sciences and Physiology
- Centre for Nutrition Research
- University of Navarra
- Pamplona
| | - Leixuri Aguirre
- Nutrition and Obesity Group
- Department of Nutrition and Food Science
- University of the Basque Country (UPV/EHU) and Lucio Lacaray Research Institute
- Vitoria
- Spain
| | - María P. Portillo
- Nutrition and Obesity Group
- Department of Nutrition and Food Science
- University of the Basque Country (UPV/EHU) and Lucio Lacaray Research Institute
- Vitoria
- Spain
| |
Collapse
|