1
|
Lambert C, Morales-Sánchez P, García AV, Villa-Fernández E, Latorre J, García-Villarino M, Turienzo Santos EO, Suárez-Gutierrez L, Uría RR, Navarro SS, Ares-Blanco J, Pujante P, Sanz Álvarez LM, Menéndez-Torre E, Moreno Gijón M, Fernandez-Real JM, Delgado E. Exploring differential miRNA expression profiles in muscular and visceral adipose tissue of patients with severe obesity. Int J Obes (Lond) 2025; 49:634-641. [PMID: 39562687 PMCID: PMC11999863 DOI: 10.1038/s41366-024-01683-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 11/04/2024] [Accepted: 11/06/2024] [Indexed: 11/21/2024]
Abstract
BACKGROUND This study aims to investigate the differential miRNA expression profile between the visceral white adipose tissue and the skeletal muscle of people with obesity undergoing bariatric surgery. METHODS Skeletal muscle and visceral adipose tissue samples of 10 controls and 38 people with obesity (50% also with type 2 diabetes) undergoing bariatric surgery were collected. miRNA expression profiles were analyzed using Next-Generation Sequencing and subsequently validated using RT-PCR. RESULTS Approximately 69% of miRNAs showed similar expression in both tissues, however, 55 miRNAs were preferentially expressed in visceral adipose tissue and 53 in skeletal muscle. miR-122b-5p was uniquely identified in skeletal muscle, while miR-1-3p and miR-206 were upregulated in skeletal muscle. Conversely, miR-224-5p and miR-335-3p exhibited upregulation in visceral adipose tissue. Notably, distinctions related to the presence of type 2 diabetes were observed solely in the expression of miR-1-3p and miR-206 in visceral adipose tissue. CONCLUSIONS This is the first study unveiling distinct miRNA expression profiles in paired samples of visceral adipose tissue and skeletal muscle in humans. The identification of obesity-specific miRNAs in these tissues opens up promising avenues for research into potential biomarkers for obesity diagnosis and treatment.
Collapse
Affiliation(s)
- Carmen Lambert
- Health Research Institute of the Principality of Asturias (ISPA), Oviedo, Asturias, Spain.
| | - Paula Morales-Sánchez
- Health Research Institute of the Principality of Asturias (ISPA), Oviedo, Asturias, Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Ana Victoria García
- Health Research Institute of the Principality of Asturias (ISPA), Oviedo, Asturias, Spain
| | - Elsa Villa-Fernández
- Health Research Institute of the Principality of Asturias (ISPA), Oviedo, Asturias, Spain
| | - Jèssica Latorre
- Department of Diabetes Endocrinology and Nutrition (UDEN) Institut d'Investigació Biomèdica de Girona (IDIBGI), Girona, Spain
- Centre for Biomedical Network Research on Obesity and Nutrition Physiopathology (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029, Madrid, Spain
| | - Miguel García-Villarino
- Health Research Institute of the Principality of Asturias (ISPA), Oviedo, Asturias, Spain
- Medicine Department, University of Oviedo, Oviedo, Asturias, Spain
| | - Estrella Olga Turienzo Santos
- Health Research Institute of the Principality of Asturias (ISPA), Oviedo, Asturias, Spain
- Asturias Central University Hospital, Oviedo, Asturias, Spain
| | - Lorena Suárez-Gutierrez
- Health Research Institute of the Principality of Asturias (ISPA), Oviedo, Asturias, Spain
- Asturias Central University Hospital, Oviedo, Asturias, Spain
| | - Raquel Rodríguez Uría
- Health Research Institute of the Principality of Asturias (ISPA), Oviedo, Asturias, Spain
- Asturias Central University Hospital, Oviedo, Asturias, Spain
| | - Sandra Sanz Navarro
- Health Research Institute of the Principality of Asturias (ISPA), Oviedo, Asturias, Spain
- Asturias Central University Hospital, Oviedo, Asturias, Spain
| | - Jessica Ares-Blanco
- Health Research Institute of the Principality of Asturias (ISPA), Oviedo, Asturias, Spain
- Medicine Department, University of Oviedo, Oviedo, Asturias, Spain
- Asturias Central University Hospital, Oviedo, Asturias, Spain
| | - Pedro Pujante
- Health Research Institute of the Principality of Asturias (ISPA), Oviedo, Asturias, Spain
- Asturias Central University Hospital, Oviedo, Asturias, Spain
| | - Lourdes María Sanz Álvarez
- Health Research Institute of the Principality of Asturias (ISPA), Oviedo, Asturias, Spain
- Asturias Central University Hospital, Oviedo, Asturias, Spain
| | - Edelmiro Menéndez-Torre
- Health Research Institute of the Principality of Asturias (ISPA), Oviedo, Asturias, Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Medicine Department, University of Oviedo, Oviedo, Asturias, Spain
- Asturias Central University Hospital, Oviedo, Asturias, Spain
| | - María Moreno Gijón
- Health Research Institute of the Principality of Asturias (ISPA), Oviedo, Asturias, Spain
- Asturias Central University Hospital, Oviedo, Asturias, Spain
| | - José Manuel Fernandez-Real
- Department of Diabetes Endocrinology and Nutrition (UDEN) Institut d'Investigació Biomèdica de Girona (IDIBGI), Girona, Spain
- Centre for Biomedical Network Research on Obesity and Nutrition Physiopathology (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029, Madrid, Spain
| | - Elías Delgado
- Health Research Institute of the Principality of Asturias (ISPA), Oviedo, Asturias, Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Medicine Department, University of Oviedo, Oviedo, Asturias, Spain
- Asturias Central University Hospital, Oviedo, Asturias, Spain
| |
Collapse
|
2
|
Gezginci-Oktayoglu S, Sancar S, Karatug-Kacar A, Bolkent S. Glucotoxicity suppresses function of pancreatic beta and duct cells via miR-335-targeted Runx2 and insulin-mediated mechanism. PROTOPLASMA 2025; 262:341-352. [PMID: 39382633 DOI: 10.1007/s00709-024-01997-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 10/01/2024] [Indexed: 10/10/2024]
Abstract
Pancreatic cell dynamics have important contributions to the development of type 2 diabetes and related diseases such as nonalcoholic fatty pancreas disease. The aim of this study was to investigate the effects of prolonged excessive glucose exposure on the functions of pancreatic beta cells and duct cells in single and co-culture conditions. In this study, we focused on the effects of glucotoxicity on insulin secretion which is the main function of beta cells and on progenitor functions of duct cells. Rat primary INS1 beta cells and ARIP duct cells were exposed to glucose (25 mM) for 72 h under single or indirect co-culture conditions. Glucotoxicity stimuli increased insulin secretion and decreased insulin expression in single beta cells while stimulating beta-cell differentiation and adipogenesis in single duct cells. On the other hand, glucotoxicity caused functional loss and increased proliferation and apoptosis in beta cells while increasing proliferation but suppressed beta-cell differentiation and adipogenesis in duct cells under co-culture conditions. The expression level of miR-335, a microRNA known to be upregulated by leptin and target Runx2, was measured. As a result, unlike single-cell culture, glucotoxicity upregulated miR-335, downregulated Runx2, and decreased insulin signaling in beta cells while downregulating miR-335 and upregulating Runx2, and decreased insulin signaling in duct cells under co-culture conditions. When the results of single and co-culture experiments are compared, insulin and miR-335 may be seen as important mediators for setting up the relation between beta and duct cells. Our findings are important for preventing the development of type 2 diabetes and nonalcoholic fatty pancreas disease, even developing new diagnosis and treatment strategies.
Collapse
Affiliation(s)
- Selda Gezginci-Oktayoglu
- Molecular Biology Section, Biology Department, Faculty of Science, Istanbul University, Vezneciler, 34134, Istanbul, Türkiye.
| | - Serap Sancar
- Molecular Biology Section, Biology Department, Faculty of Science, Istanbul University, Vezneciler, 34134, Istanbul, Türkiye
| | - Ayse Karatug-Kacar
- Molecular Biology Section, Biology Department, Faculty of Science, Istanbul University, Vezneciler, 34134, Istanbul, Türkiye
| | - Sehnaz Bolkent
- Molecular Biology Section, Biology Department, Faculty of Science, Istanbul University, Vezneciler, 34134, Istanbul, Türkiye
| |
Collapse
|
3
|
Ku HC, Kuo CC, Su TC, Yang MJ, Cheng CF, Kao YH. Molecular effects of green tea epigallocatechin gallate on the microRNA-143/MAPK7 and microRNA-let-7a/HMGA2 pathways. TEA IN HEALTH AND DISEASE PREVENTION 2025:571-580. [DOI: 10.1016/b978-0-443-14158-4.00074-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
4
|
Mondal S, Rathor R, Singh SN, Suryakumar G. miRNA and leptin signaling in metabolic diseases and at extreme environments. Pharmacol Res Perspect 2024; 12:e1248. [PMID: 39017237 PMCID: PMC11253706 DOI: 10.1002/prp2.1248] [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: 01/01/2024] [Revised: 05/27/2024] [Accepted: 07/04/2024] [Indexed: 07/18/2024] Open
Abstract
The burden of growing concern about the dysregulation of metabolic processes arises due to complex interplay between environment and nutrition that has great impact on genetics and epigenetics of an individual. Thereby, any abnormality at the level of food intake regulating hormones may contribute to the development of metabolic diseases in any age group due to malnutrition, overweight, changing lifestyle, and exposure to extreme environments such as heat stress (HS), cold stress, or high altitude (HA). Hormones such as leptin, adiponectin, ghrelin, and cholecystokinin regulate appetite and satiety to maintain energy homeostasis. Leptin, an adipokine and a pleiotropic hormone, play major role in regulating the food intake, energy gain and energy expenditure. Using in silico approach, we have identified the major genes (LEP, LEPR, JAK2, STAT3, NPY, POMC, IRS1, SOCS3) that play crucial role in leptin signaling pathway. Further, eight miRNAs (hsa-miR-204-5p, hsa-miR-211-5p, hsa-miR-30, hsa-miR-3163, hsa-miR-33a-3p, hsa-miR-548, hsa-miR-561-3p, hsa-miR-7856-5p) from TargetScan 8.0 database were screened out that commonly target these genes. The role of these miRNAs should be explored as they might play vital role in regulating the appetite, energy metabolism, metabolic diseases (obesity, type 2 diabetes, cardiovascular diseases, inflammation), and to combat extreme environments. The miRNAs regulating leptin signaling and appetite may be useful for developing novel therapeutics for metabolic diseases.
Collapse
Affiliation(s)
- Samrita Mondal
- Defence Institute of Physiology and Allied SciencesDelhiIndia
| | - Richa Rathor
- Defence Institute of Physiology and Allied SciencesDelhiIndia
| | - Som Nath Singh
- Defence Institute of Physiology and Allied SciencesDelhiIndia
| | | |
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
|
Liu C, Liu X, Li H, Kang Z. Advances in the regulation of adipogenesis and lipid metabolism by exosomal ncRNAs and their role in related metabolic diseases. Front Cell Dev Biol 2023; 11:1173904. [PMID: 37791070 PMCID: PMC10543472 DOI: 10.3389/fcell.2023.1173904] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 08/15/2023] [Indexed: 10/05/2023] Open
Abstract
Exosomes are membrane-bound extracellular vesicles released following the fusion of multivesicular bodies (MVBs) with the cell membrane. Exosomes transport diverse molecules, including proteins, lipids, DNA and RNA, and regulate distant intercellular communication. Noncoding RNA (ncRNAs) carried by exosomes regulate cell-cell communication in tissues, including adipose tissue. This review summarizes the action mechanisms of ncRNAs carried by exosomes on adipocyte differentiation and modulation of adipogenesis by exosomal ncRNAs. This study aims to provide valuable insights for developing novel therapeutics.
Collapse
Affiliation(s)
- Cong Liu
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xilin Liu
- Department of Hand and Foot Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Hong Li
- Department of Nursing, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Zhichen Kang
- Department of Rehabilitation, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
7
|
Afsharmanesh MR, Mohammadi Z, Mansourian AR, Jafari SM. A Review of micro RNAs changes in T2DM in animals and humans. J Diabetes 2023; 15:649-664. [PMID: 37329278 PMCID: PMC10415875 DOI: 10.1111/1753-0407.13431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 04/22/2023] [Accepted: 05/24/2023] [Indexed: 06/19/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) and its associated complications have become a crucial public health concern in the world. According to the literature, chronic inflammation and the progression of T2DM have a close relationship. Accumulated evidence suggests that inflammation enhances the insulin secretion lost by islets of Langerhans and the resistance of target tissues to insulin action, which are two critical features in T2DM development. Based on recently highlighted research that plasma concentration of inflammatory mediators such as tumor necrosis factor α and interleukin-6 are elevated in insulin-resistant and T2DM, and it raises novel question marks about the processes causing inflammation in both situations. Over the past few decades, microRNAs (miRNAs), a class of short, noncoding RNA molecules, have been discovered to be involved in the regulation of inflammation, insulin resistance, and T2DM pathology. These noncoding RNAs are specifically comprised of RNA-induced silencing complexes and regulate the expression of specific protein-coding genes through various mechanisms. There is extending evidence that describes the expression profile of a special class of miRNA molecules altered during T2DM development. These modifications can be observed as potential biomarkers for the diagnosis of T2DM and related diseases. In this review study, after reviewing the possible mechanisms involved in T2DM pathophysiology, we update recent information on the miRNA roles in T2DM, inflammation, and insulin resistance.
Collapse
Affiliation(s)
- Mohammad Reza Afsharmanesh
- Metabolic Disorders Research CenterGolestan University of Medical SciencesGorganIran
- Department of Biochemistry and Biophysics, School of MedicineGolestan University of Medical SciencesGorganIran
| | - Zeinab Mohammadi
- Metabolic Disorders Research CenterGolestan University of Medical SciencesGorganIran
- Department of Biochemistry and Biophysics, School of MedicineGolestan University of Medical SciencesGorganIran
| | - Azad Reza Mansourian
- Metabolic Disorders Research CenterGolestan University of Medical SciencesGorganIran
- Department of Biochemistry and Biophysics, School of MedicineGolestan University of Medical SciencesGorganIran
| | - Seyyed Mehdi Jafari
- Metabolic Disorders Research CenterGolestan University of Medical SciencesGorganIran
- Department of Biochemistry and Biophysics, School of MedicineGolestan University of Medical SciencesGorganIran
| |
Collapse
|
8
|
Macvanin MT, Gluvic Z, Bajic V, Isenovic ER. Novel insights regarding the role of noncoding RNAs in diabetes. World J Diabetes 2023; 14:958-976. [PMID: 37547582 PMCID: PMC10401459 DOI: 10.4239/wjd.v14.i7.958] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 05/01/2023] [Accepted: 05/22/2023] [Indexed: 07/12/2023] Open
Abstract
Diabetes mellitus (DM) is a group of metabolic disorders defined by hyperglycemia induced by insulin resistance, inadequate insulin secretion, or excessive glucagon secretion. In 2021, the global prevalence of diabetes is anticipated to be 10.7% (537 million people). Noncoding RNAs (ncRNAs) appear to have an important role in the initiation and progression of DM, according to a growing body of research. The two major groups of ncRNAs implicated in diabetic disorders are miRNAs and long noncoding RNAs. miRNAs are single-stranded, short (17-25 nucleotides), ncRNAs that influence gene expression at the post-transcriptional level. Because DM has reached epidemic proportions worldwide, it appears that novel diagnostic and therapeutic strategies are required to identify and treat complications associated with these diseases efficiently. miRNAs are gaining attention as biomarkers for DM diagnosis and potential treatment due to their function in maintaining physiological homeostasis via gene expression regulation. In this review, we address the issue of the gradually expanding global prevalence of DM by presenting a complete and up-to-date synopsis of various regulatory miRNAs involved in these disorders. We hope this review will spark discussion about ncRNAs as prognostic biomarkers and therapeutic tools for DM. We examine and synthesize recent research that used novel, high-throughput technologies to uncover ncRNAs involved in DM, necessitating a systematic approach to examining and summarizing their roles and possible diagnostic and therapeutic uses.
Collapse
Affiliation(s)
- Mirjana T Macvanin
- Department of Radiobiology and Molecular Genetics, Vinča Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade 11000, Serbia
| | - Zoran Gluvic
- Department of Endocrinology and Diabetes, Clinic for Internal Medicine, Zemun Clinical Hospital, School of Medicine, University of Belgrade, Belgrade 11000, Serbia
| | - Vladan Bajic
- Department of Radiobiology and Molecular Genetics, Vinča Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade 11000, Serbia
| | - Esma R Isenovic
- Department of Radiobiology and Molecular Genetics, Vinča Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade 11000, Serbia
| |
Collapse
|
9
|
Kumar P, Courtes M, Lemmers C, Le Digarcher A, Coku I, Monteil A, Hong C, Varrault A, Liu R, Wang L, Bouschet T. Functional mapping of microRNA promoters with dCas9 fused to transcriptional regulators. Front Genet 2023; 14:1147222. [PMID: 37214422 PMCID: PMC10196145 DOI: 10.3389/fgene.2023.1147222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 04/18/2023] [Indexed: 05/24/2023] Open
Abstract
MicroRNAs are small non-coding RNAs that control gene expression during development, physiology, and disease. Transcription is a key factor in microRNA abundance and tissue-specific expression. Many databases predict the location of microRNA transcription start sites and promoters. However, these candidate regions require functional validation. Here, dCas9 fused to transcriptional activators or repressors - CRISPR activation (CRISPRa) and inhibition (CRISPRi)- were targeted to the candidate promoters of two intronic microRNAs, mmu-miR-335 and hsa-miR-3662, including the promoters of their respective host genes Mest and HBS1L. We report that in mouse embryonic stem cells and brain organoids, miR-335 was downregulated upon CRISPRi of its host gene Mest. Reciprocally, CRISPRa of Mest promoter upregulated miR-335. By contrast, CRISPRa of the predicted miR-335-specific promoter (located in an intron of Mest) did not affect miR-335 levels. Thus, the expression of miR-335 only depends on the promoter activity of its host gene Mest. By contrast, miR-3662 was CRISPR activatable both by the promoter of its host gene HBS1L and an intronic sequence in HEK-293T cells. Thus, CRISPRa and CRISPRi are powerful tools to evaluate the relevance of endogenous regulatory sequences involved in microRNA transcription in defined cell types.
Collapse
Affiliation(s)
- Pradeep Kumar
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, United States
- O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Mathilde Courtes
- Institut de Génomique Fonctionnelle, CNRS, INSERM, Université de Montpellier, Montpellier, France
| | - Céline Lemmers
- Plateforme de Vectorologie de Montpellier (PVM), BioCampus Montpellier, CNRS, INSERM, Université de Montpellier, Montpellier, France
| | - Anne Le Digarcher
- Institut de Génomique Fonctionnelle, CNRS, INSERM, Université de Montpellier, Montpellier, France
| | - Ilda Coku
- Institut de Génomique Fonctionnelle, CNRS, INSERM, Université de Montpellier, Montpellier, France
| | - Arnaud Monteil
- Plateforme de Vectorologie de Montpellier (PVM), BioCampus Montpellier, CNRS, INSERM, Université de Montpellier, Montpellier, France
| | - Charles Hong
- Vanderbilt University School of Medicine Nashville, Nashville, TN, United States
| | - Annie Varrault
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Runhua Liu
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, United States
- O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Lizhong Wang
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, United States
- O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Tristan Bouschet
- Institut de Génomique Fonctionnelle, CNRS, INSERM, Université de Montpellier, Montpellier, France
| |
Collapse
|
10
|
MacCuaig WM, Thomas A, Carlos-Sorto JC, Gomez-Gutierrez JG, Alexander AC, Wellberg EA, Grizzle WE, McNally LR. Differential expression of microRNA between triple negative breast cancer patients of African American and European American descent. Biotech Histochem 2022; 97:1-10. [PMID: 34979848 PMCID: PMC9047185 DOI: 10.1080/10520295.2021.2005147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
There are racial disparities in the outcome of triple negative breast cancer (TNBC) patients between women of African ancestry and women of European ancestry, even after accounting for lifestyle, socioeconomic and clinical factors. MicroRNA (miRNA) are non-coding molecules whose level of expression is associated with cancer suppression, proliferation and drug resistance; therefore, these have potential for biomarker applications in cancers including TNBC. Historically, miRNAs up-regulated in African American (AA) patients have received less attention than for patients of European ancestry. Using laser capture microdissection (LCM) to acquire ultrapure tumor cell samples, miRNA expression was evaluated in 15 AA and 15 European American (EA) TNBC patients. Tumor sections were evaluated using RNA extraction followed by miRNA analysis and profiling. Results were compared based on ethnicity and method of tissue fixation. miRNAs that showed high differential expression in AA TNBC patients compared to EA included: miR-19a, miR-192, miR-302a, miR-302b, miR-302c, miR-335, miR-520b, miR-520f and miR-645. LCM is a useful technique for isolation of tumor cells. We found a greater abundance of RNA in frozen samples compared to formalin fixed, paraffin embedded samples. miRNA appears to be a useful biomarker for TNBC to improve diagnosis and treatment.
Collapse
Affiliation(s)
- William M. MacCuaig
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, Oklahoma,Department of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma
| | - Alexandra Thomas
- Department of Hematology Oncology, Wake Forest Baptist Health, Winston-Salem, North Carolina
| | - Juan C. Carlos-Sorto
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, Oklahoma,Department of Surgery, University of Oklahoma, Oklahoma City, Oklahoma
| | | | - Adam C. Alexander
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, Oklahoma,Department of Family and Preventive Medicine, University of Oklahoma, Oklahoma City, Oklahoma
| | - Elizabeth A. Wellberg
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, Oklahoma,Department of Pathology, University of Oklahoma, Oklahoma City, Oklahoma
| | - William E. Grizzle
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Lacey R. McNally
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, Oklahoma,Department of Surgery, University of Oklahoma, Oklahoma City, Oklahoma
| |
Collapse
|
11
|
microRNAs in Human Adipose Tissue Physiology and Dysfunction. Cells 2021; 10:cells10123342. [PMID: 34943849 PMCID: PMC8699244 DOI: 10.3390/cells10123342] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/20/2021] [Accepted: 11/26/2021] [Indexed: 12/11/2022] Open
Abstract
In recent years, there has been a large amount of evidence on the role of microRNA (miRNA) in regulating adipose tissue physiology. Indeed, miRNAs control critical steps in adipocyte differentiation, proliferation and browning, as well as lipolysis, lipogenesis and adipokine secretion. Overnutrition leads to a significant change in the adipocyte miRNOME, resulting in adipose tissue dysfunction. Moreover, via secreted mediators, dysfunctional adipocytes may impair the function of other organs and tissues. However, given their potential to control cell and whole-body energy expenditure, miRNAs also represent critical therapeutic targets for treating obesity and related metabolic complications. This review attempts to integrate present concepts on the role miRNAs play in adipose tissue physiology and obesity-related dysfunction and data from pre-clinical and clinical studies on the diagnostic or therapeutic potential of miRNA in obesity and its related complications.
Collapse
|
12
|
Yusof KM, Groen K, Rosli R, Avery-Kiejda KA. Crosstalk Between microRNAs and the Pathological Features of Secondary Lymphedema. Front Cell Dev Biol 2021; 9:732415. [PMID: 34733847 PMCID: PMC8558478 DOI: 10.3389/fcell.2021.732415] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 10/01/2021] [Indexed: 01/07/2023] Open
Abstract
Secondary lymphedema is characterized by lymphatic fluid retention and subsequent tissue swelling in one or both limbs that can lead to decreased quality of life. It often arises after loss, obstruction, or blockage of lymphatic vessels due to multifactorial modalities, such as lymphatic insults after surgery, immune system dysfunction, deposition of fat that compresses the lymphatic capillaries, fibrosis, and inflammation. Although secondary lymphedema is often associated with breast cancer, the condition can occur in patients with any type of cancer that requires lymphadenectomy such as gynecological, genitourinary, or head and neck cancers. MicroRNAs demonstrate pivotal roles in regulating gene expression in biological processes such as lymphangiogenesis, angiogenesis, modulation of the immune system, and oxidative stress. MicroRNA profiling has led to the discovery of the molecular mechanisms involved in the pathophysiology of auto-immune, inflammation-related, and metabolic diseases. Although the role of microRNAs in regulating secondary lymphedema is yet to be elucidated, the crosstalk between microRNAs and molecular factors involved in the pathological features of lymphedema, such as skin fibrosis, inflammation, immune dysregulation, and aberrant lipid metabolism have been demonstrated in several studies. MicroRNAs have the potential to serve as biomarkers for diseases and elucidation of their roles in lymphedema can provide a better understanding or new insights of the mechanisms underlying this debilitating condition.
Collapse
Affiliation(s)
- Khairunnisa’ Md Yusof
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, The University of Newcastle, Newcastle, NSW, Australia
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Kira Groen
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, The University of Newcastle, Newcastle, NSW, Australia
| | - Rozita Rosli
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
- UPM-MAKNA Cancer Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang, Malaysia
| | - Kelly A. Avery-Kiejda
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, The University of Newcastle, Newcastle, NSW, Australia
| |
Collapse
|
13
|
Yang H, Zhou J, Wang J, Zhang L, Liu Q, Luo J, Jia H, Liu L, Zhou Q. Circulating Exosomal MicroRNA Profiles Associated with Acute Soft Tissue Injury. CELL JOURNAL 2021; 23:474-484. [PMID: 34455724 PMCID: PMC8405084 DOI: 10.22074/cellj.2021.7275] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 04/04/2020] [Indexed: 12/16/2022]
Abstract
Objective This study aimed to characterize the circulating exosomal microRNA (miRNA) profiles associated with acute soft tissue injury. Materials and Methods In this experimental study, a total of 12 rats were randomly divided into control group and model group (n=6 for each group). The rats in the model group were used to establish an acute soft tissue injury following the mechanical injury of the leg. The exosomes from the peripheral blood of all the rats were isolated and then characterized by Nanosight NS300 particle size analyser (NTA), transmission electron microscopy (TEM) and western blot. Next, the exosomal miRNAs in the control and model groups were sequenced, and the differentially expressed miRNAs (DE-miRNAs) were identified using the DESeq algorithm. Functional analyses were performed using Gene Ontology (GO) terms and the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway databases. Finally, quantitative reverse-transcription polymersa chain reaction (qRT-PCR) was used to verify the expression of the DE-miRNAs. Results TEM, NTA and western blot results showed that the exosomes were approximately 100 nm in size and exhibited cup-shaped morphology. A total of 628 miRNAs were obtained by sequencing. After that, 28 DE miRNAs (DEmiRNAs) were identified, including seven down-regulated miRNAs and 21 up-regulated miRNAs. These DE-miRNAs were linked to 7539 target genes with GO. Also, KEGG analyses demonstrated that these genes were enriched for phosphorylation, VEGF signaling pathway, and MAPK signaling pathway. Additionally, the consistency rate between the qRT-PCR and sequencing results was 83.33%, which showed a high relative reliability of the sequencing results. Conclusion These findings suggest that these 28 exosomal miRNAs may be involved in the regulation of acute soft tissue injury, by one of critical biological processes (BP), phosphorylation. The findings provide valuable clues by utilizing exosomes as therapeutic targets for the effective treatment of acute soft tissue injury.
Collapse
Affiliation(s)
- Hongchang Yang
- Physical Education Department, Hohai University, Nanjing, Jiangsu, China
| | - Jing Zhou
- Department of Clinical Medicine, Jiangsu Health Vocational College, Nanjing, Jiangsu, China
| | - Junlei Wang
- Harbor, Channel and Coastal Engineering, Hohai University, Nanjing, Jiangsu, China
| | - Luoning Zhang
- Physical Education Department, Hohai University, Nanjing, Jiangsu, China
| | - Quzhi Liu
- Centre of Counseling and Psychological Services, Hohai University, Nanjing, Jiangsu, China
| | - Jing Luo
- Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hongyan Jia
- Port Channel and Coastal Engineering Department, Hohai University, Nanjing, Jiangsu, China
| | - Li Liu
- The Department of Rehabilitation, Brain Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Qiang Zhou
- Physical Education Department, Hohai University, Nanjing, Jiangsu, China.
| |
Collapse
|
14
|
Hanousková B, Vávrová G, Ambrož M, Boušová I, Karlsen TA, Skálová L, Matoušková P. MicroRNAs mediated regulation of glutathione peroxidase 7 expression and its changes during adipogenesis. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2021; 1864:194734. [PMID: 34339889 DOI: 10.1016/j.bbagrm.2021.194734] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 07/12/2021] [Accepted: 07/22/2021] [Indexed: 12/19/2022]
Abstract
Glutathione peroxidase 7 (GPx7) acts as an intracellular stress sensor/transmitter and plays an important role in adipocyte differentiation and the prevention of obesity related pathologies. For this reason, finding the regulatory mechanisms that control GPx7 expression is of great importance. As microRNAs (miRNAs) could participate in the regulation of GPx7 expression, we studied the inhibition of GPx7 expression by four selected miRNAs with relation to obesity and adipogenesis. The effect of the transfection of selected miRNAs mimics on GPx7 expression was tested in three cell models (HEK293, SW480, AT-MSC). The interaction of selected miRNAs with the 3'UTR of GPx7 was followed up on using a luciferase gene reporter assay. In addition, the levels of GPx7 and selected miRNAs in adipose tissue mesenchymal stem cells (AT-MSC) and mature adipocytes from four human donors were compared, with the changes in these levels during adipogenesis analyzed. Our results show for the first time that miR-137 and miR-29b bind to the 3'UTR region of GPx7 and inhibit the expression of this enzyme at the mRNA and protein level in all the human cells tested. However, no negative correlation between miR-137 nor miR-29b level and GPx7 was observed during adipogenesis. Despite the confirmed inhibition of GPx7 expression by miR-137 and miR-29b, the action of these two molecules in adipogenesis and mature adipocytes must be accompanied by other regulators.
Collapse
Affiliation(s)
- Barbora Hanousková
- Faculty of Pharmacy in Hradec Králové, Department of Biochemical Sciences, Charles University, Hradec Králové, Czech Republic; Norwegian Center for Stem Cell Research, Department of Immunology and Transfusion Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Gabriela Vávrová
- Faculty of Pharmacy in Hradec Králové, Department of Biochemical Sciences, Charles University, Hradec Králové, Czech Republic
| | - Martin Ambrož
- Faculty of Pharmacy in Hradec Králové, Department of Biochemical Sciences, Charles University, Hradec Králové, Czech Republic
| | - Iva Boušová
- Faculty of Pharmacy in Hradec Králové, Department of Biochemical Sciences, Charles University, Hradec Králové, Czech Republic
| | - Tommy A Karlsen
- Norwegian Center for Stem Cell Research, Department of Immunology and Transfusion Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Lenka Skálová
- Faculty of Pharmacy in Hradec Králové, Department of Biochemical Sciences, Charles University, Hradec Králové, Czech Republic
| | - Petra Matoušková
- Faculty of Pharmacy in Hradec Králové, Department of Biochemical Sciences, Charles University, Hradec Králové, Czech Republic.
| |
Collapse
|
15
|
Miura A, Ikeda A, Abe M, Seo K, Watanabe T, Ozaki-Masuzawa Y, Hosono T, Seki T. Diallyl Trisulfide Prevents Obesity and Decreases miRNA-335 Expression in Adipose Tissue in a Diet-Induced Obesity Rat Model. Mol Nutr Food Res 2021; 65:e2001199. [PMID: 34014027 DOI: 10.1002/mnfr.202001199] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 04/19/2021] [Indexed: 01/03/2023]
Abstract
SCOPE Diallyl trisulfide (DATS), an organosulfur compound generates in crushed garlic, has various beneficial health effects. A growing body of evidence indicates that miRNAs are involved in the pathology of lifestyle diseases including obesity. The anti-obesogenic effect of garlic is previously reported; however, the effects of DATS on obesity, and the relationship between garlic compounds and the involvement of miRNA remains unclear. Here, the anti-obesogenic activity of DATS and the potential role of miRNA in a diet-induced obesity rat model are investigated. METHODS AND RESULTS Oral administration of DATS suppressed body and white adipose tissue (WAT) weight gain in rats fed a high-fat diet compared with vehicle-administered rats. DATS lowered the plasma and liver triglyceride levels in obese rats, and decreased lipogenic mRNA levels including those of Srebp1c, Fasn, and Scd1 in the liver. DATS also suppressed de novo lipogenesis in the liver. Transcriptomic analyses of miRNA and mRNA in the epididymal WAT of obese rats using microarrays revealed that DATS decreased miRNA-335 expression and normalized the obesity-related mRNA transcriptomic signatures in epididymal WAT. CONCLUSION The potent anti-obesogenic effects of DATS and its possible mechanism of action was clearly demonstrated in this study.
Collapse
Affiliation(s)
- Atsushi Miura
- General Research Institute, Nihon University College of Bioresource Sciences, Fujisawa, Kanagawa, 252-0880, Japan
| | - Ayana Ikeda
- Department of Applied Life Sciences, Nihon University Graduate School of Bioresource Sciences, Fujisawa, Kanagawa, 252-0880, Japan
| | - Marina Abe
- Department of Applied Life Sciences, Nihon University Graduate School of Bioresource Sciences, Fujisawa, Kanagawa, 252-0880, Japan
| | - Kiki Seo
- Department of Applied Life Sciences, Nihon University Graduate School of Bioresource Sciences, Fujisawa, Kanagawa, 252-0880, Japan
| | - Takahiro Watanabe
- Department of Applied Life Sciences, Nihon University Graduate School of Bioresource Sciences, Fujisawa, Kanagawa, 252-0880, Japan
| | - Yori Ozaki-Masuzawa
- Department of Chemistry and Life Science, Nihon University College of Bioresource Sciences, Fujisawa, Kanagawa, 252-0880, Japan
| | - Takashi Hosono
- Department of Applied Life Sciences, Nihon University Graduate School of Bioresource Sciences, Fujisawa, Kanagawa, 252-0880, Japan
- Department of Chemistry and Life Science, Nihon University College of Bioresource Sciences, Fujisawa, Kanagawa, 252-0880, Japan
| | - Taiichiro Seki
- General Research Institute, Nihon University College of Bioresource Sciences, Fujisawa, Kanagawa, 252-0880, Japan
- Department of Applied Life Sciences, Nihon University Graduate School of Bioresource Sciences, Fujisawa, Kanagawa, 252-0880, Japan
- Department of Chemistry and Life Science, Nihon University College of Bioresource Sciences, Fujisawa, Kanagawa, 252-0880, Japan
| |
Collapse
|
16
|
Braga AA, Bortolin RH, Graciano-Saldarriaga ME, Hirata TD, Cerda A, de Freitas RC, Lin-Wang HT, Borges JB, França JI, Masi LN, Curi R, Pithon-Curi TC, Sampaio MF, Castro LR, Bastos GM, Hirata RD, Hirata MH. High serum miR-421 is associated with metabolic dysregulation and inflammation in patients with metabolic syndrome. Epigenomics 2021; 13:423-436. [PMID: 33678000 DOI: 10.2217/epi-2020-0247] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Aim: To explore the association of circulating miRNAs with adiposity, metabolic status and inflammatory biomarkers in patients with metabolic syndrome (MetS). Methods: Serum levels of 372 miRNAs were measured in patients with (n = 6) and without MetS (n = 6) by quantitative PCR array, and dysregulated miRNAs were validated in a larger cohort (MetS, n = 89; non-MetS, n = 144). Results: In the screening study, seven miRNAs were dysregulated in patients with MetS, and miR-421 remained increased in the validation study. miR-421 was associated with a high risk of MetS and insulin resistance and hypertension and correlated with glycated hemoglobin, triacylglycerols, high-sensitivity CRP, IL-6, resistin and adiponectin (p < 0.05). Conclusion: Circulating miR-421 is a potential biomarker for insulin resistance, metabolic dysregulation and inflammatory status in patients with MetS.
Collapse
Affiliation(s)
- Aécio A Braga
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Raul H Bortolin
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Magda E Graciano-Saldarriaga
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Thiago Dc Hirata
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Alvaro Cerda
- Department of Basic Sciences, Center of Excellence in Translational Medicine, CEMT-BIOREN, Universidad de La Frontera, Temuco 4810296, Chile
| | - Renata Cc de Freitas
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Hui T Lin-Wang
- Molecular Research Laboratory in Cardiology, Institute Dante Pazzanese of Cardiology, Sao Paulo 04012-909, Brazil
| | - Jessica B Borges
- Molecular Research Laboratory in Cardiology, Institute Dante Pazzanese of Cardiology, Sao Paulo 04012-909, Brazil
| | - João Id França
- Laboratory of Epidemiology and Statistics, Institute Dante Pazzanese of Cardiology, Sao Paulo 04012-909, Brazil
| | - Laureane N Masi
- Interdisciplinary Post-graduate Program in Health Sciences, Cruzeirodo Sul University, Sao Paulo 01506-000, Brazil
| | - Rui Curi
- Interdisciplinary Post-graduate Program in Health Sciences, Cruzeirodo Sul University, Sao Paulo 01506-000, Brazil
| | - Tania C Pithon-Curi
- Interdisciplinary Post-graduate Program in Health Sciences, Cruzeirodo Sul University, Sao Paulo 01506-000, Brazil
| | - Marcelo F Sampaio
- Medical Clinic Division, Institute Dante Pazzanese of Cardiology, Sao Paulo 04012-909, Brazil.,Department of Cardiology, Real e Benemerita Associação Portuguesa de Beneficiência, Sao Paulo 01323-001, Brazil
| | - Lara R Castro
- Medical Clinic Division, Institute Dante Pazzanese of Cardiology, Sao Paulo 04012-909, Brazil.,Department of Cardiology, Real e Benemerita Associação Portuguesa de Beneficiência, Sao Paulo 01323-001, Brazil
| | - Gisele M Bastos
- Molecular Research Laboratory in Cardiology, Institute Dante Pazzanese of Cardiology, Sao Paulo 04012-909, Brazil.,Department of Teaching and Research, Real e Benemerita Associação Portuguesa de Beneficiencia, Sao Paulo 01323-001, Brazil
| | - Rosario Dc Hirata
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Mario H Hirata
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| |
Collapse
|
17
|
Zhang Z, Chen L, Xu P, Xing L, Hong Y, Chen P. Gene correlation network analysis to identify regulatory factors in sepsis. J Transl Med 2020; 18:381. [PMID: 33032623 PMCID: PMC7545567 DOI: 10.1186/s12967-020-02561-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 10/03/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Sepsis is a leading cause of mortality and morbidity in the intensive care unit. Regulatory mechanisms underlying the disease progression and prognosis are largely unknown. The study aimed to identify master regulators of mortality-related modules, providing potential therapeutic target for further translational experiments. METHODS The dataset GSE65682 from the Gene Expression Omnibus (GEO) database was utilized for bioinformatic analysis. Consensus weighted gene co-expression netwoek analysis (WGCNA) was performed to identify modules of sepsis. The module most significantly associated with mortality were further analyzed for the identification of master regulators of transcription factors and miRNA. RESULTS A total number of 682 subjects with various causes of sepsis were included for consensus WGCNA analysis, which identified 27 modules. The network was well preserved among different causes of sepsis. Two modules designated as black and light yellow module were found to be associated with mortality outcome. Key regulators of the black and light yellow modules were the transcription factor CEBPB (normalized enrichment score = 5.53) and ETV6 (NES = 6), respectively. The top 5 miRNA regulated the most number of genes were hsa-miR-335-5p (n = 59), hsa-miR-26b-5p (n = 57), hsa-miR-16-5p (n = 44), hsa-miR-17-5p (n = 42), and hsa-miR-124-3p (n = 38). Clustering analysis in 2-dimension space derived from manifold learning identified two subclasses of sepsis, which showed significant association with survival in Cox proportional hazard model (p = 0.018). CONCLUSIONS The present study showed that the black and light-yellow modules were significantly associated with mortality outcome. Master regulators of the module included transcription factor CEBPB and ETV6. miRNA-target interactions identified significantly enriched miRNA.
Collapse
Affiliation(s)
- Zhongheng Zhang
- Department of Emergency Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No 3, East Qingchun Road, Hangzhou, 310016 Zhejiang Province China
| | - Lin Chen
- Department of Critical Care Medicine, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Ping Xu
- Emergency Department, Zigong Fourth People’s Hospital, 19 Tanmulin Road, Zigong, Sichuan China
| | - Lifeng Xing
- Department of Emergency Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No 3, East Qingchun Road, Hangzhou, 310016 Zhejiang Province China
| | - Yucai Hong
- Department of Emergency Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No 3, East Qingchun Road, Hangzhou, 310016 Zhejiang Province China
| | - Pengpeng Chen
- Department of Emergency Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No 3, East Qingchun Road, Hangzhou, 310016 Zhejiang Province China
| |
Collapse
|
18
|
Szczepankiewicz D, Langwiński W, Kołodziejski P, Pruszyńska-Oszmałek E, Sassek M, Nowakowska J, Chmurzyńska A, Nowak KW, Szczepankiewicz A. Allergic Inflammation Alters microRNA Expression Profile in Adipose Tissue in the Rat. Genes (Basel) 2020; 11:genes11091034. [PMID: 32887419 PMCID: PMC7564923 DOI: 10.3390/genes11091034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/29/2020] [Accepted: 08/31/2020] [Indexed: 01/28/2023] Open
Abstract
Adipose tissue is a major source of circulating exosomal microRNAs (miRNAs) that are modulators of the immune response in various types of tissues and organs, including airways. Still, no evidence exists if allergic airway inflammation may affect fat tissue inflammation via alterations in the miRNA expression profile. Therefore, we investigated the miRNA expression profile in the adipose tissue upon induced allergic inflammation in the airways in the rat. Brown Norway rats were chronically sensitized to house dust mite extract for seven weeks. Body composition was performed using MiniSpec Plus. The eosinophil count and the total IgE level were determined to confirm the induction of allergic inflammation. MiRNA expression profiling was done using the next-generation sequencing with validation by qPCR. We found that allergic airway inflammation significantly increased fat in adipose tissue, glucose concentration, and the gene expression of adipose tissue-derived proinflammatory peptides (leptin, TNFα). In miRNA-seq analysis, we showed significant differences in the expression of 36 mature miRNAs, three precursors, and two miRNA families in adipose tissue of allergic rats. Two miRNAs—miRNA-151-5p and miRNA-423-3p—showed significantly increased expression in qPCR in adipose tissue and lungs of sensitized animals. Allergic airway inflammation affects fat tissue and alters miRNA expression profile in adipose tissue in the rat.
Collapse
Affiliation(s)
- Dawid Szczepankiewicz
- Department of Animal Physiology, Biochemistry and Biostructure, Poznań University of Life Sciences, 60-637 Poznań, Poland; (D.S.); (P.K.); (E.P.-O.); (M.S.); (K.W.N.)
| | - Wojciech Langwiński
- Molecular and Cell Biology Unit, Department of Pediatric Pulmonology, Allergy and Clinical Immunology, Poznan University of Medical Sciences, 60-572 Poznań, Poland; (W.L.); (J.N.)
| | - Paweł Kołodziejski
- Department of Animal Physiology, Biochemistry and Biostructure, Poznań University of Life Sciences, 60-637 Poznań, Poland; (D.S.); (P.K.); (E.P.-O.); (M.S.); (K.W.N.)
| | - Ewa Pruszyńska-Oszmałek
- Department of Animal Physiology, Biochemistry and Biostructure, Poznań University of Life Sciences, 60-637 Poznań, Poland; (D.S.); (P.K.); (E.P.-O.); (M.S.); (K.W.N.)
| | - Maciej Sassek
- Department of Animal Physiology, Biochemistry and Biostructure, Poznań University of Life Sciences, 60-637 Poznań, Poland; (D.S.); (P.K.); (E.P.-O.); (M.S.); (K.W.N.)
| | - Joanna Nowakowska
- Molecular and Cell Biology Unit, Department of Pediatric Pulmonology, Allergy and Clinical Immunology, Poznan University of Medical Sciences, 60-572 Poznań, Poland; (W.L.); (J.N.)
| | - Agata Chmurzyńska
- Institute of Human Nutrition and Dietetics, Poznań University of Life Sciences, 60-624 Poznań, Poland;
| | - Krzysztof W. Nowak
- Department of Animal Physiology, Biochemistry and Biostructure, Poznań University of Life Sciences, 60-637 Poznań, Poland; (D.S.); (P.K.); (E.P.-O.); (M.S.); (K.W.N.)
| | - Aleksandra Szczepankiewicz
- Molecular and Cell Biology Unit, Department of Pediatric Pulmonology, Allergy and Clinical Immunology, Poznan University of Medical Sciences, 60-572 Poznań, Poland; (W.L.); (J.N.)
- Correspondence: ; Tel.: +48-61-88547643; Fax: +48-618547663
| |
Collapse
|
19
|
Epigenetic modulation of tenascin C in the heart: implications on myocardial ischemia, hypertrophy and metabolism. J Hypertens 2020; 37:1861-1870. [PMID: 30950975 DOI: 10.1097/hjh.0000000000002097] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
BACKGROUND Tenascin C (TN-C) is considered to play a pathophysiological role in maladaptive left ventricular remodeling. Yet, the mechanism underlying TN-C-dependent cardiac dysfunction remains elusive. METHOD The present study was designed to investigate the effect of hypoxia and hypertrophic stimuli on TN-C expression in H9c2 cells and its putative regulation by epigenetic mechanisms, namely DNA promoter methylation and microRNAs. In addition, rats subjected to myocardial infarction (MI) were investigated. H9c2 cells were subjected to oxygen and glucose deprivation; incubated with angiotensin II (Ang II); or human TN-C (hTN-C) purified protein. Hypertrophic and fibrotic markers, TN-C promoter methylation as well as mir-335 expression were assessed by reverse transcription and quantitative polymerase chain reaction while TN-C protein levels were assessed by ELISA. RESULTS Tn-C mRNA expression was markedly increased by both oxygen and glucose deprivation and Ang II (P < 0.01, respectively). In addition, Ang-II-dependent TN-C upregulation was explained by reduced promoter methylation (P < 0.05). Cells treated with hTN-C displayed upregulation of Bnp, Mmp2, β-Mhc, integrin α6 and integrin β1. Furthermore, hTN-C treated cells showed a significant reduction in adenosine monophosphate and adenosine triphosphate levels. In vivo, plasma and myocardial TN-C levels were increased 7 days post MI (P < 0.05, respectively). This increment in TN-C was accompanied by upregulation of mir-335 (P < 0.01). In conclusion, both hypoxic and hypertrophic stimuli lead to epigenetically driven TN-C upregulation and subsequent impairment of cellular energy metabolism in cardiomyoblasts. CONCLUSION These findings might enlighten our understanding on maladaptive left ventricular remodeling and direct towards a strong involvement of TN-C.
Collapse
|
20
|
Mir BA, Reyer H, Komolka K, Ponsuksili S, Kühn C, Maak S. Differentially Expressed miRNA-Gene Targets Related to Intramuscular Fat in Musculus Longissimus Dorsi of Charolais × Holstein F 2-Crossbred Bulls. Genes (Basel) 2020; 11:genes11060700. [PMID: 32630492 PMCID: PMC7348786 DOI: 10.3390/genes11060700] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/22/2020] [Accepted: 06/23/2020] [Indexed: 12/12/2022] Open
Abstract
Intramuscular fat (IMF) is a meat quality indicator associated with taste and juiciness. IMF deposition, influenced by genetic and non-genetic factors, occurs through a transcriptionally coordinated process of adipogenesis. MicroRNAs (miRNAs) are transcriptional regulators of vital biological processes, including lipid metabolism and adipogenesis. However, in bovines, limited data on miRNA profiling and association with divergent intramuscular fat content, regulated exclusively by genetic parameters, have been reported. Here, a microarray experiment was performed to identify and characterize the miRNA expression pattern in the Musculus longissimus dorsi of F2-cross (Charolais × German Holstein) bulls with high and low IMF. A total of 38 differentially expressed miRNAs (DE miRNAs), including 33 upregulated and 5 downregulated (corrected p-value ≤ 0.05, FC ≥ ±1.2), were reported. Among DE miRNAs, the upregulated miRNAs miR-105a/b, miR-695, miR-1193, miR-1284, miR-1287-5p, miR-3128, miR-3178, miR-3910, miR-4443, miR-4445 and miR-4745, and the downregulated miRNAs miR-877-5p, miR-4487 and miR-4706 were identified as novel fat deposition regulators. DE miRNAs were further analyzed, along with previously identified differentially expressed genes (DEGs) from the same samples and predicted target genes, using multiple bioinformatic approaches, including target prediction tools and co-expression networks, as well as Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment. We identified DE miRNAs and their gene targets associated with bovine intramuscular adipogenesis, and we provide a basis for further functional investigations.
Collapse
Affiliation(s)
- Bilal Ahmad Mir
- Institute of Muscle Biology and Growth, Leibniz Institute for Farm Animal Biology (FBN), D-18196 Dummerstorf, Germany; (K.K.); (S.M.)
- Correspondence: ; Tel.: +49-38208-68885
| | - Henry Reyer
- Institute of Genome Biology, Leibniz Institute for Farm Animal Biology (FBN), D-18196 Dummerstorf, Germany; (H.R.); (S.P.); (C.K.)
| | - Katrin Komolka
- Institute of Muscle Biology and Growth, Leibniz Institute for Farm Animal Biology (FBN), D-18196 Dummerstorf, Germany; (K.K.); (S.M.)
| | - Siriluck Ponsuksili
- Institute of Genome Biology, Leibniz Institute for Farm Animal Biology (FBN), D-18196 Dummerstorf, Germany; (H.R.); (S.P.); (C.K.)
| | - Christa Kühn
- Institute of Genome Biology, Leibniz Institute for Farm Animal Biology (FBN), D-18196 Dummerstorf, Germany; (H.R.); (S.P.); (C.K.)
| | - Steffen Maak
- Institute of Muscle Biology and Growth, Leibniz Institute for Farm Animal Biology (FBN), D-18196 Dummerstorf, Germany; (K.K.); (S.M.)
| |
Collapse
|
21
|
Abstract
Obstructive sleep apnea (OSA), characterized by recurrent episodes of apnea during sleep and daytime sleepiness, seriously affects human health and may lead to systemic organ dysfunction. The pathogenesis of OSA is complex and still uncertain, but multiple surveys have shown that obesity is an important factor, and the incidence of OSA in people with obesity is as high as 30%. Adipokines are a group of proteins secreted from adipocytes, which are dysregulated in obesity and may contribute to OSA. Here, we review the most important and representative research results regarding the correlation between obesity-related adipokines including leptin, adiponectin, omentin-1, chemerin, and resistin and OSA in the past 5 years, provide an overview of these key adipokines, and analyze possible intrinsic mechanisms and influencing factors. The existing research shows that OSA is associated with an increase in the serum levels of leptin, chemerin, and resistin and a decrease in the levels of adiponectin and omentin-1; the findings presented here can be used to monitor the development of OSA and obesity, prevent future comorbidities, and identify risk factors for cardiovascular and other diseases, while different adipokines can be linked to OSA through different pathways such as insulin resistance, intermittent hypoxia, and inflammation, among others. We hope our review leads to a deeper and more comprehensive understanding of OSA based on the relevant literature, which will also provide directions for future clinical research.
Collapse
Affiliation(s)
- Xiongye Xu
- The First Clinical Medical College of Nanchang University, Nanchang, People's Republic of China
| | - Jixiong Xu
- The First Clinical Medical College of Nanchang University, Nanchang, People's Republic of China
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| |
Collapse
|
22
|
Gu X, Yao X, Liu D. Up-regulation of microRNA-335-5p reduces inflammation via negative regulation of the TPX2-mediated AKT/GSK3β signaling pathway in a chronic rhinosinusitis mouse model. Cell Signal 2020; 70:109596. [PMID: 32156642 DOI: 10.1016/j.cellsig.2020.109596] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 03/05/2020] [Accepted: 03/06/2020] [Indexed: 01/19/2023]
Abstract
Chronic rhinosinusitis (CRS) is featured with chronic symptoms of inflammation or infection in the nasal and sinus tissues. MicroRNAs (miRNAs/miRs), such as dysregulated expression of miR-125b and miR-26a, has been previously demonstrated to be related to CRS. The present study is intended to define the role of miR-335-5p in inflammation and the related mechanism in a mouse model of CRS. The differentially expressed genes associated with CRS were screened by microarray analysis. The targeting relationship between miR-335-5p and TPX2 was analyzed by target prediction program and dual luciferase reporter gene assay. The mouse model of CRS was established, and mice were introduced with miR-335-5p mimics, miR-335-5p inhibitors, or siRNA against TPX2 to explore the regulatory functions of miR-335-5p. The regulatory effect of miR-335-5p on inflammation with the involvement of the AKT signaling pathway was also analyzed with the expression of inflammatory cytokines and AKT signaling pathway-related factors measured. It was indicated that miR-335-5p regulated the TPX2 gene-mediated AKT signaling pathway. TPX2 was identified as a target gene of miR-335-5p, and miR-335-5p elevation inhibited the activation of the AKT signaling pathway. In mice with CRS, up-regulation of miR-335-5p or silence of TPX2 inhibited the inflammation, as evidenced by decreased levels of TNF-α, IL-6 and IL-8, and higher levels of GSK3β and IL-10. Collectively, miR-335-5p inhibits the activation of AKT signaling pathway by negatively mediating TPX2, which may confer anti-inflammatory protection in CRS.
Collapse
Affiliation(s)
- Xiao Gu
- Department of E.N.T, Linyi People's Hospital, Linyi 276000, PR China
| | - Xiaocui Yao
- Clinical Laboratory, Linyi People's Hospital, Linyi 276000, PR China
| | - Dengtao Liu
- Clinical Laboratory, Linyi People's Hospital, Linyi 276000, PR China.
| |
Collapse
|
23
|
Green Tea Prevents NAFLD by Modulation of miR-34a and miR-194 Expression in a High-Fat Diet Mouse Model. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:4168380. [PMID: 31885789 PMCID: PMC6914886 DOI: 10.1155/2019/4168380] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 10/11/2019] [Accepted: 11/05/2019] [Indexed: 02/07/2023]
Abstract
Background/Aims Nonalcoholic fatty liver disease (NAFLD) is considered the hepatic manifestation of metabolic syndrome. It is currently the most common chronic liver disease with complex pathogenesis and challenging treatment. Here, we investigated the hepatoprotective role of green tea (GT) and determined the involvement of miRNAs and its mechanism of action. Methods Male C57Bl/6 mice were fed with a high-fat diet for 4 weeks. After this period, the animals received gavage with GT (500 mg/kg body weight) over 12 weeks (5 days/week). HepG2 cell lines were transfected with miR-34a or miR-194 mimetics and inhibitors to validate the in vivo results or were treated with TNF-α to evaluate miRNA regulation. Results GT supplementation protects against NAFLD development by altering lipid metabolism, increasing gene expression involved in triglycerides and fatty acid catabolism, and decreasing uptake and lipid accumulation. This phenotype was accompanied by miR-34a downregulation and an increase in their mRNA targets Sirt1, Pparα, and Insig2. GT upregulated hepatic miR-194 by inhibiting TNF-α action leading to a decrease in miR-194 target genes Hmgcs/Apoa5. Conclusion Our study identified for the first time that the beneficial effects of GT in the liver can be due to the modulation of miRNAs, opening new perspectives for the treatment of NAFLD focusing on epigenetic regulation of miR-34a and miR-194 as green tea targets.
Collapse
|
24
|
Gholami M, Larijani B, Zahedi Z, Mahmoudian F, Bahrami S, Omran SP, Saadatian Z, Hasani-Ranjbar S, Taslimi R, Bastami M, Amoli MM. Inflammation related miRNAs as an important player between obesity and cancers. J Diabetes Metab Disord 2019; 18:675-692. [PMID: 31890692 PMCID: PMC6915181 DOI: 10.1007/s40200-019-00459-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 10/23/2019] [Indexed: 12/13/2022]
Abstract
The growing trend in addition to their burden, prevalence, and death has made obesity and cancer two of the most concerning diseases worldwide. Obesity is an important risk factor for common types of cancers where the risk of some cancers is directly related to the obesity. Various inflammatory mechanisms and increased level of pro-inflammatory cytokines have been investigated in many previous studies, which play key roles in the pathophysiology and development of both of these conditions. On the other hand, in the recent years, many studies have individually focused on the biomarker's role and therapeutic targeting of microRNAs (miRNAs) in different types of cancers and obesity including newly discovered small noncoding RNAs (sncRNAs) which regulate gene expression and RNA silencing. This study is a comprehensive review of the main inflammation related miRNAs in obesity/obesity related traits. For the first time, the main roles of miRNAs in obesity related cancers have been discussed in response to the question raised in the following hypothesis; do the main inflammatory miRNAs link obesity with obesity-related cancers regarding their role as biomarkers? Graphical abstractConceptual design of inflammatory miRNAs which provide link between obesity and cancers.
Collapse
Affiliation(s)
- Morteza Gholami
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Zhila Zahedi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Mahmoudian
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Samira Bahrami
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sima Parvizi Omran
- Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, 5th floor, Shariati Hospital, North Kargar Ave, Tehran, Iran
| | - Zahra Saadatian
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shirin Hasani-Ranjbar
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Taslimi
- Department of Gastroenterology, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, IR Iran
| | - Milad Bastami
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahsa M. Amoli
- Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, 5th floor, Shariati Hospital, North Kargar Ave, Tehran, Iran
| |
Collapse
|
25
|
Androgen-Regulated microRNAs (AndroMiRs) as Novel Players in Adipogenesis. Int J Mol Sci 2019; 20:ijms20225767. [PMID: 31744106 PMCID: PMC6888160 DOI: 10.3390/ijms20225767] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 11/11/2019] [Accepted: 11/12/2019] [Indexed: 12/14/2022] Open
Abstract
The development, homeostasis, or increase of the adipose tissue is driven by the induction of the adipogenic differentiation (adipogenesis) of undifferentiated mesenchymal stem cells (MSCs). Adipogenesis can be inhibited by androgen stimulation of these MSCs resulting in the transcription initiation or repression of androgen receptor (AR) regulated genes. AR not only regulates the transcription of protein-coding genes but also the transcription of several non-coding microRNAs involved in the posttranscriptional gene regulation (herein designated as AndroMiRs). As microRNAs are largely involved in differentiation processes such as adipogenesis, the involvement of AndroMiRs in the androgen-mediated inhibition of adipogenesis is likely, however, not yet intensively studied. In this review, existing knowledge about adipogenesis-related microRNAs and AndroMiRs is summarized, and putative cross-links are drawn, which are still prone to experimental validation.
Collapse
|
26
|
Ghasemi A, Hashemy SI, Azimi-Nezhad M, Dehghani A, Saeidi J, Mohtashami M. The cross-talk between adipokines and miRNAs in health and obesity-mediated diseases. Clin Chim Acta 2019; 499:41-53. [PMID: 31476303 DOI: 10.1016/j.cca.2019.08.028] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 08/28/2019] [Accepted: 08/28/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND Multiple studies have revealed a direct correlation between obesity and the development of multiple comorbidities, including metabolic diseases, cardiovascular disorders, chronic inflammatory disease, and cancers. However, the molecular mechanism underlying the link between obesity and the progression of these diseases is not completely understood. Adipokines are factors that are secreted by adipocytes and play a key role in whole body homeostasis. Collaboratively, miRNAs are suggested to have key functions in the development of obesity and obesity-related disorders. Based on recently emerging evidence, obesity leads to the dysregulation of both adipokines and obesity-related miRNAs. In the present study, we described the correlations between obesity and its related diseases that are mediated by the mutual regulatory effects of adipokines and miRNAs. METHODS We reviewed current knowledge of the modulatory effects of adipokines on miRNAs activity and their relevant functions in pathological conditions and vice versa. RESULTS Our research reveals the ability of adipokines and miRNAs to control the expression and activity of the other class of molecules, and their effects on obesity-related diseases. CONCLUSIONS This study may help researchers develop a roadmap for future investigations and provide opportunities to develop new therapeutic and diagnostic methods for treating obesity-related diseases.
Collapse
Affiliation(s)
- Ahmad Ghasemi
- Non-communicable Disease Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran.
| | - Seyed Isaac Hashemy
- Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Mohsen Azimi-Nezhad
- Non-communicable Disease Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran; UMR INSERM U 1122, IGE-PCV, Interactions Gène-Environment en Physiopathologie Cardiovascular Université de Lorraine, France
| | - Alireza Dehghani
- Institute of Biochemistry and Molecular Biology, University of Bonn, Bonn, Germany
| | - Jafar Saeidi
- Department of Physiology, School of Basic Science, Neyshabur Branch, Islamic Azad University, Neyshabur, Iran
| | - Mahnaz Mohtashami
- Department of Biology, School of Basic Science, Neyshabur Branch, Islamic Azad University, Neyshabur, Iran
| |
Collapse
|
27
|
Wróblewski A, Strycharz J, Świderska E, Drewniak K, Drzewoski J, Szemraj J, Kasznicki J, Śliwińska A. Molecular Insight into the Interaction between Epigenetics and Leptin in Metabolic Disorders. Nutrients 2019; 11:nu11081872. [PMID: 31408957 PMCID: PMC6723573 DOI: 10.3390/nu11081872] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 08/07/2019] [Accepted: 08/09/2019] [Indexed: 12/16/2022] Open
Abstract
Nowadays, it is well-known that the deregulation of epigenetic machinery is a common biological event leading to the development and progression of metabolic disorders. Moreover, the expression level and actions of leptin, a vast adipocytokine regulating energy metabolism, appear to be strongly associated with epigenetics. Therefore, the aim of this review was to summarize the current knowledge of the epigenetic regulation of leptin as well as the leptin-induced epigenetic modifications in metabolic disorders and associated phenomena. The collected data indicated that the deregulation of leptin expression and secretion that occurs during the course of metabolic diseases is underlain by a variation in the level of promoter methylation, the occurrence of histone modifications, along with miRNA interference. Furthermore, leptin was proven to epigenetically regulate several miRNAs and affect the activity of the histone deacetylases. These epigenetic modifications were observed in obesity, gestational diabetes, metabolic syndrome and concerned various molecular processes like glucose metabolism, insulin sensitivity, liver fibrosis, obesity-related carcinogenesis, adipogenesis or fetal/early postnatal programming. Moreover, the circulating miRNA profiles were associated with the plasma leptin level in metabolic syndrome, and miRNAs were found to be involved in hypothalamic leptin sensitivity. In summary, the evidence suggests that leptin is both a target and a mediator of epigenetic changes that develop in numerous tissues during metabolic disorders.
Collapse
Affiliation(s)
- Adam Wróblewski
- Department of Medical Biochemistry, Medical University of Lodz, 6/8 Mazowiecka Str., 92-215 Lodz, Poland.
| | - Justyna Strycharz
- Department of Medical Biochemistry, Medical University of Lodz, 6/8 Mazowiecka Str., 92-215 Lodz, Poland
| | - Ewa Świderska
- Department of Medical Biochemistry, Medical University of Lodz, 6/8 Mazowiecka Str., 92-215 Lodz, Poland
| | - Karolina Drewniak
- Student Scientific Society of the Civilization Diseases, Medical University of Lodz, 251 Pomorska Str., 92-213 Lodz, Poland
| | - Józef Drzewoski
- Central Teaching Hospital of the Medical University of Lodz, 251 Pomorska Str., 92-213 Lodz, Poland
| | - Janusz Szemraj
- Department of Medical Biochemistry, Medical University of Lodz, 6/8 Mazowiecka Str., 92-215 Lodz, Poland
| | - Jacek Kasznicki
- Department of Internal Diseases, Diabetology and Clinical Pharmacology, Medical University of Lodz, 251 Pomorska Str., 92-213 Lodz, Poland
| | - Agnieszka Śliwińska
- Department of Nucleic Acid Biochemistry, Medical University of Lodz, 251 Pomorska Str., 92-213 Lodz, Poland.
| |
Collapse
|
28
|
Landrier JF, Derghal A, Mounien L. MicroRNAs in Obesity and Related Metabolic Disorders. Cells 2019; 8:cells8080859. [PMID: 31404962 PMCID: PMC6721826 DOI: 10.3390/cells8080859] [Citation(s) in RCA: 146] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 08/01/2019] [Accepted: 08/07/2019] [Indexed: 12/14/2022] Open
Abstract
Metabolic disorders are characterized by the inability to properly use and/or store energy. The burdens of metabolic disease, such as obesity or diabetes, are believed to arise through a complex interplay between genetics and epigenetics predisposition, environment and nutrition. Therefore, understanding the molecular mechanisms for the onset of metabolic disease will provide new insights for prevention and treatment. There is growing concern about the dysregulation of micro-RNAs (miRNAs) in metabolic diseases. MiRNAs are short non-coding RNA molecules that post-transcriptionally repress the expression of genes by binding to untranslated regions and coding sequences of the target mRNAs. This review aims to provide recent data about the potential involvement of miRNAs in metabolic diseases, particularly obesity and type 2 diabetes.
Collapse
Affiliation(s)
| | - Adel Derghal
- Aix Marseille Univ, INSERM, INRA, C2VN, 13005 Marseille, France
| | - Lourdes Mounien
- Aix Marseille Univ, INSERM, INRA, C2VN, 13005 Marseille, France.
| |
Collapse
|
29
|
Gao Y, Wang Y, Chen X, Peng Y, Chen F, He Y, Pang W, Yang G, Yu T. MiR‐127 attenuates adipogenesis by targeting MAPK4 and HOXC6 in porcine adipocytes. J Cell Physiol 2019; 234:21838-21850. [DOI: 10.1002/jcp.28660] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 03/21/2019] [Accepted: 03/25/2019] [Indexed: 01/16/2023]
Affiliation(s)
- Yun Gao
- Laboratory of Animal Gennetics, Breeding and Reproducation of Shaanxi Province, Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology Northwest A&F University Yangling Shaanxi 712100 China
- Department of Molecular Biosciences, The Wenner‐Gren Institute Stockholm University Stockholm Sweden
| | - Yingqian Wang
- Laboratory of Animal Gennetics, Breeding and Reproducation of Shaanxi Province, Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology Northwest A&F University Yangling Shaanxi 712100 China
| | - Xiaochang Chen
- Laboratory of Animal Gennetics, Breeding and Reproducation of Shaanxi Province, Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology Northwest A&F University Yangling Shaanxi 712100 China
| | - Ying Peng
- Laboratory of Animal Gennetics, Breeding and Reproducation of Shaanxi Province, Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology Northwest A&F University Yangling Shaanxi 712100 China
| | - Fenfen Chen
- Laboratory of Animal Gennetics, Breeding and Reproducation of Shaanxi Province, Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology Northwest A&F University Yangling Shaanxi 712100 China
- Faculty of Life Sciences Southwest Forestry University Kunming China
| | - Yulin He
- Laboratory of Animal Gennetics, Breeding and Reproducation of Shaanxi Province, Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology Northwest A&F University Yangling Shaanxi 712100 China
| | - Weijun Pang
- Laboratory of Animal Gennetics, Breeding and Reproducation of Shaanxi Province, Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology Northwest A&F University Yangling Shaanxi 712100 China
| | - Gongshe Yang
- Laboratory of Animal Gennetics, Breeding and Reproducation of Shaanxi Province, Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology Northwest A&F University Yangling Shaanxi 712100 China
| | - Taiyong Yu
- Laboratory of Animal Gennetics, Breeding and Reproducation of Shaanxi Province, Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology Northwest A&F University Yangling Shaanxi 712100 China
| |
Collapse
|
30
|
Jiang N, Li Y, Shu T, Wang J. Cytokines and inflammation in adipogenesis: an updated review. Front Med 2019; 13:314-329. [PMID: 30066061 DOI: 10.1007/s11684-018-0625-0] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 12/12/2017] [Indexed: 02/07/2023]
Abstract
The biological relevance of cytokines is known for more than 20 years. Evidence suggests that adipogenesis is one of the biological events involved in the regulation of cytokines, and pro-inflammatory cytokines (e.g., TNFα and IL-1β) inhibit adipogenesis through various pathways. This inhibitory effect can constrain the hyperplastic expandability of adipose tissues. Meanwhile, chronic low-grade inflammation is commonly observed in obese populations. In some individuals, the impaired ability of adipose tissues to recruit new adipocytes to adipose depots during overnutrition results in adipocyte hypertrophy, ectopic lipid accumulation, and insulin resistance. Intervention studies showed that pro-inflammatory cytokine antagonists improve metabolism in patients with metabolic syndrome. This review focuses on the cytokines currently known to regulate adipogenesis under physiological and pathophysiological circumstances. Recent studies on how inhibited adipogenesis leads to metabolic disorders were summarized. Although the interplay of cytokines and lipid metabolism is yet incompletely understood, cytokines represent a class of potential therapeutic targets in the treatment of metabolic disorders.
Collapse
Affiliation(s)
- Ning Jiang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing, 100730, China
| | - Yao Li
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing, 100730, China
| | - Ting Shu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing, 100730, China
| | - Jing Wang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
31
|
Abstract
One of the biggest challenges in the management of obesity is the prevention of weight regain after successful weight loss. Weight regain after weight loss has large interindividual variation. Although many factors probably contribute to this variation, we hypothesize that variability in biological responses associated with weight loss-induced shrinking of subcutaneous adipocytes has an important role. In this Review, we show that weight loss-induced variations in cellular stress, extracellular matrix remodelling, inflammatory responses, adipokine secretion and lipolysis seem to be associated with the amount of weight that is regained after successful weight loss. Weight regain could therefore, at least in part, depend on a combination of these factors. Further research on the causality of these associations could aid the development of effective strategies to prevent weight regain after successful weight loss.
Collapse
Affiliation(s)
- Marleen A van Baak
- NUTRIM School for Nutrition and Translational Research in Metabolism, Department of Human Biology, Maastricht University, Maastricht, Netherlands.
| | - Edwin C M Mariman
- NUTRIM School for Nutrition and Translational Research in Metabolism, Department of Human Biology, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
32
|
Hanousková B, Neprašová B, Skálová L, Maletínská L, Zemanová K, Ambrož M, Matoušková P. High-fructose drinks affect microRNAs expression differently in lean and obese mice. J Nutr Biochem 2019; 68:42-50. [PMID: 31030166 DOI: 10.1016/j.jnutbio.2019.03.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 12/17/2018] [Accepted: 03/08/2019] [Indexed: 12/19/2022]
Abstract
High fructose intake from soft drinks and sweets is assumed to have a negative impact on human health. Yet in spite of intensive research, the molecular mechanisms of these effects have not been fully elucidated yet, for example, the effect of high fructose intake could be different in normal and obese individuals. Four groups of mice were used in this study: control groups of lean mice and mice with obesity induced by a high-fat diet, then both of these groups with or without fructose administration in drinks. In plasma of each group, triacylglycerol, cholesterol, free fatty acids, alanine aminotransferase, insulin and adiponectin were measured. The expression levels of selected microRNAs (miRNAs) in plasma, the liver, white adipose tissue, brown adipose tissue and subcutaneous adipose tissue were quantified. In both lean and obese mice, high fructose intake increased cholesterol amount in the liver, up-regulated hepatic miR-27a, down-regulated miR-33a in white adipose tissue and increased plasmatic level of miR-21. The effect of high fructose intake on other miRNAs in the liver, plasma and adipose tissues differed in normal and obese mice. Fructose intake led to hepatic hypercholesterolemia and aberrant expression of several miRNAs participating in lipid metabolism, adipocytes differentiation and nonalcoholic fatty liver disease promotion. The effect of fructose on miRNAs expression differed in normal and obese mice. Nevertheless, plasmatic miR-21, which was induced by fructose in both lean and obese mice, may be considered as a potential biomarker of excessive fructose intake.
Collapse
Affiliation(s)
- Barbora Hanousková
- Department of Biochemical Sciences, Faculty of Pharmacy, Charles University, Akademika Heyrovského 1203/8, Hradec Králové, Czech Republic.
| | - Barbora Neprašová
- Czech Academy of Sciences, Institute of Organic Chemistry and Biochemistry, Flemingovo náměstí 542/2, Prague, Czech Republic.
| | - Lenka Skálová
- Department of Biochemical Sciences, Faculty of Pharmacy, Charles University, Akademika Heyrovského 1203/8, Hradec Králové, Czech Republic.
| | - Lenka Maletínská
- Czech Academy of Sciences, Institute of Organic Chemistry and Biochemistry, Flemingovo náměstí 542/2, Prague, Czech Republic.
| | - Kateřina Zemanová
- Department of Biochemical Sciences, Faculty of Pharmacy, Charles University, Akademika Heyrovského 1203/8, Hradec Králové, Czech Republic.
| | - Martin Ambrož
- Department of Biochemical Sciences, Faculty of Pharmacy, Charles University, Akademika Heyrovského 1203/8, Hradec Králové, Czech Republic.
| | - Petra Matoušková
- Department of Biochemical Sciences, Faculty of Pharmacy, Charles University, Akademika Heyrovského 1203/8, Hradec Králové, Czech Republic.
| |
Collapse
|
33
|
Palhinha L, Liechocki S, Hottz ED, Pereira JADS, de Almeida CJ, Moraes-Vieira PMM, Bozza PT, Maya-Monteiro CM. Leptin Induces Proadipogenic and Proinflammatory Signaling in Adipocytes. Front Endocrinol (Lausanne) 2019; 10:841. [PMID: 31920961 PMCID: PMC6923660 DOI: 10.3389/fendo.2019.00841] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 11/19/2019] [Indexed: 12/12/2022] Open
Abstract
Background: Leptin is an adipokine with well-known effects on the central nervous system including the induction of energy expenditure and satiety. Leptin also has major relevance when activating immune cells and modulating inflammatory response. In obesity, increases in white adipose tissue accumulation and leptin levels are accompanied by hypothalamic resistance to leptin. Even though the adipose tissue is a leptin-rich environment, the local actions of leptin regarding adipogenesis were not thoroughly investigated until now. Here we evaluate the contributions of leptins direct signaling in preadipocytes and adipose tissue-derived stromal cells (ASCs) for adipogenesis. Methods: Adipocytes were differentiated from the murine lineage of preadipocytes 3T3-L1 or ASCs from subcutaneous and visceral (retroperitoneal) fat depots from C57Bl/6J mice. Differentiating cells were treated with leptin in addition to or in replacement of insulin. The advance of adipogenesis was assessed by the expression and secretion of adipogenesis- and lipogenesis-related proteins by Western blot and immunoenzimatic assays, and the accumulation of lipid droplets by fluorescence microscopy. Results: Leptin treatment in 3T3-L1 preadipocytes or ASCs increased the production of the adipogenesis- and lipogenesis-related proteins PLIN1, CAV-1, PPARγ, SREBP1C, and/or adiponectin at earlier stages of differentiation. In 3T3-L1 preadipocytes, we found that leptin induced lipid droplets' formation in an mTOR-dependent manner. Also, leptin induced a proinflammatory cytokine profile in 3T3-L1 and ASCs, modulating the production of TNF-α, IL-10, and IL-6. Since insulin is considered an essential factor for preadipocyte differentiation, we asked whether leptin would support adipogenesis in the absence of insulin. Importantly, leptin induced the formation of lipid droplets and the expression of adipogenesis-related proteins independently of insulin during the differentiation of 3T3-L1 cells and ASCs. Conclusions: Our results demonstrate that leptin induces intracellular signaling in preadipocytes and adipocytes promoting adipogenesis and modulating the secretion of inflammatory mediators. Also, leptin restores adipogenesis in the absence of insulin. These findings contribute to the understanding of the local signaling of leptin in precursor and mature adipose cells. The proadipogenic role of leptin unraveled here may be of especial relevance during obesity, when its central signaling is defective.
Collapse
Affiliation(s)
- Lohanna Palhinha
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, Brazil
| | - Sally Liechocki
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, Brazil
| | - Eugenio D. Hottz
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, Brazil
- Laboratory of Glycoconjugates Analysis, Department of Biochemistry, Federal University of Juiz de Fora (UFJF), Juiz de Fora, Brazil
| | - Jéssica Aparecida da Silva Pereira
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, Brazil
- Post-Graduate Program in Immunology, Institute of Biological Sciences, University of Sao Paulo, São Paulo, Brazil
| | - Cecília J. de Almeida
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, Brazil
| | - Pedro Manoel M. Moraes-Vieira
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, Brazil
- Post-Graduate Program in Immunology, Institute of Biological Sciences, University of Sao Paulo, São Paulo, Brazil
- Experimental Medicine Research Cluster, EMRC, University of Cammpinas, Campinas, Brazil
| | - Patrícia T. Bozza
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, Brazil
| | - Clarissa Menezes Maya-Monteiro
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, Brazil
- *Correspondence: Clarissa Menezes Maya-Monteiro ;
| |
Collapse
|
34
|
Jasinski-Bergner S, Kielstein H. Adipokines Regulate the Expression of Tumor-Relevant MicroRNAs. Obes Facts 2019; 12:211-225. [PMID: 30999294 PMCID: PMC6547259 DOI: 10.1159/000496625] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 01/05/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Increasing prevalence of obesity requires the investigation of respective comorbidities, including tumor diseases like colorectal, renal, post-menopausal breast, prostate cancer, and leukemia. To date, molecular mechanisms of the malignant transformation of these peripheral tissues induced by obesity remain unclear. Adipose tissue secretes factors with hormone-like functions, the adipokines, and is therefore categorized as an endocrine organ. Current research demonstrates the ability of adipose tissue to alter DNA methylation and gene expression in peripheral tissues, probably affecting microRNA (miR) expression. METHODS Literature was analyzed for adipokine-regulated miRs. Many of these adipokine upregulated or downregulated miRs exert either oncogenic or anti-tumoral potential. RESULTS The three selected and analyzed adipokines, adiponectin, leptin, and resistin, induce more strongly oncogenic miRs and simultaneously reduce anti-tumoral miRs than vice versa. This effect is not only true for the pure number of regulated miRs, it is also the case by consideration of the abundance of the respective miR expression based on actual data sets derived from next-generation sequencing. CONCLUSION The link of obesity and cancer is analyzed under the aspect of adipokine-regulated miRs. At the same time the impact of miR abundance is considered as a regulatory variable. This context offers new strategies for tumor therapy and diagnostics.
Collapse
Affiliation(s)
- Simon Jasinski-Bergner
- Department of Anatomy and Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany,
| | - Heike Kielstein
- Department of Anatomy and Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| |
Collapse
|
35
|
Corrêa TA, Rogero MM. Polyphenols regulating microRNAs and inflammation biomarkers in obesity. Nutrition 2018; 59:150-157. [PMID: 30471527 DOI: 10.1016/j.nut.2018.08.010] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 07/30/2018] [Accepted: 08/01/2018] [Indexed: 12/22/2022]
Abstract
Obesity is one of the most prevalent health problems worldwide. It is a complex disease that is generally accompanied by insulin resistance, increases in oxidative stress and inflammation biomarkers, and potentially, microRNA (miRNA) dysregulation. Polyphenols may act on obesity and its metabolic consequences. Circulating miRNAs have been studied as potential biomarkers for inflammatory and metabolic diseases, and their use may improve the diagnostic tools currently available and the ability to diagnose specific diseases. To our knowledge, data regarding the link between the consumption of polyphenols from food sources, miRNA expression, and inflammation biomarkers related to obesity is scarce, and most data available describing this relationship are found in cancer studies. This review focuses on the polyphenols that modulate the metabolism, inflammation, or both related to obesity to understand the extent to which miRNA expression can be modulated by dietary interventions.
Collapse
Affiliation(s)
- Telma Af Corrêa
- Department of Nutrition, School of Public Health, University of São Paulo, São Paulo, Brazil
| | - Marcelo M Rogero
- Department of Nutrition, School of Public Health, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
36
|
Salunkhe VA, Ofori JK, Gandasi NR, Salö SA, Hansson S, Andersson ME, Wendt A, Barg S, Esguerra JLS, Eliasson L. MiR-335 overexpression impairs insulin secretion through defective priming of insulin vesicles. Physiol Rep 2018; 5:5/21/e13493. [PMID: 29122960 PMCID: PMC5688784 DOI: 10.14814/phy2.13493] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 10/11/2017] [Accepted: 10/13/2017] [Indexed: 01/01/2023] Open
Abstract
MicroRNAs contribute to the maintenance of optimal cellular functions by fine‐tuning protein expression levels. In the pancreatic β‐cells, imbalances in the exocytotic machinery components lead to impaired insulin secretion and type 2 diabetes (T2D). We hypothesize that dysregulated miRNA expression exacerbates β‐cell dysfunction, and have earlier shown that islets from the diabetic GK‐rat model have increased expression of miRNAs, including miR‐335‐5p (miR‐335). Here, we aim to determine the specific role of miR‐335 during development of T2D, and the influence of this miRNA on glucose‐stimulated insulin secretion and Ca2+‐dependent exocytosis. We found that the expression of miR‐335 negatively correlated with secretion index in human islets of individuals with prediabetes. Overexpression of miR‐335 in human EndoC‐βH1 and in rat INS‐1 832/13 cells (OE335) resulted in decreased glucose‐stimulated insulin secretion, and OE335 cells showed concomitant reduction in three exocytotic proteins: SNAP25, Syntaxin‐binding protein 1 (STXBP1), and synaptotagmin 11 (SYT11). Single‐cell capacitance measurements, complemented with TIRF microscopy of the granule marker NPY‐mEGFP demonstrated a significant reduction in exocytosis in OE335 cells. The reduction was not associated with defective docking or decreased Ca2+ current. More likely, it is a direct consequence of impaired priming of already docked granules. Earlier reports have proposed reduced granular priming as the cause of reduced first‐phase insulin secretion during prediabetes. Here, we show a specific role of miR‐335 in regulating insulin secretion during this transition period. Moreover, we can conclude that miR‐335 has the capacity to modulate insulin secretion and Ca2+‐dependent exocytosis through effects on granular priming.
Collapse
Affiliation(s)
- Vishal A Salunkhe
- Department of Clinical Sciences Malmö, Islet Cell Exocytosis Lund University Diabetes Centre Lund University, Malmö, Sweden
| | - Jones K Ofori
- Department of Clinical Sciences Malmö, Islet Cell Exocytosis Lund University Diabetes Centre Lund University, Malmö, Sweden
| | - Nikhil R Gandasi
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Sofia A Salö
- Department of Clinical Sciences Malmö, Islet Cell Exocytosis Lund University Diabetes Centre Lund University, Malmö, Sweden
| | - Sofia Hansson
- Department of Clinical Sciences Malmö, Islet Cell Exocytosis Lund University Diabetes Centre Lund University, Malmö, Sweden
| | - Markus E Andersson
- Department of Clinical Sciences Malmö, Islet Cell Exocytosis Lund University Diabetes Centre Lund University, Malmö, Sweden
| | - Anna Wendt
- Department of Clinical Sciences Malmö, Islet Cell Exocytosis Lund University Diabetes Centre Lund University, Malmö, Sweden
| | - Sebastian Barg
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Jonathan L S Esguerra
- Department of Clinical Sciences Malmö, Islet Cell Exocytosis Lund University Diabetes Centre Lund University, Malmö, Sweden
| | - Lena Eliasson
- Department of Clinical Sciences Malmö, Islet Cell Exocytosis Lund University Diabetes Centre Lund University, Malmö, Sweden
| |
Collapse
|
37
|
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
|
38
|
Gao XL, Li JQ, Dong YT, Cheng EJ, Gong JN, Qin YL, Huang YQ, Yang JJ, Wang SJ, An DD. Upregulation of microRNA-335-5p reduces inflammatory responses by inhibiting FASN through the activation of AMPK/ULK1 signaling pathway in a septic mouse model. Cytokine 2018; 110:466-478. [PMID: 29866515 DOI: 10.1016/j.cyto.2018.05.016] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 04/28/2018] [Accepted: 05/17/2018] [Indexed: 12/22/2022]
Abstract
Sepsis, as a systemic inflammatory response syndrome (SIRS) subtype, is generally characterized by infection. Emerging evidence has highlighted dysregulated microRNAs (miRNAs) are involved in the progression of sepsis. The aim of the study was to investigate the effects of miR-335-5p on inflammatory responses in a septic mouse model. The hypothesis was subsequently asserted that the FASN gene and AMPK/ULK1 signaling pathway may participate in the regulation of miR-335-5p. A septic mouse model was established in order to validate the effect of miR-335-5p on the inflammatory response by means of suppressing the endogenous expression of FASN by siRNA against FASN in endothelial cells. A target prediction program and luciferase activity was employed to ascertain as to whether miR--335-5p targets FASN. The levels of inflammatory factors including IL-6 and IL-1β were determined by means of ELISA assay. RT-qPCR and western blot analysis were used to determine the AMPK/ULK1 signaling pathway-, apoptosis- and autophagy-related genes. Flow cytometry was employed in order to evaluate sepsis-induced cell apoptosis in response to miR-335-5p and FASN alternations. FASN was identified as a target gene of miR--335-5p. Gain- and loss-of-function studies revealed that miR-335-5p acted to enhance autophagy, reduce cell apoptosis, promote cell cycle entry in endothelial cells, and reduce inflammatory response through the modulation of pro- and anti-apoptotic factors in endothelial cells. The effect of miR-335-5p on endothelial cells was increased when FASN was suppressed by siRNA as well as when the AMPK/ULK1 signaling pathway was activated, suggesting that miR-335-5p influences sepsis by targeting and inhibiting FASN, and activating the AMPK/ULK1 signaling pathway. Our study provides evidence indicating that overexpressed miR-335-5p enhances autophagy by targeting FASN through activation of the AMPK/ULK1 signaling pathway working to alleviate the inflammatory response in septic mouse models, emphasizing the value of the functional upregulation of miR-335-5p as therapeutic strategy for sepsis.
Collapse
Affiliation(s)
- Xiao-Ling Gao
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Shanxi Medical University, Taiyuan 030001, PR China
| | - Jian-Qiang Li
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Shanxi Medical University, Taiyuan 030001, PR China.
| | - Yan-Ting Dong
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Shanxi Medical University, Taiyuan 030001, PR China
| | - Er-Jing Cheng
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Shanxi Medical University, Taiyuan 030001, PR China
| | - Jian-Nan Gong
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Shanxi Medical University, Taiyuan 030001, PR China
| | - Yi-Li Qin
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Shanxi Medical University, Taiyuan 030001, PR China
| | | | | | | | - Dong-Dong An
- Shanxi Medical University, Taiyuan 030000, PR China
| |
Collapse
|
39
|
Matoušková P, Hanousková B, Skálová L. MicroRNAs as Potential Regulators of Glutathione Peroxidases Expression and Their Role in Obesity and Related Pathologies. Int J Mol Sci 2018; 19:ijms19041199. [PMID: 29662007 PMCID: PMC5979329 DOI: 10.3390/ijms19041199] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 04/08/2018] [Accepted: 04/10/2018] [Indexed: 12/19/2022] Open
Abstract
Glutathione peroxidases (GPxs) belong to the eight-member family of phylogenetically related enzymes with different cellular localization, but distinct antioxidant function. Several GPxs are important selenoproteins. Dysregulated GPx expression is connected with severe pathologies, including obesity and diabetes. We performed a comprehensive bioinformatic analysis using the programs miRDB, miRanda, TargetScan, and Diana in the search for hypothetical microRNAs targeting 3′untranslated regions (3´UTR) of GPxs. We cross-referenced the literature for possible intersections between our results and available reports on identified microRNAs, with a special focus on the microRNAs related to oxidative stress, obesity, and related pathologies. We identified many microRNAs with an association with oxidative stress and obesity as putative regulators of GPxs. In particular, miR-185-5p was predicted by a larger number of programs to target six GPxs and thus could play the role as their master regulator. This microRNA was altered by selenium deficiency and can play a role as a feedback control of selenoproteins’ expression. Through the bioinformatics analysis we revealed the potential connection of microRNAs, GPxs, obesity, and other redox imbalance related diseases.
Collapse
Affiliation(s)
- Petra Matoušková
- Faculty of Pharmacy, Department of Biochemical Sciences, Charles University, 500 05, Hradec Králové, Czech Republic.
| | - Barbora Hanousková
- Faculty of Pharmacy, Department of Biochemical Sciences, Charles University, 500 05, Hradec Králové, Czech Republic.
| | - Lenka Skálová
- Faculty of Pharmacy, Department of Biochemical Sciences, Charles University, 500 05, Hradec Králové, Czech Republic.
| |
Collapse
|
40
|
Otton R, Bolin AP, Ferreira LT, Marinovic MP, Rocha ALS, Mori MA. Polyphenol-rich green tea extract improves adipose tissue metabolism by down-regulating miR-335 expression and mitigating insulin resistance and inflammation. J Nutr Biochem 2018; 57:170-179. [PMID: 29734116 DOI: 10.1016/j.jnutbio.2018.03.024] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 03/19/2018] [Accepted: 03/23/2018] [Indexed: 01/07/2023]
Abstract
Obesity leads to changes in miRNA expression in adipose tissue, and this modulation is linked to the pathophysiology of the disease. Green tea (GT) is a natural source of polyphenols that have been shown to confer health benefits, particularly preventing metabolic diseases. Here, we investigated if the beneficial effects of GT in obesity results from changes in the miRNA profile in white adipose tissue. GT treatment [500 mg/body weight (BW)/12 weeks] increased energy expenditure of high-fat diet-fed mice (16 weeks), leading to reduced weight gain, decreased adiposity, reduced inflammation and improved insulin sensitivity. These phenotypes were associated with a decrease in the expression of miR-335 in the adipose tissue. miR-335 was up-regulated by TNF-α in adipocytes and, in turn, down-regulated genes involved in insulin signaling and lipid metabolism. On the other hand, GT inhibited TNF-α effect. In conclusion, miR-335 serves as a link between inflammation and impaired metabolism in adipose tissue, providing an important mechanistic insight into the molecular basis underlying GT action during obesity.
Collapse
Affiliation(s)
- Rosemari Otton
- Interdisciplinary Post-graduate Program in Health Sciences, CBS, Cruzeiro do Sul University, São Paulo, 03342-000, Brazil.
| | - Anaysa Paola Bolin
- Department of Pharmacology, Biomedical Sciences Institute, University of São Paulo, São Paulo, 05508-000, Brazil
| | - Leticia Torres Ferreira
- Interdisciplinary Post-graduate Program in Health Sciences, CBS, Cruzeiro do Sul University, São Paulo, 03342-000, Brazil
| | - Marcelo Paradiso Marinovic
- Interdisciplinary Post-graduate Program in Health Sciences, CBS, Cruzeiro do Sul University, São Paulo, 03342-000, Brazil
| | - Andrea Livia Silva Rocha
- Department of Biochemistry and Tissue Biology, Universidade Estadual de Campinas, Campinas, 13083-862, Brazil
| | - Marcelo Alves Mori
- Department of Biochemistry and Tissue Biology, Universidade Estadual de Campinas, Campinas, 13083-862, Brazil
| |
Collapse
|
41
|
Davoli R, Gaffo E, Zappaterra M, Bortoluzzi S, Zambonelli P. Identification of differentially expressed small RNAs and prediction of target genes in Italian Large White pigs with divergent backfat deposition. Anim Genet 2018; 49:205-214. [DOI: 10.1111/age.12646] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/10/2018] [Indexed: 01/21/2023]
Affiliation(s)
- R. Davoli
- Department of Agricultural and-Food Sciences (DISTAL); University of Bologna; Viale G. Fanin 46 40127 Bologna Italy
| | - E. Gaffo
- Department of Molecular Medicine; University of Padova; Viale G. Colombo 3 35131 Padova Italy
| | - M. Zappaterra
- Department of Agricultural and-Food Sciences (DISTAL); University of Bologna; Viale G. Fanin 46 40127 Bologna Italy
| | - S. Bortoluzzi
- Department of Molecular Medicine; University of Padova; Viale G. Colombo 3 35131 Padova Italy
| | - P. Zambonelli
- Department of Agricultural and-Food Sciences (DISTAL); University of Bologna; Viale G. Fanin 46 40127 Bologna Italy
| |
Collapse
|
42
|
Bao CX, Zhang DX, Wang NN, Zhu XK, Zhao Q, Sun XL. MicroRNA-335-5p suppresses lower extremity deep venous thrombosis by targeted inhibition of PAI-1 via the TLR4 signalingpathway. J Cell Biochem 2018; 119:4692-4710. [PMID: 29278662 DOI: 10.1002/jcb.26647] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 12/20/2017] [Indexed: 12/21/2022]
Abstract
This study aims to investigate the effects of microRNA-335-5p (miR-335-5p) on lower-extremity deep vein thrombosis (LEDVT) by targeting PAI-1 through the TLR4 signaling pathway in rat models. siRNA, mimic, and inhibitor were used for transfection. The miR-335-5p expression was detected by in situ hybridization. CCK-8 assay and flow cytometry were adopted to detect proliferation, cell cycle, and apoptosis, respectively. Scratch test and Matrigel-based tube formation assay were used to detect the effect of miR-335-5p on cell migration ability and tube formation ability. A miR-335-5p lentivirus plasmid was constructed and injected into LEDVT rats. The length and weight of thrombus were measured, changes of thrombus recanalization were observed by CD34 immunohistochemistry, and levels of PAI-1 and inflammatory factors in femoral vein blood were detected by ELISA. LEDVT rats showed a higher AOD value of PAI-1, higher expression of PAI-1, NF-κB, Rac1, IL-1β, and TLR4 and a lower miR-335-5p expression. PAI-1 and miR-335-5p were negatively correlated. Compared to the blank and siRNA-NC groups, the miR-335-5p mimic and siRNA-PAI-1 groups showed declined expression of PAI-1, TLR4, NF-κB, Rac1, and IL-1β, increased proliferation and tube formation abilities, less cells in G0/G1 phase, and decreased apoptosis, decreased length and weight of thrombus, organized thrombus, increased new blood vessels, and decreased levels of PAI-1, IL-1, IL-6, and Tnf-a. miR-335-5p may suppress the occurrence and development of LEDVT in rats by repressing the activation of the TLR4 signaling pathway by targeted inhibition of PAI-1.
Collapse
Affiliation(s)
- Cui-Xia Bao
- Clinical Laboratory, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, P. R. China
| | - Dong-Xia Zhang
- Department of Cardiovascular Medicine, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, P. R. China
| | - Na-Na Wang
- Clinical Laboratory, Yantai Yeda Hospital, Yantai, P. R. China
| | - Xiang-Kui Zhu
- Department of Radiology, Yantai Stomatological Hospital, Yantai, P. R. China
| | - Qi Zhao
- Clinical Laboratory, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, P. R. China
| | - Xiao-Lei Sun
- Department of Cardiology, Yantai Hospital of Traditional Chinese Medicine, Yantai, P. R. China
| |
Collapse
|
43
|
Wijayatunga NN, Pahlavani M, Kalupahana NS, Kottapalli KR, Gunaratne PH, Coarfa C, Ramalingam L, Moustaid-Moussa N. An integrative transcriptomic approach to identify depot differences in genes and microRNAs in adipose tissues from high fat fed mice. Oncotarget 2018; 9:9246-9261. [PMID: 29507687 PMCID: PMC5823660 DOI: 10.18632/oncotarget.24226] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 01/01/2018] [Indexed: 01/08/2023] Open
Abstract
Obesity contributes to metabolic disorders such as diabetes and cardiovascular disease. Characterization of differences between the main adipose tissue depots, white (WAT) [including subcutaneous (SAT) and visceral adipose tissue (VAT)] and brown adipose tissue (BAT) helps to identify their roles in obesity. Thus, we studied depot-specific differences in whole transcriptome and miRNA profiles of SAT, VAT and BAT from high fat diet (HFD/45% of calories from fat) fed mice using RNA sequencing and small RNA-Seq. Using quantitative real-time polymerase chain reaction, we validated depot-specific differences in endoplasmic reticulum (ER) stress related genes and miRNAs using mice fed a HFD vs. low fat diet (LFD/10% of calories from fat). According to the transcriptomic analysis, lipogenesis, adipogenesis, inflammation, endoplasmic reticulum (ER) stress and unfolded protein response (UPR) were higher in VAT compared to BAT, whereas energy expenditure, fatty acid oxidation and oxidative phosphorylation were higher in BAT than in VAT of the HFD fed mice. In contrast to BAT, ER stress marker genes were significantly upregulated in VAT of HFD fed mice than the LFD fed mice. For the first time, we report depot specific differences in ER stress related miRNAs including; downregulation of miR-125b-5p, upregulation miR-143-3p, and miR-222-3p in VAT following HFD and upregulation of miR-30c-2-3p only in BAT following a HFD in mice than the LFD mice. In conclusion, HFD differentially regulates miRNAs and genes in different adipose depots with significant induction of genes related to lipogenesis, adipogenesis, inflammation, ER stress, and UPR in WAT compared to BAT.
Collapse
Affiliation(s)
- Nadeeja N. Wijayatunga
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA
- Obesity Research Cluster, Texas Tech University, Lubbock, TX, USA
| | - Mandana Pahlavani
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA
- Obesity Research Cluster, Texas Tech University, Lubbock, TX, USA
| | - Nishan S. Kalupahana
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA
- Obesity Research Cluster, Texas Tech University, Lubbock, TX, USA
- Department of Physiology, University of Peradeniya, Peradeniya, Sri Lanka
| | - Kameswara Rao Kottapalli
- Obesity Research Cluster, Texas Tech University, Lubbock, TX, USA
- Center for Biotechnology and Genomics, Texas Tech University, Lubbock, TX, USA
| | | | - Cristian Coarfa
- Department of Molecular and Cell Biology, Baylor College of Medicine Hospital, Houston, TX, USA
| | - Latha Ramalingam
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA
- Obesity Research Cluster, Texas Tech University, Lubbock, TX, USA
| | - Naima Moustaid-Moussa
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA
- Obesity Research Cluster, Texas Tech University, Lubbock, TX, USA
| |
Collapse
|
44
|
Shi C, Huang F, Gu X, Zhang M, Wen J, Wang X, You L, Cui X, Ji C, Guo X. Adipogenic miRNA and meta-signature miRNAs involved in human adipocyte differentiation and obesity. Oncotarget 2018; 7:40830-40845. [PMID: 27049726 PMCID: PMC5130048 DOI: 10.18632/oncotarget.8518] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Accepted: 03/28/2016] [Indexed: 01/08/2023] Open
Abstract
MicroRNAs (miRNAs) have been identified as a new class of regulatory molecules that influence many biological functions, including metabolism, adipocyte differentiation. To determine the role of adipogenic miRNAs in the adipocyte differentiation process, we used microarray technology to monitor miRNA levels in human adipose-derived mesenchymal stem cells (hMSCs-Ad), human stromal vascular cells (SVCs) and differentiated adipocytes. 79 miRNAs were found to be differentially expressed, most of which are located in obesity related chromosomal regions but have not been previously linked to adipocyte differentiation process. A systematic search was made for relevant studies in academic data bases, involving the Gene Expression Omnibus (GEO) ArrayExpress, Pubmed and Embase database. Eight studies on human adipocyte differentiation or obesity were included in the final analysis. After combining our microarray data with meta-analysis of published microarray data, we detected 42 differently expressed miRNAs (meta-signature miRNAs) in mature adipocytes compared to SVCs or hMSCs-Ad. Our study shows meta-signature miRNAs specific for adipogenesis, several of which are correlated with key gene targets demonstrating functional relationships to pathways in BMP signaling pathway, Cell differentiation, Wnt signaling, insulin receptor signaling pathway, MAPK signaling, Cell cycle and lipid metabolic process. Our study shows that the first evidence of hsa-let-7 family, hsa-miR-15a-5p, hsa-miR-27a-3p, hsa-miR-106b-5p, hsa-miR-148a-3p and hsa-miR-26b-5p got a great weight in adipogenesis. We concluded that meta-signature miRNAs involved in adipocyte differentiation and provided pathophysiological roles and novel insight into obesity and its related metabolic diseases.
Collapse
Affiliation(s)
- Chunmei Shi
- Department of Children Health Care, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing, China.,Institute of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Fangyan Huang
- Department of Children Health Care, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing, China.,Institute of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Xiaohong Gu
- Department of Children Health Care, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing, China.,Institute of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Min Zhang
- Department of Children Health Care, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Juan Wen
- Department of Children Health Care, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Xing Wang
- Department of Children Health Care, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Lianghui You
- Department of Children Health Care, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Xianwei Cui
- Department of Children Health Care, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Chenbo Ji
- Department of Children Health Care, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing, China.,Institute of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Xirong Guo
- Department of Children Health Care, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing, China.,Institute of Pediatrics, Nanjing Medical University, Nanjing, China
| |
Collapse
|
45
|
MicroRNAs and adipocytokines: Promising biomarkers for pharmacological targets in diabetes mellitus and its complications. Biomed Pharmacother 2017; 93:1326-1336. [DOI: 10.1016/j.biopha.2017.07.059] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 07/08/2017] [Accepted: 07/11/2017] [Indexed: 02/06/2023] Open
|
46
|
Zhou YF, Fu ZY, Chen XH, Cui Y, Ji CB, Guo XR. Tumor necrosis factor‑α and interleukin‑6 suppress microRNA‑1275 transcription in human adipocytes through nuclear factor‑κB. Mol Med Rep 2017; 16:5965-5971. [PMID: 28901460 PMCID: PMC5865775 DOI: 10.3892/mmr.2017.7392] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2016] [Accepted: 06/16/2017] [Indexed: 12/30/2022] Open
Abstract
Obesity is a confirmed risk factor for hyperlipidemia, type-II diabetes, hypertension, and cardiovascular disease. MicroRNAs (miRs) have emerged as an important field of study within energy metabolism and obesity. A previous study demonstrated miR-1275 to be markedly down-regulated during maturation of human preadipocytes. It has been reported that miR-1275 dysregulates expression in several types of cancer and infections. Little is currently known about the regulation of miR-1275 transcription. The aim of the current study was to explore the mechanism underlying the expression of miR-1275 in mature human adipocytes. After differentiation, human adipocytes were incubated with tumor necrosis factor (TNF)-α and interleukin-6. The results of reverse transcription-quantitative polymerase chain reaction demonstrated that miR-1275 can be down-regulated by TNF-α and IL-6, in human mature adipocytes. Bioinformatic analysis was used to predict nuclear factor (NF)-κB binding sites of miR-1275′s promoter region. Luciferase assay and rescue experiments were performed in HEK293T cells. NF-κB was involved in regulating miR-1275 transcription by binding to its promoter. In response to TNF-α, NF-κB was bound to the promoter of miR-1275 and inhibited its transcription. These results indicated that inflammatory factors could regulate miR-1275 transcription through NF-κB and influencing miR-1275 effects on obesity.
Collapse
Affiliation(s)
- Yu-Feng Zhou
- School of Nursing, Nanjing Medical University, Nanjing, Jiangsu 210004, P.R. China
| | - Zi-Yi Fu
- Department of Children Health Care, Nanjing Maternal and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210004, P.R. China
| | - Xiao-Hui Chen
- Department of Children Health Care, Nanjing Maternal and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210004, P.R. China
| | - Yan Cui
- School of Nursing, Nanjing Medical University, Nanjing, Jiangsu 210004, P.R. China
| | - Chen-Bo Ji
- Department of Children Health Care, Nanjing Maternal and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210004, P.R. China
| | - Xi-Rong Guo
- Department of Children Health Care, Nanjing Maternal and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210004, P.R. China
| |
Collapse
|
47
|
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
|
48
|
Oldenburg A, Briand N, Sørensen AL, Cahyani I, Shah A, Moskaug JØ, Collas P. A lipodystrophy-causing lamin A mutant alters conformation and epigenetic regulation of the anti-adipogenic MIR335 locus. J Cell Biol 2017; 216:2731-2743. [PMID: 28751304 PMCID: PMC5584164 DOI: 10.1083/jcb.201701043] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 05/04/2017] [Accepted: 06/20/2017] [Indexed: 12/30/2022] Open
Abstract
Mutations in the Lamin A/C (LMNA) gene-encoding nuclear LMNA cause laminopathies, which include partial lipodystrophies associated with metabolic syndromes. The lipodystrophy-associated LMNA p.R482W mutation is known to impair adipogenic differentiation, but the mechanisms involved are unclear. We show in this study that the lamin A p.R482W hot spot mutation prevents adipogenic gene expression by epigenetically deregulating long-range enhancers of the anti-adipogenic MIR335 microRNA gene in human adipocyte progenitor cells. The R482W mutation results in a loss of function of differentiation-dependent lamin A binding to the MIR335 locus. This impairs H3K27 methylation and instead favors H3K27 acetylation on MIR335 enhancers. The lamin A mutation further promotes spatial clustering of MIR335 enhancer and promoter elements along with overexpression of the MIR355 gene after adipogenic induction. Our results link a laminopathy-causing lamin A mutation to an unsuspected deregulation of chromatin states and spatial conformation of an miRNA locus critical for adipose progenitor cell fate.
Collapse
Affiliation(s)
- Anja Oldenburg
- Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Nolwenn Briand
- Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Anita L Sørensen
- Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Inswasti Cahyani
- Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Akshay Shah
- Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Jan Øivind Moskaug
- Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway.,Norwegian Center for Stem Cell Research, Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Philippe Collas
- Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway .,Norwegian Center for Stem Cell Research, Department of Immunology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
49
|
Anunciado-Koza RP, Manuel J, Mynatt RL, Zhang J, Kozak LP, Koza RA. Diet-induced adipose tissue expansion is mitigated in mice with a targeted inactivation of mesoderm specific transcript (Mest). PLoS One 2017. [PMID: 28640866 PMCID: PMC5481029 DOI: 10.1371/journal.pone.0179879] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Interindividual variation of white adipose tissue (WAT) expression of mesoderm specific transcript (Mest), a paternally-expressed imprinted gene belonging to the α/β-hydrolase fold protein family, becomes apparent among genetically inbred mice fed high fat diet (HFD) and is positively associated with adipose tissue expansion (ATE). To elucidate a role for MEST in ATE, mice were developed with global and adipose tissue inactivation of Mest. Mice with homozygous (MestgKO) and paternal allelic (MestpKO) inactivation of Mest were born at expected Mendelian frequencies, showed no behavioral or physical abnormalities, and did not perturb expression of the Mest locus-derived microRNA miR-335. MestpKO mice fed HFD showed reduced ATE and adipocyte hypertrophy, improved glucose tolerance, and reduced WAT expression of genes associated with hypoxia and inflammation compared to littermate controls. Remarkably, caloric intake and energy expenditure were unchanged between genotypes. Mice with adipose tissue inactivation of Mest were phenotypically similar to MestpKO, supporting a role for WAT MEST in ATE. Global profiling of WAT gene expression of HFD-fed control and MestpKO mice detected few differences between genotypes; nevertheless, genes with reduced expression in MestpKO mice were associated with immune processes and consistent with improved glucose homeostasis. Ear-derived mesenchymal stem cells (EMSC) from MestgKO mice showed no differences in adipogenic differentiation compared to control cells unless challenged by shRNA knockdown of Gpat4, an enzyme that mediates lipid accumulation in adipocytes. Reduced adipogenic capacity of EMSC from MestgKO after Gpat4 knockdown suggests that MEST facilitates lipid accumulation in adipocytes. Our data suggests that reduced diet-induced ATE in MEST-deficient mice diminishes hypoxia and inflammation in WAT leading to improved glucose tolerance and insulin sensitivity. Since inactivation of Mest in mice has minimal additional effects aside from reduction of ATE, an intervention that mitigates MEST function in adipocytes is a plausible strategy to obviate obesity and type-2-diabetes.
Collapse
Affiliation(s)
- Rea P. Anunciado-Koza
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, Maine, United States of America
| | - Justin Manuel
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, Maine, United States of America
| | - Randall L. Mynatt
- Transgenics Core Facility, Pennington Biomedical Research Center, LSU System, Baton Rouge, Louisiana, United States of America
| | - Jingying Zhang
- Transgenics Core Facility, Pennington Biomedical Research Center, LSU System, Baton Rouge, Louisiana, United States of America
| | - Leslie P. Kozak
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Robert A. Koza
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, Maine, United States of America
- * E-mail:
| |
Collapse
|
50
|
Yue J, Wang P, Hong Q, Liao Q, Yan L, Xu W, Chen X, Zheng Q, Zhang L, Huang D. MicroRNA-335-5p Plays Dual Roles in Periapical Lesions by Complex Regulation Pathways. J Endod 2017; 43:1323-1328. [PMID: 28578884 DOI: 10.1016/j.joen.2017.03.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 02/02/2017] [Accepted: 03/12/2017] [Indexed: 02/05/2023]
Abstract
INTRODUCTION MicroRNA-335-5p has been reported to regulate osteogenic and chondrogenic differentiations of mesenchymal stem cells. The aim of this study was to explore the function and regulation mechanism of miR-335-5p in apical periodontitis (AP). METHODS Total RNAs were extracted from human periodontal ligament fibroblasts (HPDLFs), 10 AP tissues, and 6 healthy periodontal ligament tissues using lysis buffer. Gene expression was detected using real-time polymerase chain reaction. The Dual Luciferase Assay (Promega, Madison, WI) was used to test miR-335-5p directly targeted urokinase-type plasminogen activator receptor (uPAR) and the receptor activator of nuclear factor kappa-B ligand (RANKL). Western Blot was used to detect protein expressions of RANKL, uPAR, and the fragile X-related 1 gene (FXR1). The enzyme-linked immunosorbent assay was used to detect the secretions of interleukin 6, tumor necrosis factor alpha, and RANKL. Data were analyzed using the Student t test. RESULTS miR-335-5p acted as a positive mediator in HPDLF inflammation (P < .05). Two targets of miR-335-5p, uPAR and RANKL, were identified. Interestingly, uPAR was repressed by miR-335-5p at the basal level, but it can be relieved from miR-335-5p-mediated repression, which is called derepression, when HPDLFs were subjected to lipopolysaccharide stimulation. miR-335-5p promoted RANKL in HPDLFs regardless of whether or not it was under inflammatory conditions (P < .05). We proved FXR1 was responsible for the derepression of uPAR from miR-335-5p (P < .01). Both FXR1 and uPAR were positive mediators in HPDLF inflammation (P < .05). miR-335-5p, uPAR, RANKL, and FXR1 had the same expression profiles in HPDLF inflammation and AP tissues (P < .05). CONCLUSIONS Our data showed that miR-335-5p may play dual roles in AP, and it might be considered as a target for therapeutic potency in clinical applications.
Collapse
Affiliation(s)
- Junli Yue
- State Key Laboratory of Oral Diseases, Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, China
| | - Puyu Wang
- State Key Laboratory of Oral Diseases, Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, China
| | - Qingchun Hong
- State Key Laboratory of Oral Diseases, Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, China
| | - Qian Liao
- State Key Laboratory of Oral Diseases, Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, China
| | - Li Yan
- State Key Laboratory of Oral Diseases, Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, China
| | - Weizhe Xu
- State Key Laboratory of Oral Diseases, Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, China
| | - Xi Chen
- State Key Laboratory of Oral Diseases, Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, China
| | - Qinghua Zheng
- State Key Laboratory of Oral Diseases, Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, China
| | - Lan Zhang
- State Key Laboratory of Oral Diseases, Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, China.
| | - Dingming Huang
- State Key Laboratory of Oral Diseases, Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, China.
| |
Collapse
|