1
|
Huang W, Wu X, Xiang S, Qiao M, Li H, Zhu Y, Zhu Z, Zhao Z. Regulatory of miRNAs in tri-lineage differentiation of C3H10T1/2. Stem Cell Res Ther 2022; 13:521. [PMID: 36414991 PMCID: PMC9682817 DOI: 10.1186/s13287-022-03205-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 10/28/2022] [Indexed: 11/24/2022] Open
Abstract
MicroRNAs (miRNAs) are non-coding single-stranded RNA molecules encoded by endogenous genes, which play a vital role in cell generation, metabolism, apoptosis and stem cell differentiation. C3H10T1/2, a mesenchymal cell extracted from mouse embryos, is capable of osteogenic differentiation, adipogenic differentiation and chondrogenic differentiation. Extensive studies have shown that not only miRNAs can directly trigger targeted genes to regulate the tri-lineage differentiation of C3H10T1/2, but it also can indirectly regulate the differentiation by triggering different signaling pathways or various downstream molecules. This paper aims to clarify the regulatory roles of different miRNAs on C3H10T1/2 differentiation, and discussing their balance effect among osteogenic differentiation, adipogenic differentiation and chondrogenic differentiation of C3H10T1/2. We also review the biogenesis of miRNAs, Wnt signaling pathways, MAPK signaling pathways and BMP signaling pathways and provide some specific examples of how these signaling pathways act on C3H10T1/2 tri-lineage differentiation. On this basis, we hope that a deeper understanding of the differentiation and regulation mechanism of miRNAs in C3H10T1/2 can provide a promising therapeutic method for the clinical treatment of bone defects, osteoporosis, osteoarthritis and other diseases.
Collapse
Affiliation(s)
- Wei Huang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Xiaoyue Wu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Shuaixi Xiang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Mingxin Qiao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Hanfei Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Yujie Zhu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Zhou Zhu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, People's Republic of China.
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, People's Republic of China.
| |
Collapse
|
2
|
Yan Y, Yuan J, Luo X, Yu X, Lu J, Hou W, He X, Zhang L, Cao J, Wang H. microRNA-140 Regulates PDGFRα and Is Involved in Adipocyte Differentiation. Front Mol Biosci 2022; 9:907148. [PMID: 35832736 PMCID: PMC9271708 DOI: 10.3389/fmolb.2022.907148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 05/16/2022] [Indexed: 12/23/2022] Open
Abstract
In recent years, the studies of the role of microRNAs in adipogenesis and adipocyte development and the corresponding molecular mechanisms have received great attention. In this work, we investigated the function of miR-140 in the process of adipogenesis and the molecular pathways involved, and we found that adipogenic treatment promoted the miR-140-5p RNA level in preadipocytes. Over-expression of miR-140-5p in preadipocytes accelerated lipogenesis along with adipogenic differentiation by transcriptional modulation of adipogenesis-linked genes. Meanwhile, silencing endogenous miR-140-5p dampened adipogenesis. Platelet-derived growth factor receptor alpha (PDGFRα) was shown to be a miR-140-5p target gene. miR-140-5p over-expression in preadipocyte 3T3-L1 diminished PDGFRα expression, but silencing of miR-140-5p augmented it. In addition, over-expression of PDGFRα suppressed adipogenic differentiation and lipogenesis, while its knockdown enhanced these biological processes of preadipocyte 3T3-L1. Altogether, our current findings reveal that miR-140-5p induces lipogenesis and adipogenic differentiation in 3T3-L1 cells by targeting PDGFRα, therefore regulating adipogenesis. Our research provides molecular targets and a theoretical basis for the treatment of obesity-related metabolic diseases.
Collapse
Affiliation(s)
- Yi Yan
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - Jiahui Yuan
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - Xiaomao Luo
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - Xiuju Yu
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - Jiayin Lu
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - Wei Hou
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - Xiaoyan He
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - Liping Zhang
- Department of Medicine, Nephrology Division, Baylor College of Medicine, Houston, GA, United States
| | - Jing Cao
- Department of Animal Husbandry and Veterinary Medicine, Beijing Vocational College of Agriculture, Beijing, China
| | - Haidong Wang
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
- *Correspondence: Haidong Wang,
| |
Collapse
|
3
|
Meruvu S, Zhang J, Choudhury M. Butyl Benzyl Phthalate Promotes Adipogenesis in 3T3-L1 Cells via the miRNA-34a-5p Signaling Pathway in the Absence of Exogenous Adipogenic Stimuli. Chem Res Toxicol 2021; 34:2251-2260. [PMID: 34520170 DOI: 10.1021/acs.chemrestox.1c00115] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Phthalates, a plasticizer group, are used extensively in many of the products we use every day. Public health concerns are growing as recent studies have implicated butyl benzyl phthalate (BBP) as an obesogen. However, BBP-induced epigenetic regulation during adipogenesis is still unknown. We investigated if BBP altered miR-34a-5p, a key miRNA involved in obesity, and regulated its downstream pathway. Differentiating 3T3-L1 cells were exposed to various doses of BBP without exogenous adipogenic stimuli, tested for adipogenesis markers (PPARγ and aP2), and stained for lipid accumulation with Oil Red O staining. We then measured the expression of miR-34a-5p and its target genes, Nampt and Sirt1, along with another significant epigenetic modulator, Sirt3. Furthermore, using antagomiR, we examined whether miR-34a-5p knockdown decreased adipogenesis. BBP exposure resulted in augmented expression levels of miR-34a-5p with an associated increase in adipogenesis. BBP significantly decreased the Nampt, Sirt1, and Sirt3 gene expression levels. However, a decrease in the protein expression was observed only for Nampt, indicating that miR-34a-5p under BBP exposure may regulate Sirt1/Sirt3 only at the transcriptional level. Interestingly, in the presence of BBP, knockdown of miR-34a-5p decreased adipogenesis in the differentiating 3T3-L1 cells. Furthermore, miR-34a-5p knockdown increased the Nampt protein expression levels as well as NAD+ levels, indicating that miR-34a-5p regulates Nampt during BBP exposure. Additionally, the NAD+-dependent sirtuin activity decreased in BBP-treated cells and increased in miR-34a-5p knockdown cells with BBP treatment. BBP exposure demonstrated the involvement of epigenetic regulation by altering the expression patterns of miR-34a-5p and its target Nampt, which may perturb the energy homeostasis of the differentiating adipocytes by altering NAD+ levels and sirtuin activity, resulting in increased adipogenesis.
Collapse
Affiliation(s)
- Sunitha Meruvu
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M Health Science Center, College Station, Texas 77843, United States
| | - Jian Zhang
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M Health Science Center, College Station, Texas 77843, United States
| | - Mahua Choudhury
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M Health Science Center, College Station, Texas 77843, United States
| |
Collapse
|
4
|
Gao T, Ma D, Chen S, Zhang X, Han Y, Liu M. Acupuncture for the treatment of leptin resistance in obesity: A protocol for systematic review and meta-analysis. Medicine (Baltimore) 2021; 100:e26244. [PMID: 34260523 PMCID: PMC8284727 DOI: 10.1097/md.0000000000026244] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 05/19/2021] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Recently, there has been a global increase in obesity and obesity-related diseases. The prevention and treatment of obesity have become one of the most significant public health challenges worldwide in the 21st century, and how to effectively curb the occurrence of obesity has become a major global concern. Numerous studies have shown that the majority of obese individuals do not respond to leptin, and instead demonstrate leptin resistance. Clinical studies have found that acupuncture is widely used in the clinical treatment of obesity in recent years, but whether it can improve leptin resistance has not been systematically reviewed. This study is aimed to investigate the effectiveness of acupuncture in obesity with leptin resistance (LR). METHODS We searched PubMed, Cochrane Library, Embase, China National Knowledge Infrastructure (CNKI), Wan Fang, Technology Journal Database (VIP), and China Biology Medicine disc (CBM). Chinese Clinical Trial Registry. The time was from the establishment of the database to March 19, 2021. RevMan 5.3 software was used to assess the quality and risk of the included studies. RESULTS This study will be conducted in terms of clinical efficacy, serum leptin content, and body weight change. The current evidence shows that the incidence of the disease is high and the comprehensive quality is high. CONCLUSION The conclusion of this review will provide a basis for judging whether acupuncture therapy is effective in the treatment of leptin resistance in obesity.
Collapse
Affiliation(s)
| | - Dehui Ma
- Changchun University of Chinese Medicine
| | - Shaotao Chen
- Changchun University of Chinese Medicine
- Acupuncture and Massage Center of the Third Affiliated Clinical Hospital of Changchun University of Chinese Medicine, Changchun, China
| | | | - Yiran Han
- Changchun University of Chinese Medicine
| | - Mingjun Liu
- Changchun University of Chinese Medicine
- Acupuncture and Massage Center of the Third Affiliated Clinical Hospital of Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
5
|
Osteopetrosis-Associated Transmembrane Protein 1 Recruits RNA Exosome To Restrict Hepatitis B Virus Replication. J Virol 2020; 94:JVI.01800-19. [PMID: 32188736 DOI: 10.1128/jvi.01800-19] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 01/22/2020] [Indexed: 02/07/2023] Open
Abstract
Hepatitis B virus (HBV) chronically infects approximately 350 million people worldwide, and 600,000 deaths are caused by HBV-related hepatic failure, liver cirrhosis, and hepatocellular carcinoma annually. It is important to reveal the mechanism underlying the regulation of HBV replication. This study demonstrated that osteopetrosis-associated transmembrane protein 1 (Ostm1) plays an inhibitory role in HBV replication. Ostm1 represses the levels of HBeAg and HBsAg proteins, HBV 3.5-kb and 2.4/2.1-kb RNAs, and core-associated DNA in HepG2, Huh7, and NTCP-HepG2 cells. Notably, Ostm1 has no direct effect on the activity of HBV promoters or the transcription of HBV RNAs; instead, Ostm1 binds to HBV RNA to facilitate RNA decay. Detailed studies further demonstrated that Ostm1 binds to and recruits the RNA exosome complex to promote the degradation of HBV RNAs, and knockdown of the RNA exosome component exonuclease 3 (Exosc3) leads to the elimination of Ostm1-mediated repression of HBV replication. Mutant analyses revealed that the N-terminal domain, the transmembrane domain, and the C-terminal domain are responsible for the repression of HBV replication, and the C-terminal domain is required for interaction with the RNA exosome complex. Moreover, Ostm1 production is not regulated by interferon-α (IFN-α) or IFN-γ, and the expression of IFN signaling components is not affected by Ostm1, suggesting that Ostm1 anti-HBV activity is independent of the IFN signaling pathway. In conclusion, this study revealed a distinct mechanism underlying the repression of HBV replication, in which Ostm1 binds to HBV RNA and recruits RNA exosomes to degrade viral RNA, thereby restricting HBV replication.IMPORTANCE Hepatitis B virus (HBV) is a human pathogen infecting the liver to cause a variety of diseases ranging from acute hepatitis to advanced liver diseases, fulminate hepatitis, liver cirrhosis, and hepatocellular carcinoma, thereby causing a major health problem worldwide. In this study, we demonstrated that Ostm1 plays an inhibitory role in HBV protein production, RNA expression, and DNA replication. However, Ostm1 has no effect on the activities of the four HBV promoters; instead, it binds to HBV RNA and recruits RNA exosomes to promote HBV RNA degradation. We further demonstrated that the anti-HBV activity of Ostm1 is independent of the interferon signaling pathway. In conclusion, this study reveals a distinct mechanism underlying the repression of HBV replication and suggests that Ostm1 is a potential therapeutic agent for HBV infection.
Collapse
|
6
|
Zhang Z, Gao Y, Xu MQ, Wang CJ, Fu XH, Liu JB, Han DX, Jiang H, Yuan B, Zhang JB. miR-181a regulate porcine preadipocyte differentiation by targeting TGFBR1. Gene 2019; 681:45-51. [DOI: 10.1016/j.gene.2018.09.046] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 09/20/2018] [Accepted: 09/24/2018] [Indexed: 12/12/2022]
|
7
|
Lo PK, Wolfson B, Zhou Q. Adipogenesis and Noncoding RNAs. HANDBOOK OF NUTRITION, DIET, AND EPIGENETICS 2019:623-645. [DOI: 10.1007/978-3-319-55530-0_41] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
8
|
Sun Y, Kuek V, Liu Y, Tickner J, Yuan Y, Chen L, Zeng Z, Shao M, He W, Xu J. MiR-214 is an important regulator of the musculoskeletal metabolism and disease. J Cell Physiol 2018; 234:231-245. [PMID: 30076721 DOI: 10.1002/jcp.26856] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 05/10/2018] [Indexed: 12/21/2022]
Abstract
MiR-214 belongs to a family of microRNA (small, highly conserved noncoding RNA molecules) precursors that play a pivotal role in biological functions, such as cellular function, tissue development, tissue homeostasis, and pathogenesis of diseases. Recently, miR-214 emerged as a critical regulator of musculoskeletal metabolism. Specifically, miR-214 can mediate skeletal muscle myogenesis and vascular smooth muscle cell proliferation, migration, and differentiation. MiR-214 also modulates osteoblast function by targeting specific molecular pathways and the expression of various osteoblast-related genes; promotes osteoclast activity by targeting phosphatase and tensin homolog (Pten); and mediates osteoclast-osteoblast intercellular crosstalk via an exosomal miRNA paracrine mechanism. Importantly, dysregulation in miR-214 expression is associated with pathological bone conditions such as osteoporosis, osteosarcoma, multiple myeloma, and osteolytic bone metastasis of breast cancer. This review discusses the cellular targets of miR-214 in bone, the molecular mechanisms governing the activities of miR-214 in the musculoskeletal system, and the putative role of miR-214 in skeletal diseases. Understanding the biology of miR-214 could potentially lead to the development of miR-214 as a possible biomarker and a therapeutic target for musculoskeletal diseases.
Collapse
Affiliation(s)
- Youqiang Sun
- The Department of Orthopedics, First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.,Division of Pathology and Laboratory Medicine, School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia.,The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Vincent Kuek
- Division of Pathology and Laboratory Medicine, School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
| | - Yuhao Liu
- The Department of Orthopedics, First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.,Division of Pathology and Laboratory Medicine, School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia.,The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jennifer Tickner
- Division of Pathology and Laboratory Medicine, School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
| | - Yu Yuan
- School of Physical Education and Sports Science, South China Normal University, Guangzhou, Guangdong, China
| | - Leilei Chen
- The Department of Orthopedics, First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.,The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Zhikui Zeng
- The Department of Orthopedics, First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.,The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Min Shao
- The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.,Department of Orthopedics, Third Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Wei He
- The Department of Orthopedics, First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.,The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jiake Xu
- Division of Pathology and Laboratory Medicine, School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia.,The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
9
|
MicroRNA-140 Suppresses Human Chondrocytes Hypertrophy by Targeting SMAD1 and Controlling the Bone Morphogenetic Protein Pathway in Osteoarthritis. Am J Med Sci 2018; 355:477-487. [DOI: 10.1016/j.amjms.2018.01.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 01/06/2018] [Accepted: 01/18/2018] [Indexed: 12/19/2022]
|
10
|
Integrated analysis of mRNA and miRNA expression profiles in livers of Yimeng black pigs with extreme phenotypes for backfat thickness. Oncotarget 2017; 8:114787-114800. [PMID: 29383120 PMCID: PMC5777732 DOI: 10.18632/oncotarget.21918] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 09/21/2017] [Indexed: 12/20/2022] Open
Abstract
Fat deposition is an important economic trait in farm animals as well as obesity related diseases in humans, and the liver is a central organ involved in regulating lipid synthesis and metabolism in mammals. In this study, the pig liver transcriptome of two groups (H and L) showing differences in backfat thickness were profiled using RNA-Seq and miRNA-Seq to further explore the molecular mechanism of fat deposition. A total of 238 differentially expressed genes (DEGs) and 58 differentially expressed miRNAs were identified between the H and L group. These genes and miRNAs were functionally related to lipid metabolism, including CYP1A1/2, HMGCS2, ACSS2, UBE2L6, miR-27a, and miR-31. Functional enrichment analysis revealed that genes associated with oxidative stress might be responsible for fat deposition in pigs. Two miRNA-mRNA interaction networks involved in lipid metabolism were identified, and these provided new insights into the molecular regulation that determines fat content in these pigs. Overall, our study furthers our understanding of the molecular mechanisms involved in fat deposition, and these results may help in the design of selection strategies to improve the quality of pork meat and to combat obesity in humans.
Collapse
|
11
|
Subcutaneous adipose tissue gene expression and DNA methylation respond to both short- and long-term weight loss. Int J Obes (Lond) 2017; 42:412-423. [PMID: 28978976 DOI: 10.1038/ijo.2017.245] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 08/24/2017] [Accepted: 09/26/2017] [Indexed: 12/16/2022]
Abstract
BACKGROUND Few studies have examined both gene expression and DNA methylation profiles in subcutaneous adipose tissue (SAT) during long-term weight loss. Thus, molecular mechanisms in weight loss and regain remain elusive. PARTICIPANTS/METHODS We performed a 1-year weight loss intervention on 19 healthy obese participants (mean body mass index (BMI) 34.6 kg m-2) and studied longitudinal gene expression (Affymetrix Human Genome U133 Plus 2.0) and DNA methylation (Infinium HumanMethylation450 BeadChip) in SAT at 0, 5 and 12 months. To examine whether weight loss and acquired obesity produce reciprocal profiles, we verified our findings in 26 BMI-discordant monozygotic twin pairs. RESULTS We found altered expression of 69 genes from 0 to 5' months (short-term) weight loss. Sixty of these genes showed reversed expression in acquired obesity (twins). Altogether 21/69 genes showed significant expression-DNA methylation correlations. Pathway analyses revealed increased high-density lipoprotein-mediated lipid transport characteristic to short-term weight loss. After the fifth month, two groups of participants evolved: weight losers (WLs) and weight regainers (WRs). In WLs five genes were differentially expressed in 5 vs 12 months, three of which significantly correlated with methylation. Signaling by insulin receptor pathway showed increased expression. We further identified 35 genes with differential expression in WLs from 0 to 12 months (long-term) weight loss, with 20 showing opposite expression patterns in acquired obesity, and 16/35 genes with significant expression-DNA methylation correlations. Pathway analyses demonstrated changes in signal transduction, metabolism, immune system and cell cycle. Notably, seven genes (UCHL1, BAG3, TNMD, LEP, BHMT2, EPDR1 and OSTM1) were found to be downregulated during both short- and long-term weight loss. CONCLUSIONS Our study indicates short- and long-term weight loss influences in transcription and DNA methylation in SAT of healthy participants. Moreover, we demonstrate that same genes react in an opposite manner in weight loss and acquired obesity.
Collapse
|
12
|
Wa Q, Liu Y, Huang S, He P, Zuo J, Li X, Li Z, Dong L, Peng J, Wu S, Chen F, Cai D, Zou X, Liao W. miRNA-140 inhibits C3H10T1/2 mesenchymal stem cell proliferation by targeting CXCL12 during transforming growth factor-β3-induced chondrogenic differentiation. Mol Med Rep 2017; 16:1389-1394. [PMID: 29067438 DOI: 10.3892/mmr.2017.6720] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 03/10/2017] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to investigate the role of microRNA (miRNA or miR)-140 in C3H10T1/2 mesenchymal stem cells (MSCs). Cluster analysis was used to evaluate the miRNA expression profile. The expression level of miRNA‑140 was validated by reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR). TargetScan and microRNA.org databases were used to predict target miRNAs and cartilage‑associated target genes. Binding sites between miR‑140 and the target gene were predicted by bioinformatics software. A dual‑luciferase reporter assay was performed to determine whether miR‑140 could target C‑X‑C motif chemokine ligand 12 (CXCL12). Following the promotion/inhibition of miR‑140, 1, 7 and 14 days following transforming growth factor‑β3 (TGF‑β3)‑induction, western blotting was utilized to evaluate CXCL12 protein levels. MTT assays and alcian blue staining were applied to assess C3H10T1/2 MSC viability and chondrogenic differentiation, respectively. In the TGF‑β3‑induced group, RT‑qPCR verified that the mRNA level of Mus musculus (mmu)‑miR‑140 was significantly elevated when compared with the control group. miR‑140 was predicted to recognize and interact with CXCL12‑3'UTR and the dual luciferase reporter assay further validated that miR‑140 targeted the predicted region of CXCL12. CXCL12 was markedly decreased following miR‑140 overexpression and visibly increased following miR‑140 inhibition. In addition, the level of CXCL12 expression declined as the duration of induction increased. Following the promotion/inhibition of miR‑140, at 1 and 7 days following TGF‑β3‑induction, C3H10T1/2 MSCs inhibited or promoted cell viability, respectively, when compared with the control groups. In addition, in pellets achieved by chondrogenic differentiation following the induction of C3H10T1/2 MSCs for 7 days, alcian blue staining revealed no significant difference in characteristic extracellular matrix glycosaminoglycans between the miR‑140 up and downregulated groups, and their respective control groups. The present study concludes that miRNA‑140 inhibition promoted C3H10T1/2 MSC viability however, not C3H10T1/2 MSC differentiation by targeting and reducing CXCL12 protein levels during the process of TGF‑β3‑induced chondrogenic differentiation. In conclusion, the present study provided a potential target for the treatment of cartilage defection.
Collapse
Affiliation(s)
- Qingde Wa
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou 563003, P.R. China
| | - Yi Liu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou 563003, P.R. China
| | - Shuai Huang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Peiheng He
- Department of Orthopedic Surgery, The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Jianwei Zuo
- Department of Sports Medicine, Shenzhen Hospital of Peking University, Shenzhen, Guangdong 518036, P.R. China
| | - Xing Li
- Department of Orthopedic Surgery, The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Ziqing Li
- Department of Orthopedic Surgery, The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Liming Dong
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou 563003, P.R. China
| | - Jiachen Peng
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou 563003, P.R. China
| | - Shuhong Wu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou 563003, P.R. China
| | - Fang Chen
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou 563003, P.R. China
| | - Dongfeng Cai
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou 563003, P.R. China
| | - Xuenong Zou
- Department of Orthopedic Surgery, The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Wenbo Liao
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou 563003, P.R. China
| |
Collapse
|
13
|
Gan S, Huang Z, Liu N, Su R, Xie G, Zhong B, Zhang K, Wang S, Hu X, Zhang J, Xiang S. MicroRNA-140-5p impairs zebrafish embryonic bone development via targeting BMP-2. FEBS Lett 2017; 590:1438-46. [PMID: 27130837 DOI: 10.1002/1873-3468.12190] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 04/14/2016] [Accepted: 04/18/2016] [Indexed: 12/21/2022]
Abstract
MicroRNA-140-5p (miRNA-140-5p) is important for embryonic bone development. In this study, we found that miRNA-140-5p and its binding site in the 3'UTR of bone morphogenetic protein 2 (BMP-2) are highly conserved among vertebrates, and miRNA-140-5p negatively regulates both zebrafish and human BMP-2 genes. Microinjection of miRNA-140-5p or BMP-2b morpholino into zebrafish embryos led to a similar phenotype, including shortened tails, curved trunks, and defects in cranial cartilage. Moreover, miRNA-140-5p injection induced zebrafish embryo malformation that could be significantly rescued by microinjection of BMP-2 mRNA. In conclusion, our results indicated that miRNA-140-5p regulates zebrafish embryonic bone development via targeting BMP-2.
Collapse
Affiliation(s)
- Shiquan Gan
- Key Laboratory of Protein Chemistry and Developmental Biology of State Education Ministry of China, Department of Biochemistry and Molecular Biology, College of Life Science, Hunan Normal University, Changsha, Hunan, China.,The Cooperative Innovation Center of Engineering and New Products for Developmental Biology of Hunan Province (20134486), Changsha, Hunan, China
| | - Zhaoqin Huang
- Key Laboratory of Protein Chemistry and Developmental Biology of State Education Ministry of China, Department of Biochemistry and Molecular Biology, College of Life Science, Hunan Normal University, Changsha, Hunan, China.,The Cooperative Innovation Center of Engineering and New Products for Developmental Biology of Hunan Province (20134486), Changsha, Hunan, China
| | - Ning Liu
- Key Laboratory of Protein Chemistry and Developmental Biology of State Education Ministry of China, Department of Biochemistry and Molecular Biology, College of Life Science, Hunan Normal University, Changsha, Hunan, China.,College of Medicine, Hunan Normal University, Changsha, Hunan, China
| | - Renxiang Su
- Key Laboratory of Protein Chemistry and Developmental Biology of State Education Ministry of China, Department of Biochemistry and Molecular Biology, College of Life Science, Hunan Normal University, Changsha, Hunan, China.,The Cooperative Innovation Center of Engineering and New Products for Developmental Biology of Hunan Province (20134486), Changsha, Hunan, China
| | - Guie Xie
- Key Laboratory of Protein Chemistry and Developmental Biology of State Education Ministry of China, Department of Biochemistry and Molecular Biology, College of Life Science, Hunan Normal University, Changsha, Hunan, China.,The Cooperative Innovation Center of Engineering and New Products for Developmental Biology of Hunan Province (20134486), Changsha, Hunan, China
| | - Beibei Zhong
- Key Laboratory of Protein Chemistry and Developmental Biology of State Education Ministry of China, Department of Biochemistry and Molecular Biology, College of Life Science, Hunan Normal University, Changsha, Hunan, China.,The Cooperative Innovation Center of Engineering and New Products for Developmental Biology of Hunan Province (20134486), Changsha, Hunan, China
| | - Kai Zhang
- Key Laboratory of Protein Chemistry and Developmental Biology of State Education Ministry of China, Department of Biochemistry and Molecular Biology, College of Life Science, Hunan Normal University, Changsha, Hunan, China.,The Cooperative Innovation Center of Engineering and New Products for Developmental Biology of Hunan Province (20134486), Changsha, Hunan, China
| | - Shang Wang
- Key Laboratory of Protein Chemistry and Developmental Biology of State Education Ministry of China, Department of Biochemistry and Molecular Biology, College of Life Science, Hunan Normal University, Changsha, Hunan, China.,The Cooperative Innovation Center of Engineering and New Products for Developmental Biology of Hunan Province (20134486), Changsha, Hunan, China
| | - Xiang Hu
- Key Laboratory of Protein Chemistry and Developmental Biology of State Education Ministry of China, Department of Biochemistry and Molecular Biology, College of Life Science, Hunan Normal University, Changsha, Hunan, China.,The Cooperative Innovation Center of Engineering and New Products for Developmental Biology of Hunan Province (20134486), Changsha, Hunan, China
| | - Jian Zhang
- Key Laboratory of Protein Chemistry and Developmental Biology of State Education Ministry of China, Department of Biochemistry and Molecular Biology, College of Life Science, Hunan Normal University, Changsha, Hunan, China.,The Cooperative Innovation Center of Engineering and New Products for Developmental Biology of Hunan Province (20134486), Changsha, Hunan, China
| | - Shuanglin Xiang
- Key Laboratory of Protein Chemistry and Developmental Biology of State Education Ministry of China, Department of Biochemistry and Molecular Biology, College of Life Science, Hunan Normal University, Changsha, Hunan, China.,The Cooperative Innovation Center of Engineering and New Products for Developmental Biology of Hunan Province (20134486), Changsha, Hunan, China
| |
Collapse
|
14
|
Seeliger C, Balmayor ER, van Griensven M. miRNAs Related to Skeletal Diseases. Stem Cells Dev 2016; 25:1261-81. [PMID: 27418331 DOI: 10.1089/scd.2016.0133] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
miRNAs as non-coding, short, double-stranded RNA segments are important for cellular biological functions, such as proliferation, differentiation, and apoptosis. miRNAs mainly contribute to the inhibition of important protein translations through their cleavage or direct repression of target messenger RNAs expressions. In the last decade, miRNAs got in the focus of interest with new publications on miRNAs in the context of different diseases. For many types of cancer or myocardial damage, typical signatures of local or systemically circulating miRNAs have already been described. However, little is known about miRNA expressions and their molecular effect in skeletal diseases. An overview of published studies reporting miRNAs detection linked with skeletal diseases was conducted. All regulated miRNAs were summarized and their molecular interactions were illustrated. This review summarizes the involvement and interaction of miRNAs in different skeletal diseases. Thereby, 59 miRNAs were described to be deregulated in tissue, cells, or in the circulation of osteoarthritis (OA), 23 miRNAs deregulated in osteoporosis, and 107 miRNAs deregulated in osteosarcoma (OS). The molecular influences of miRNAs regarding OA, osteoporosis, and OS were illustrated. Specific miRNA signatures for skeletal diseases are described in the literature. Some overlapped, but also unique ones for each disease exist. These miRNAs may present useful targets for the development of new therapeutic approaches and are candidates for diagnostic evaluations.
Collapse
Affiliation(s)
- Claudine Seeliger
- Experimental Trauma Surgery, Department of Trauma Surgery, Klinikum Rechts der Isar, Technical University of Munich , Munich, Germany
| | - Elizabeth R Balmayor
- Experimental Trauma Surgery, Department of Trauma Surgery, Klinikum Rechts der Isar, Technical University of Munich , Munich, Germany
| | - Martijn van Griensven
- Experimental Trauma Surgery, Department of Trauma Surgery, Klinikum Rechts der Isar, Technical University of Munich , Munich, Germany
| |
Collapse
|
15
|
Barrallo-Gimeno A, Gradogna A, Zanardi I, Pusch M, Estévez R. Regulatory-auxiliary subunits of CLC chloride channel-transport proteins. J Physiol 2016; 593:4111-27. [PMID: 25762128 DOI: 10.1113/jp270057] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 02/15/2015] [Indexed: 02/06/2023] Open
Abstract
The CLC family of chloride channels and transporters is composed by nine members, but only three of them, ClC-Ka/b, ClC-7 and ClC-2, have been found so far associated with auxiliary subunits. These CLC regulatory subunits are small proteins that present few common characteristics among them, both structurally and functionally, and their effects on the corresponding CLC protein are different. Barttin, a protein with two transmembrane domains, is essential for the membrane localization of ClC-K proteins and their activity in the kidney and inner ear. Ostm1 is a protein with a single transmembrane domain and a highly glycosylated N-terminus. Unlike the other two CLC auxiliary subunits, Ostm1 shows a reciprocal relationship with ClC-7 for their stability. The subcellular localization of Ostm1 depends on ClC-7 and not the other way around. ClC-2 is active on its own, but GlialCAM, a transmembrane cell adhesion molecule with two extracellular immunoglobulin (Ig)-like domains, regulates its subcellular localization and activity in glial cells. The common theme for these three proteins is their requirement for a proper homeostasis, since their malfunction leads to distinct diseases. We will review here their properties and their role in normal chloride physiology and the pathological consequences of their improper function.
Collapse
Affiliation(s)
- Alejandro Barrallo-Gimeno
- Sección de Fisiología, Departamento de Ciencias Fisiológicas II, University of Barcelona, Barcelona, Spain.,U-750, Centro de investigación en red de enfermedades raras (CIBERER), ISCIII, Barcelona, Spain
| | | | - Ilaria Zanardi
- Istituto di Biofisica, Consiglio Nazionale delle Ricerche, Genoa, Italy
| | - Michael Pusch
- Istituto di Biofisica, Consiglio Nazionale delle Ricerche, Genoa, Italy
| | - Raúl Estévez
- Sección de Fisiología, Departamento de Ciencias Fisiológicas II, University of Barcelona, Barcelona, Spain.,U-750, Centro de investigación en red de enfermedades raras (CIBERER), ISCIII, Barcelona, Spain
| |
Collapse
|
16
|
Martin E, Qureshi A, Dasa V, Freitas M, Gimble J, Davis T. MicroRNA regulation of stem cell differentiation and diseases of the bone and adipose tissue: Perspectives on miRNA biogenesis and cellular transcriptome. Biochimie 2016; 124:98-111. [DOI: 10.1016/j.biochi.2015.02.012] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 02/17/2015] [Indexed: 12/19/2022]
|
17
|
Gernapudi R, Wolfson B, Zhang Y, Yao Y, Yang P, Asahara H, Zhou Q. MicroRNA 140 Promotes Expression of Long Noncoding RNA NEAT1 in Adipogenesis. Mol Cell Biol 2016; 36:30-8. [PMID: 26459763 PMCID: PMC4702608 DOI: 10.1128/mcb.00702-15] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 08/15/2015] [Accepted: 10/02/2015] [Indexed: 02/06/2023] Open
Abstract
More than 40% of the U.S. population are clinically obese and suffer from metabolic syndrome with an increased risk of postmenopausal estrogen receptor-positive breast cancer. Adipocytes are the primary component of adipose tissue and are formed through adipogenesis from precursor mesenchymal stem cells. While the major molecular pathways of adipogenesis are understood, little is known about the noncoding RNA signaling networks involved in adipogenesis. Using adipocyte-derived stem cells (ADSCs) isolated from wild-type and microRNA 140 (miR-140) knockout mice, we identify a novel miR-140/long noncoding RNA (lncRNA) NEAT1 signaling network necessary for adipogenesis. miR-140 knockout ADSCs have dramatically decreased adipogenic capabilities associated with downregulation of NEAT1 expression. We identified a miR-140 binding site in NEAT1 and found that mature miR-140 in the nucleus can physically interact with NEAT1, leading to increased NEAT1 expression. We demonstrated that reexpression of NEAT1 in miR-140 knockout ADSCs is sufficient to restore their ability to undergo differentiation. Our results reveal an exciting new noncoding RNA signaling network that regulates adipogenesis and that is a potential new target in the prevention or treatment of obesity.
Collapse
Affiliation(s)
- Ramkishore Gernapudi
- Department of Biochemistry and Molecular Biology, Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Benjamin Wolfson
- Department of Biochemistry and Molecular Biology, Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Yongshu Zhang
- Department of Biochemistry and Molecular Biology, Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Yuan Yao
- Department of Biochemistry and Molecular Biology, Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Peixin Yang
- Department of Obstetrics, Gynecology and Reproductive Sciences at University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Hiroshi Asahara
- The Scripps Research Institute Department of Molecular and Experimental Medicine, La Jolla, California, USA Department of Systems Biomedicine, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Qun Zhou
- Department of Biochemistry and Molecular Biology, Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
18
|
miR-140-5p regulates adipocyte differentiation by targeting transforming growth factor-β signaling. Sci Rep 2015; 5:18118. [PMID: 26657345 PMCID: PMC4676041 DOI: 10.1038/srep18118] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 11/12/2015] [Indexed: 12/21/2022] Open
Abstract
Recent emerging studies of miRNAs in adipocyte commitment provide new insights to understand the molecular basis of adipogenesis. The current study indicated that miR-140-5p was altered in primary cultured marrow stromal cells and established progenitor lines after adipogenic and/or osteogenic treatment. miR-140-5p was increased in adipose tissue in db/db obese mice vs. lean mice. Supplementing miR-140-5p activity induced stromal cell ST2 and preadipocyte 3T3-L1 to differentiate into mature adipocytes. Conversely, inhibition of the endogenous miR-140-5p repressed ST2 and 3T3-L1 to fully differentiate. By contrast, knockdown of the endogenous miR-140-5p enhanced osteoblast differentiation. Transforming growth factor-β receptor I (Tgfbr1) was shown to be a direct target of miR-140-5p. Supplementing miR-140-5p in ST2 reduced the level of TGFBR1 protein, while suppression of endogenous miR-140-5p increased TGFBR1. Overexpression of Tgfbr1 inhibited, whereas knockdown of Tgfbr1 promoted adipogenic differentiation of ST2 cells. Further investigation of mechanisms that control miR-140-5p expression revealed that C/EBPα induced transcriptional activity of the miR-140-5p promoter. Removal of the putative response element of C/EBP from the promoter abolished the enhancement of the promoter activity by C/EBPα, suggesting that C/EBPα transcriptionally controls miR-140-5p expression. Taken together, our study provides evidences that miR-140-5p regulates adipocyte differentiation through a C/EBP/miR-140-5p/TGFBR1 regulatory feedback loop.
Collapse
|
19
|
Barter MJ, Tselepi M, Gómez R, Woods S, Hui W, Smith GR, Shanley DP, Clark IM, Young DA. Genome-Wide MicroRNA and Gene Analysis of Mesenchymal Stem Cell Chondrogenesis Identifies an Essential Role and Multiple Targets for miR-140-5p. Stem Cells 2015; 33:3266-80. [PMID: 26175215 PMCID: PMC4737122 DOI: 10.1002/stem.2093] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 05/20/2015] [Accepted: 06/01/2015] [Indexed: 12/21/2022]
Abstract
microRNAs (miRNAs) are abundantly expressed in development where they are critical determinants of cell differentiation and phenotype. Accordingly miRNAs are essential for normal skeletal development and chondrogenesis in particular. However, the question of which miRNAs are specific to the chondrocyte phenotype has not been fully addressed. Using microarray analysis of miRNA expression during mesenchymal stem cell chondrogenic differentiation and detailed examination of the role of essential differentiation factors, such as SOX9, TGF-β, and the cell condensation phase, we characterize the repertoire of specific miRNAs involved in chondrocyte development, highlighting in particular miR-140 and miR-455. Further with the use of mRNA microarray data we integrate miRNA expression and mRNA expression during chondrogenesis to underline the particular importance of miR-140, especially the -5p strand. We provide a detailed identification and validation of direct targets of miR-140-5p in both chondrogenesis and adult chondrocytes with the use of microarray and 3'UTR analysis. This emphasizes the diverse array of targets and pathways regulated by miR-140-5p. We are also able to confirm previous experimentally identified targets but, additionally, identify a novel positive regulation of the Wnt signaling pathway by miR-140-5p. Wnt signaling has a complex role in chondrogenesis and skeletal development and these findings illustrate a previously unidentified role for miR-140-5p in regulation of Wnt signaling in these processes. Together these developments further highlight the role of miRNAs during chondrogenesis to improve our understanding of chondrocyte development and guide cartilage tissue engineering.
Collapse
Affiliation(s)
- Matt J. Barter
- Institute of Cellular MedicineNewcastle UniversityNewcastle upon TyneUnited Kingdom
| | - Maria Tselepi
- Institute of Cellular MedicineNewcastle UniversityNewcastle upon TyneUnited Kingdom
| | - Rodolfo Gómez
- Institute of Cellular MedicineNewcastle UniversityNewcastle upon TyneUnited Kingdom
| | - Steven Woods
- Institute of Cellular MedicineNewcastle UniversityNewcastle upon TyneUnited Kingdom
| | - Wang Hui
- Institute of Cellular MedicineNewcastle UniversityNewcastle upon TyneUnited Kingdom
| | - Graham R. Smith
- Institute of Cellular MedicineNewcastle UniversityNewcastle upon TyneUnited Kingdom
| | - Daryl P. Shanley
- Institute for Ageing and HealthNewcastle UniversityNewcastle upon TyneUnited Kingdom
| | - Ian M. Clark
- School of Biological SciencesUniversity of East AngliaNorwichUnited Kingdom
| | - David A. Young
- Institute of Cellular MedicineNewcastle UniversityNewcastle upon TyneUnited Kingdom
| |
Collapse
|
20
|
Effects of canola proteins and hydrolysates on adipogenic differentiation of C3H10T/2 mesenchymal stem cells. Food Chem 2015; 185:226-32. [DOI: 10.1016/j.foodchem.2015.03.054] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 02/23/2015] [Accepted: 03/17/2015] [Indexed: 11/20/2022]
|
21
|
Chen K, He H, Xie Y, Zhao L, Zhao S, Wan X, Yang W, Mo Z. miR-125a-3p and miR-483-5p promote adipogenesis via suppressing the RhoA/ROCK1/ERK1/2 pathway in multiple symmetric lipomatosis. Sci Rep 2015; 5:11909. [PMID: 26148871 PMCID: PMC4493643 DOI: 10.1038/srep11909] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 04/22/2015] [Indexed: 01/08/2023] Open
Abstract
Multiple symmetric lipomatosis (MSL) is a rare disease characterized by symmetric and abnormal distribution of subcutaneous adipose tissue (SAT); however, the etiology is largely unknown. We report here that miR-125a-3p and miR-483-5p are upregulated in the SAT of MSL patients, promoting adipogenesis through suppressing the RhoA/ROCK1/ERK1/2 pathway. TaqMan microRNA (miR) array analysis revealed that 18 miRs were upregulated in the SAT of MSL patients. Transfection of human adipose-derived mesenchymal stem cells (hADSCs) with the individual agomirs of these 18 miRs showed that miR-125a-3p and miR-483-5p significantly promoted adipogenesis. A dual-luciferase assay showed that RhoA and ERK1 were the targets of miR-125a-3p and miR-483-5p, respectively. Moreover, transfection of hADSCs with mimics of miR-125a-3p and miR-483-5p resulted in a pronounced decrease of ERK1/2 phosphorylation in the nucleus; conversely, transfection of hADSCs with inhibitors of miR-125a-3p and miR-483-5p led to a significant increase of ERK1/2 phosphorylation in the nucleus. Most importantly, we found that miR-125a-3p and miR-483-5p promoted de novo adipose tissue formation in nude mice. These results demonstrated that miR-125a-3p and miR-483-5p coordinately promoted adipogenesis through suppressing the RhoA/ROCK1/ERK1/2 pathway. Our findings may provide novel strategies for the management and treatment of MSL or obesity.
Collapse
Affiliation(s)
- Ke Chen
- Department of Endocrinology, Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, China
| | - Honghui He
- Department of Endocrinology, Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, China
| | - Yanhong Xie
- Department of Endocrinology, Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, China
| | - Liling Zhao
- Department of Endocrinology, Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, China
| | - Shaoli Zhao
- Department of Endocrinology, Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, China
| | - Xinxing Wan
- Department of Endocrinology, Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, China
| | - Wenjun Yang
- Department of Endocrinology, Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, China
| | - Zhaohui Mo
- Department of Endocrinology, Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, China
| |
Collapse
|
22
|
Shi XE, Li YF, Jia L, Ji HL, Song ZY, Cheng J, Wu GF, Song CC, Zhang QL, Zhu JY, Yang GS. MicroRNA-199a-5p affects porcine preadipocyte proliferation and differentiation. Int J Mol Sci 2014; 15:8526-38. [PMID: 24830555 PMCID: PMC4057746 DOI: 10.3390/ijms15058526] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Revised: 03/31/2014] [Accepted: 05/04/2014] [Indexed: 11/16/2022] Open
Abstract
MicroRNAs (miRNAs), a class of small non-coding RNAs, have emerged as novel and potent regulators of adipogenesis. However, few miRNAs have been fully investigated in porcine adipogenesis, given the fact that pig is not only an apropos model of human obesity research, but also a staple meat source of human diet. In this study, we showed that miRNA-199a-5p is highly expressed in porcine subcutaneous fat deposits compared to several other tissue types and organs measured alongside. Overexpression of miR-199a-5p in porcine preadipocytes significantly promoted cell proliferation while attenuating the lipid deposition in porcine adipocytes. By target gene prediction and experimental validation, we demonstrated that caveolin-1 (Cav-1) may be a bona fide target of miR-199a-5p in porcine adipocytes, accounting for some of miR-199a-5p’s functions. Taken together, our data established a role of miR-199a-5p in porcine preadipocyte proliferation and differentiation, which is at least partially played by downregulating Cav-1.
Collapse
Affiliation(s)
- Xin-E Shi
- Laboratory of Animal Fat Deposition and Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China.
| | - Yue-Feng Li
- Laboratory of Animal Fat Deposition and Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China.
| | - Long Jia
- Laboratory of Animal Fat Deposition and Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China.
| | - Hong-Lei Ji
- Laboratory of Animal Fat Deposition and Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China.
| | - Zi-Yi Song
- Laboratory of Animal Fat Deposition and Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China.
| | - Jia Cheng
- Vitamin D Research Institute, Shaanxi University of Technology, Hanzhong 723000, China.
| | - Guo-Fang Wu
- Laboratory of Animal Fat Deposition and Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China.
| | - Cheng-Chuang Song
- Laboratory of Animal Fat Deposition and Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China.
| | - Qiang-Ling Zhang
- Laboratory of Animal Fat Deposition and Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China.
| | - Jia-Yu Zhu
- Laboratory of Animal Fat Deposition and Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China.
| | - Gong-She Yang
- Laboratory of Animal Fat Deposition and Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China.
| |
Collapse
|
23
|
Chiang DY, Cuthbertson DW, Ruiz FR, Li N, Pereira FA. A coregulatory network of NR2F1 and microRNA-140. PLoS One 2013; 8:e83358. [PMID: 24349493 PMCID: PMC3857795 DOI: 10.1371/journal.pone.0083358] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Accepted: 11/11/2013] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Both nuclear receptor subfamily 2 group F member 1 (NR2F1) and microRNAs (miRNAs) have been shown to play critical roles in the developing and functional inner ear. Based on previous studies suggesting interplay between NR2F1 and miRNAs, we investigated the coregulation between NR2F1 and miRNAs to better understand the regulatory mechanisms of inner ear development and functional maturation. RESULTS Using a bioinformatic approach, we identified 11 potential miRNAs that might coregulate target genes with NR2F1 and analyzed their targets and potential roles in physiology and disease. We selected 6 miRNAs to analyze using quantitative real-time (qRT) -PCR and found that miR-140 is significantly down-regulated by 4.5-fold (P=0.004) in the inner ear of NR2F1 knockout (Nr2f1(-/-)) mice compared to wild-type littermates but is unchanged in the brain. Based on this, we performed chromatin-immunoprecipitation followed by qRT-PCR and confirmed that NR2F1 directly binds and regulates both miR-140 and Klf9 in vivo. Furthermore, we performed luciferase reporter assay and showed that miR-140 mimic directly regulates KLF9-3'UTR, thereby establishing and validating an example coregulatory network involving NR2F1, miR-140, and Klf9. CONCLUSIONS We have described and experimentally validated a novel tissue-dependent coregulatory network for NR2F1, miR-140, and Klf9 in the inner ear and we propose the existence of many such coregulatory networks important for both inner ear development and function.
Collapse
Affiliation(s)
- David Y. Chiang
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, United States of America
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas, United States of America
| | - David W. Cuthbertson
- Bobby R. Alford Department of Otolaryngology- Head and Neck Surgery, Baylor College of Medicine, Houston, Texas, United States of America
| | - Fernanda R. Ruiz
- Huffington Center on Aging and Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Na Li
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Fred A. Pereira
- Bobby R. Alford Department of Otolaryngology- Head and Neck Surgery, Baylor College of Medicine, Houston, Texas, United States of America
- Huffington Center on Aging and Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, United States of America
| |
Collapse
|