1
|
Yan Z, Ruan B, Zhu Z, Cao X, Lu Z. Azoramide, a novel regulator, favors adipogenesis against osteogenesis through inhibiting the GLP-1 receptor-PKA-β-catenin pathway. Hum Cell 2025; 38:73. [PMID: 40108027 PMCID: PMC11923014 DOI: 10.1007/s13577-025-01192-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 02/22/2025] [Indexed: 03/22/2025]
Abstract
The reciprocal fate decision of mesenchymal stem cells (MSCs) to either bone or adipocytes is determined by Wnt-related signaling and the glucagon-like peptide-1 receptor (GLP-1R). Azoramide, an ER stress alleviator, was reported to have an antidiabetic effect. In this study, we investigated the function of azoramide in regulating the lineage determination of MSCs for either adipogenic or osteogenic differentiation. Microcomputed tomography and histological analysis on bone morphogenetic protein (BMP)2-induced parietal periosteum bone formation assays, C3H10T1/2 and mouse bone marrow MSC-derived bone formation and adipogenesis assays, and specific staining for bone tissue and lipid droplets were used to evaluate the role of azoramide on the lineage determination of MSC differentiation. Cells were harvested for Western blot and quantitative real-time polymerase chain reaction (PCR), and immunofluorescence staining was used to explore the potential mechanism of azoramide for regulating MSC differentiation. Based on MSC-derived bone formation assays both in vivo and in vitro, azoramide treatment displayed a cell fate determining ability in favor of adipogenesis over osteogenesis. Further mechanistic characterizations disclosed that both the GLP-1R agonist peptide exendin-4 (Ex-4) and GLP-1R small interfering (si)RNA abrogated azoramide dual effects. Moreover, cAMP-protein kinase A (PKA)-mediated nuclear β-catenin activity was responsible for the negative function of azoramide on bone formation in favor of adipogenesis. These data provide the first evidence to show that azoramide may serve as an inhibitor against GLP-1R in MSC lineage determination.
Collapse
Affiliation(s)
- Zhao Yan
- Department of Anatomy and K.K. Leung Brain Research Centre, Fourth Military Medical University, Xi'an, 710032, China
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Banjun Ruan
- State Key Laboratory of Cancer Biology, Department of Pharmacogenomics, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Zheng Zhu
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shanxi, China
| | - Xiaorui Cao
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China.
- Innovation Research Institute, Xijing Hospital, Airforce Medical University, Xi'an, 710032, Shanxi, China.
| | - Zifan Lu
- State Key Laboratory of Cancer Biology, Department of Pharmacogenomics, Fourth Military Medical University, Xi'an, 710032, People's Republic of China.
| |
Collapse
|
2
|
Iacobini C, Vitale M, Haxhi J, Menini S, Pugliese G. Impaired Remodeling of White Adipose Tissue in Obesity and Aging: From Defective Adipogenesis to Adipose Organ Dysfunction. Cells 2024; 13:763. [PMID: 38727299 PMCID: PMC11083890 DOI: 10.3390/cells13090763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 04/22/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
The adipose organ adapts and responds to internal and environmental stimuli by remodeling both its cellular and extracellular components. Under conditions of energy surplus, the subcutaneous white adipose tissue (WAT) is capable of expanding through the enlargement of existing adipocytes (hypertrophy), followed by de novo adipogenesis (hyperplasia), which is impaired in hypertrophic obesity. However, an impaired hyperplastic response may result from various defects in adipogenesis, leading to different WAT features and metabolic consequences, as discussed here by reviewing the results of the studies in animal models with either overexpression or knockdown of the main molecular regulators of the two steps of the adipogenesis process. Moreover, impaired WAT remodeling with aging has been associated with various age-related conditions and reduced lifespan expectancy. Here, we delve into the latest advancements in comprehending the molecular and cellular processes underlying age-related changes in WAT function, their involvement in common aging pathologies, and their potential as therapeutic targets to influence both the health of elderly people and longevity. Overall, this review aims to encourage research on the mechanisms of WAT maladaptation common to conditions of both excessive and insufficient fat tissue. The goal is to devise adipocyte-targeted therapies that are effective against both obesity- and age-related disorders.
Collapse
|
3
|
Lee LMY, Lin ZQ, Zheng LX, Tu YF, So YH, Zheng XH, Feng TJ, Wang XY, Wong WT, Leung YC. Lysine Deprivation Suppresses Adipogenesis in 3T3-L1 Cells: A Transcriptome Analysis. Int J Mol Sci 2023; 24:ijms24119402. [PMID: 37298352 DOI: 10.3390/ijms24119402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/21/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023] Open
Abstract
Growing evidence proves that amino acid restriction can reverse obesity by reducing adipose tissue mass. Amino acids are not only the building blocks of proteins but also serve as signaling molecules in multiple biological pathways. The study of adipocytes' response to amino acid level changes is crucial. It has been reported that a low concentration of lysine suppresses lipid accumulation and transcription of several adipogenic genes in 3T3-L1 preadipocytes. However, the detailed lysine-deprivation-induced cellular transcriptomic changes and the altered pathways have yet to be fully studied. Here, using 3T3-L1 cells, we performed RNA sequencing on undifferentiated and differentiated cells, and differentiated cells under a lysine-free environment, and the data were subjected to KEGG enrichment. We found that the differentiation process of 3T3-L1 cells to adipocytes required the large-scale upregulation of metabolic pathways, mainly on the mitochondrial TCA cycle, oxidative phosphorylation, and downregulation of the lysosomal pathway. Single amino acid lysine depletion suppressed differentiation dose dependently. It disrupted the metabolism of cellular amino acids, which could be partially reflected in the changes in amino acid levels in the culture medium. It inhibited the mitochondria respiratory chain and upregulated the lysosomal pathway, which are essential for adipocyte differentiation. We also noticed that cellular interleukin 6 (IL6) expression and medium IL6 level were dramatically increased, which was one of the targets for suppressing adipogenesis induced by lysine depletion. Moreover, we showed that the depletion of some essential amino acids such as methionine and cystine could induce similar phenomena. This suggests that individual amino acid deprivation may share some common pathways. This descriptive study dissects the pathways for adipogenesis and how the cellular transcriptome was altered under lysine depletion.
Collapse
Affiliation(s)
- Leo Man-Yuen Lee
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
- Lo Ka Chung Research Centre for Natural Anti-Cancer Drug Development and State Key Laboratory of Chemical Biology and Drug Discovery, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
- School of Biomedical Science, The Chinese University of Hong Kong, Shatin, New Territory, Hong Kong, China
| | - Zhi-Qiang Lin
- School of Biomedical Science, The Chinese University of Hong Kong, Shatin, New Territory, Hong Kong, China
| | - Lu-Xi Zheng
- School of Biomedical Science, The Chinese University of Hong Kong, Shatin, New Territory, Hong Kong, China
| | - Yi-Fan Tu
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
- Lo Ka Chung Research Centre for Natural Anti-Cancer Drug Development and State Key Laboratory of Chemical Biology and Drug Discovery, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
- Department of Obstetrics and Gynecology, The Chinese University of Hong Kong, New Territory, Hong Kong, China
| | - Yik-Hing So
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
- Lo Ka Chung Research Centre for Natural Anti-Cancer Drug Development and State Key Laboratory of Chemical Biology and Drug Discovery, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| | - Xiu-Hua Zheng
- School of Biomedical Science, The Chinese University of Hong Kong, Shatin, New Territory, Hong Kong, China
| | - Tie-Jun Feng
- School of Biomedical Science, The Chinese University of Hong Kong, Shatin, New Territory, Hong Kong, China
| | - Xi-Yue Wang
- Shenzhen Research Institute, The Hong Kong Polytechnic University, Shenzhen 518000, China
| | - Wai-Ting Wong
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| | - Yun-Chung Leung
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
- Lo Ka Chung Research Centre for Natural Anti-Cancer Drug Development and State Key Laboratory of Chemical Biology and Drug Discovery, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| |
Collapse
|
4
|
A Wrong Fate Decision in Adipose Stem Cells upon Obesity. Cells 2023; 12:cells12040662. [PMID: 36831329 PMCID: PMC9954614 DOI: 10.3390/cells12040662] [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: 01/30/2023] [Revised: 02/16/2023] [Accepted: 02/17/2023] [Indexed: 02/22/2023] Open
Abstract
Progress has been made in identifying stem cell aging as a pathological manifestation of a variety of diseases, including obesity. Adipose stem cells (ASCs) play a core role in adipocyte turnover, which maintains tissue homeostasis. Given aberrant lineage determination as a feature of stem cell aging, failure in adipogenesis is a culprit of adipose hypertrophy, resulting in adiposopathy and related complications. In this review, we elucidate how ASC fails in entering adipogenic lineage, with a specific focus on extracellular signaling pathways, epigenetic drift, metabolic reprogramming, and mechanical stretch. Nonetheless, such detrimental alternations can be reversed by guiding ASCs towards adipogenesis. Considering the pathological role of ASC aging in obesity, targeting adipogenesis as an anti-obesity treatment will be a key area of future research, and a strategy to rejuvenate tissue stem cell will be capable of alleviating metabolic syndrome.
Collapse
|
5
|
Song Z, Liu N, He Y, Chen J, Li J, Wang F, Wu Z. Knockout of ICAT in Adipose Tissue Alleviates Fibro-inflammation in Obese Mice. Inflammation 2023; 46:404-417. [PMID: 36181623 DOI: 10.1007/s10753-022-01742-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/08/2022] [Accepted: 09/14/2022] [Indexed: 11/24/2022]
Abstract
The E2 promoter binding factor 1 (E2F1) and the Wnt/β-catenin signaling are crucial in regulating metabolic homeostasis including obesity. The β-catenin interacting protein 1 (CTNNBIP1), also known as the inhibitor of β-catenin and TCF4 (ICAT), is required for E2F1 to inhibit the activity of β-catenin. However, the role of ICAT in E2F1 regulating obesity-related metabolic disorders remains unknown. In the present study, male adipose tissue-specific ICAT knockout (ICATadi-/-) C57BL/6 J mice and control littermates aged 6-8 weeks were fed with high-fat diet (HFD) for 12 weeks to explore the effect of ICAT on lipid metabolism and obesity-related disorders. Results showed that the adipose tissue-specific ICAT knockout had negligible effect on lipid metabolism, reflected by no difference in body weight, fat mass, and the expression of proteins involved in lipid metabolism in white adipose tissue (WAT) and the liver between the ICATadi-/- mice and the control littermate (ICATfl/fl) mice. However, the knockout of ICAT reduced inflammatory response in WAT and the liver. Additionally, Sirius red staining results showed that deletion of ICAT attenuated fibrosis and reduced mRNA levels of transforming growth factor β1(TGF-β1), matrix metallopeptidase 2 (Mmp2), Mmp3, and collagen, type V, alpha 1 (Col5a1) in WAT and the liver. These results suggested that knockout of ICAT improved the metabolic abnormalities of obese mice through attenuating adipose tissue and the liver inflammation as well as fibrosis. Our findings may provide a new insight to understand the role of ICAT in inflammation and fibrosis.
Collapse
Affiliation(s)
- Zhuan Song
- Department of Animal Nutrition and Feed Science, State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China
| | - Ning Liu
- Department of Animal Nutrition and Feed Science, State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China.,Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, 100193, China
| | - Yu He
- Department of Animal Nutrition and Feed Science, State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China
| | - Jingqing Chen
- Laboratory Animal Center of the Academy of Military Medical Sciences, Beijing, 100193, China
| | - Jun Li
- Department of Animal Nutrition and Feed Science, State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China
| | - Fengchao Wang
- National Institute of Biological Sciences (NIBS), Beijing, 102206, China
| | - Zhenlong Wu
- Department of Animal Nutrition and Feed Science, State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, 100193, China. .,Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
6
|
Integrative Analysis of miRNAs Involved in Fat Deposition in Different Pig Breeds. Genes (Basel) 2022; 14:genes14010094. [PMID: 36672834 PMCID: PMC9859024 DOI: 10.3390/genes14010094] [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: 11/19/2022] [Revised: 12/17/2022] [Accepted: 12/20/2022] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND miRNAs are a set of small, noncoding RNAs that bind to partially complementary sequences on target mRNAs. This leads to the post-transcriptional regulation of gene expression. Many studies have shown that microRNAs play critical roles in adipose cell differentiation and fat metabolism. The aim of this study was to explore the regulatory functions of miRNAs in fat deposition for the prevention and therapy of lipid metabolism-related diseases. METHODS The significant differences in the fat deposition of Laiwu (LW) pigs and Large White (LY) pigs were studied. To investigate the genetic relationships of miRNAs that regulate fat deposition, we performed a genome-wide analysis of miRNAs derived from subcutaneous adipose tissue of LW and LY pigs using RNA-seq. RESULTS There were 39 known miRNAs and 56 novel miRNAs significantly differential expressed between the two breeds of pigs. In the analysis of the Gene Ontology and KEGG pathways, predicted targets of these differentially expressed miRNAs were involved in several fat-associated pathways, such as the peroxisome proliferator-activated receptor (PPAR), mitogen-activated protein kinases (MAPK) and Wnt signaling pathways. In addition, ssc-miR-133a-3p, ssc-miR-486 and ssc-miR-1 each had a great impact on the development of porcine subcutaneous fat through the PPAR signaling pathway. CONCLUSIONS We explored the role of differentially expressed miRNAs and studied the mechanisms of adipogenesis and fat deposition between two different pig breeds. In addition, these results also contribute to research relevant to human obesity.
Collapse
|
7
|
Khan F, Khan H, Khan A, Yamasaki M, Moustaid-Moussa N, Al-Harrasi A, Rahman SM. Autophagy in adipogenesis: Molecular mechanisms and regulation by bioactive compounds. Biomed Pharmacother 2022; 155:113715. [PMID: 36152415 DOI: 10.1016/j.biopha.2022.113715] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/19/2022] [Accepted: 09/19/2022] [Indexed: 11/02/2022] Open
Abstract
White adipose tissue expands rapidly due to increased adipocyte number (hyperplasia) and size (hypertrophy), which results in obesity. Adipogenesis is a process of the formation of mature adipocytes from precursor cells. Additionally, obesity-related metabolic complications, such as fatty liver and insulin resistance, are linked to adipogenesis. On the contrary, autophagy is a catabolic process; essential to maintain cellular homeostasis via the degradation or recycling of unnecessary or damaged components. Importantly, autophagy dictates obesity and adipogenesis. Hence, a clear understanding of how autophagy regulates adipogenesis is crucial for drug development and the prevention and treatment of obesity and its associated disorders, such as type 2 diabetes, cardiovascular disease, and cancer. In this review, we highlighted recent findings regarding the crosstalk between adipogenesis and autophagy, as well as the molecules involved. Furthermore, the review discussed how bioactive compounds regulate adipogenesis by manipulating autophagy and underlying molecular mechanisms. Based on in vitro and animal studies, we summarized the effects of bioactive compounds on adipogenesis and autophagy. Hence, human studies are necessary to validate the effectiveness and optimal dosage of these bioactive compounds.
Collapse
Affiliation(s)
- Faizullah Khan
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa 616, Sultanate of Oman; Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan 23200 Khyber Pakhtunkhwa, Pakistan
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan 23200 Khyber Pakhtunkhwa, Pakistan
| | - Ajmal Khan
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa 616, Sultanate of Oman
| | - Masao Yamasaki
- Department of Biochemistry and Applied Biosciences, Faculty of Agriculture, University of Miyazaki, Miyazaki, Japan
| | - Naima Moustaid-Moussa
- Texas Tech University, Nutritional Sciences, Lubbock, TX 79409, USA; Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA
| | - Ahmed Al-Harrasi
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa 616, Sultanate of Oman
| | - Shaikh Mizanoor Rahman
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa 616, Sultanate of Oman.
| |
Collapse
|
8
|
Silva-Vignato B, Cesar ASM, Afonso J, Moreira GCM, Poleti MD, Petrini J, Garcia IS, Clemente LG, Mourão GB, Regitano LCDA, Coutinho LL. Integrative Analysis Between Genome-Wide Association Study and Expression Quantitative Trait Loci Reveals Bovine Muscle Gene Expression Regulatory Polymorphisms Associated With Intramuscular Fat and Backfat Thickness. Front Genet 2022; 13:935238. [PMID: 35991540 PMCID: PMC9386181 DOI: 10.3389/fgene.2022.935238] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 06/23/2022] [Indexed: 11/13/2022] Open
Abstract
Understanding the architecture of gene expression is fundamental to unravel the molecular mechanisms regulating complex traits in bovine, such as intramuscular fat content (IMF) and backfat thickness (BFT). These traits are economically important for the beef industry since they affect carcass and meat quality. Our main goal was to identify gene expression regulatory polymorphisms within genomic regions (QTL) associated with IMF and BFT in Nellore cattle. For that, we used RNA-Seq data from 193 Nellore steers to perform SNP calling analysis. Then, we combined the RNA-Seq SNP and a high-density SNP panel to obtain a new dataset for further genome-wide association analysis (GWAS), totaling 534,928 SNPs. GWAS was performed using the Bayes B model. Twenty-one relevant QTL were associated with our target traits. The expression quantitative trait loci (eQTL) analysis was performed using Matrix eQTL with the complete SNP dataset and 12,991 genes, revealing a total of 71,033 cis and 36,497 trans-eQTL (FDR < 0.05). Intersecting with QTL for IMF, we found 231 eQTL regulating the expression levels of 117 genes. Within those eQTL, three predicted deleterious SNPs were identified. We also identified 109 eQTL associated with BFT and affecting the expression of 54 genes. This study revealed genomic regions and regulatory SNPs associated with fat deposition in Nellore cattle. We highlight the transcription factors FOXP4, FOXO3, ZSCAN2, and EBF4, involved in lipid metabolism-related pathways. These results helped us to improve our knowledge about the genetic architecture behind important traits in cattle.
Collapse
Affiliation(s)
- Bárbara Silva-Vignato
- Department of Animal Science, College of Agriculture “Luiz de Queiroz”, University of São Paulo, Piracicaba, Brazil
| | - Aline Silva Mello Cesar
- Department of Agroindustry, Food, and Nutrition, College of Agriculture “Luiz de Queiroz”, University of São Paulo, Piracicaba, Brazil
| | | | | | - Mirele Daiana Poleti
- College of Animal Science and Food Engineering, University of São Paulo, Pirassununga, Brazil
| | - Juliana Petrini
- Department of Animal Science, College of Agriculture “Luiz de Queiroz”, University of São Paulo, Piracicaba, Brazil
| | - Ingrid Soares Garcia
- Department of Animal Science, College of Agriculture “Luiz de Queiroz”, University of São Paulo, Piracicaba, Brazil
| | - Luan Gaspar Clemente
- Department of Animal Science, College of Agriculture “Luiz de Queiroz”, University of São Paulo, Piracicaba, Brazil
| | - Gerson Barreto Mourão
- Department of Animal Science, College of Agriculture “Luiz de Queiroz”, University of São Paulo, Piracicaba, Brazil
| | | | - Luiz Lehmann Coutinho
- Department of Animal Science, College of Agriculture “Luiz de Queiroz”, University of São Paulo, Piracicaba, Brazil
- *Correspondence: Luiz Lehmann Coutinho,
| |
Collapse
|
9
|
Wen Q, Xie X, Ren Q, Du Y. Polybrominated diphenyl ether congener 99 (PBDE 99) promotes adipocyte lineage commitment of C3H10T1/2 mesenchymal stem cells. CHEMOSPHERE 2022; 290:133312. [PMID: 34919914 DOI: 10.1016/j.chemosphere.2021.133312] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 11/21/2021] [Accepted: 12/13/2021] [Indexed: 06/14/2023]
Abstract
Obesogens are defined as chemicals that trigger obesity partially by stimulating adipogenesis. Adipogenesis consists of two successive processes: the adipocyte lineage commitment of pluripotent stem cells and the differentiation of preadipocytes. Compared with the differentiation of preadipocytes, the effects of most environmental obesogens on adipocyte lineage commitment remain largely unknown. In this study, investigations are performed to explore the influences of PBDE 99 on the adipocyte lineage commitment based on C3H10T1/2, which has been widely used as a mesenchymal stem cell (MSC) model. Our results indicated that exposure to PBDE 99 during commitment stage resulted in significant up-regulation of subsequent adipogenesis in C3H10T1/2 MSCs. Interestingly, PBDE 99 did not affect the osteogenesis of C3H10T1/2 MSCs, although the adipogenesis and osteogenesis of MSCs are typically reciprocal. PBDE 99 was further demonstrated to significantly decrease the expression of Pref1, the marker of very early adipose mesenchymal precursor, and its downstream effector, Sox9. This result strongly suggested that PBDE 99 facilitated adipocyte commitment to exert adipogenic effect on C3H10T1/2 MSCs. Mechanistic studies revealed that PBDE 99 efficiently inhibited Hedgehog signaling transduction, a conserved negative regulator of the adipocyte lineage commitment. Furthermore, the effects of PBDE 99 on adipogenesis were abrogated by the co-treatment with SAG, a specific Hedgehog signaling activator, suggesting inhibition of Hedgehog signaling is responsible for the effect of PBDE 99 on adipocyte commitment. Taking together, these results strongly suggested enhanced adipocyte lineage commitment was involved in potential obesogenic effect of PBDE 99, presumably through repressing Hedgehog signalling during commitment stage. Moreover, the results of this study indicated that C3H10T1/2 can be used as a feasible MSCs cell model to evaluate the capabilities of potential obesogens on adipocyte commitment.
Collapse
Affiliation(s)
- Qing Wen
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, 100190, China
| | - Xinni Xie
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China.
| | - Qidong Ren
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, 100190, China
| | - Yuguo Du
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, 100190, China.
| |
Collapse
|
10
|
Barilla S, Treuter E, Venteclef N. Transcriptional and epigenetic control of adipocyte remodeling during obesity. Obesity (Silver Spring) 2021; 29:2013-2025. [PMID: 34813171 DOI: 10.1002/oby.23248] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 04/27/2021] [Accepted: 05/07/2021] [Indexed: 01/05/2023]
Abstract
The rising prevalence of obesity over the past decades coincides with the rising awareness that a detailed understanding of both adipose tissue biology and obesity-associated remodeling is crucial for developing therapeutic and preventive strategies. Substantial progress has been made in identifying the signaling pathways and transcriptional networks that orchestrate alterations of adipocyte gene expression linked to diverse phenotypes. Owing to recent advances in epigenomics, we also gained a better appreciation for the fact that different environmental cues can epigenetically reprogram adipocyte fate and function, mainly by altering DNA methylation and histone modification patterns. Intriguingly, it appears that transcription factors and chromatin-modifying coregulator complexes are the key regulatory components that coordinate both signaling-induced transcriptional and epigenetic alterations in adipocytes. In this review, we summarize and discuss current molecular insights into how these alterations and the involved regulatory components trigger adipogenesis and adipose tissue remodeling in response to energy surplus.
Collapse
Affiliation(s)
- Serena Barilla
- Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden
| | - Eckardt Treuter
- Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden
| | - Nicolas Venteclef
- Cordeliers Research Center, Inserm, University of Paris, IMMEDIAB Laboratory, Paris, France
- Inovarion, Paris, France
| |
Collapse
|
11
|
Wang Z, Zhu M, Wang M, Gao Y, Zhang C, Liu S, Qu S, Liu Z, Zhang C. Integrated Multiomic Analysis Reveals the High-Fat Diet Induced Activation of the MAPK Signaling and Inflammation Associated Metabolic Cascades via Histone Modification in Adipose Tissues. Front Genet 2021; 12:650863. [PMID: 34262592 PMCID: PMC8273343 DOI: 10.3389/fgene.2021.650863] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 05/10/2021] [Indexed: 11/13/2022] Open
Abstract
Background The number of diet induced obese population is increasing every year, and the incidence of type 2 diabetes is also on the rise. Histone methylation and acetylation have been shown to be associated with lipogenesis and obesity by manipulating gene expression via the formation of repression or activation domains on chromosomes. Objective In this study, we aimed to explore gene activation or repression and related biological processes by histone modification across the whole genome on a high-fat diet (HFD) condition. We also aimed to elucidate the correlation of these genes that modulated by histone modification with energy metabolism and inflammation under both short-term and long-term HFD conditions. Method We performed ChIP-seq analysis of H3K9me2 and H3K9me3 in brown and white adipose tissues (WATs; subcutaneous adipose tissue) from mice fed with a standard chow diet (SCD) or HFD and a composite analysis of the histone modification of H3K9me2, H3K9me3, H3K4me1 and H3K27ac throughout the whole genome. We also employed and integrated two bulk RNA-seq and a single-nuclei RNA sequencing dataset and performed western blotting (WB) to confirm the gene expression levels in adipose tissue of the SCD and HFD groups. Results The ChIP-seq and transcriptome analysis of mouse adipose tissues demonstrated that a series of genes were activated by the histone modification of H3K9me2, H3K9me3, H3K4me1, and H3K27ac in response to HFD condition. These genes were enriched in Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways involved in lipogenesis, energy metabolism and inflammation. Several genes in the activated mitogen-activated protein kinase (MAPK) pathway might be related to both inflammation and energy metabolism in mice, rats and humans fed with HFD for a short or long term, as showed by bulk RNA-seq and single nuclei RNA-seq datasets. Western blot analyses further confirmed the increased expression of MET, VEGFA and the enhanced phosphorylation ratio of p44/42 MAPK upon HFD treatment. Conclusion This study expanded our understanding of the influence of eating behavior on obesity and could assist the identification of putative therapeutic targets for the prevention and treatment of metabolic disorders in the future.
Collapse
Affiliation(s)
- Zhe Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Institute of Precision Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ming Zhu
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Meng Wang
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Yihui Gao
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Cong Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Institute of Precision Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shangyun Liu
- Department of Plastic and Reconstructive Surgery, Shanghai Institute of Precision Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shen Qu
- Department of Endocrinology and Metabolism, National Metabolic Management Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhongmin Liu
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China.,Department of Cardiac Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chao Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Institute of Precision Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| |
Collapse
|
12
|
Zhong Y, Li X, Wang F, Wang S, Wang X, Tian X, Bai S, Miao D, Fan J. Emerging Potential of Exosomes on Adipogenic Differentiation of Mesenchymal Stem Cells. Front Cell Dev Biol 2021; 9:649552. [PMID: 34239869 PMCID: PMC8258133 DOI: 10.3389/fcell.2021.649552] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 05/28/2021] [Indexed: 12/20/2022] Open
Abstract
The mesenchymal stem cells have multidirectional differentiation potential and can differentiate into adipocytes, osteoblasts, cartilage tissue, muscle cells and so on. The adipogenic differentiation of mesenchymal stem cells is of great significance for the construction of tissue-engineered fat and the treatment of soft tissue defects. Exosomes are nanoscale vesicles secreted by cells and widely exist in body fluids. They are mainly involved in cell communication processes and transferring cargo contents to recipient cells. In addition, exosomes can also promote tissue and organ regeneration. Recent studies have shown that various exosomes can influence the adipogenic differentiation of stem cells. In this review, the effects of exosomes on stem cell differentiation, especially on adipogenic differentiation, will be discussed, and the mechanisms and conclusions will be drawn. The main purpose of studying the role of these exosomes is to understand more comprehensively the influencing factors existing in the process of stem cell differentiation into adipocytes and provide a new idea in adipose tissue engineering research.
Collapse
Affiliation(s)
- Yuxuan Zhong
- Department of Tissue Engineering, School of Fundamental Science, China Medical University, Shenyang, China
| | - Xiang Li
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| | - Fanglin Wang
- Department of Tissue Engineering, School of Fundamental Science, China Medical University, Shenyang, China
| | - Shoushuai Wang
- Department of Tissue Engineering, School of Fundamental Science, China Medical University, Shenyang, China
| | - Xiaohong Wang
- Department of Tissue Engineering, School of Fundamental Science, China Medical University, Shenyang, China
| | - Xiaohong Tian
- Department of Tissue Engineering, School of Fundamental Science, China Medical University, Shenyang, China
| | - Shuling Bai
- Department of Tissue Engineering, School of Fundamental Science, China Medical University, Shenyang, China
| | - Di Miao
- China Medical University-The Queen's University of Belfast Joint College-Combination, Shenyang, China
| | - Jun Fan
- Department of Tissue Engineering, School of Fundamental Science, China Medical University, Shenyang, China
| |
Collapse
|
13
|
Wongtrakul-Kish K, Herbert BR, Packer NH. Bisecting GlcNAc Protein N-Glycosylation Is Characteristic of Human Adipogenesis. J Proteome Res 2020; 20:1313-1327. [PMID: 33383989 DOI: 10.1021/acs.jproteome.0c00702] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Human adipose tissue contains a major source of adipose-derived stem cells (ADSCs) that have the ability to differentiate into various cell types: in vitro, ADSCs can differentiate into mesenchymal lineages including adipocytes, while in vivo, ADSCs become mature adipocytes. Protein glycosylation has been shown to change in stem cell differentiation, and while ADSCs have been acknowledged for their therapeutic potential, little is known about protein glycosylation during human ADSC adipogenic differentiation. In the present study, the global membrane protein glycosylation of native adipocytes was compared to ADSCs from the same individuals as a model of in vivo adipogenesis. For in vitro adipogenesis, ADSCs were adipogenically differentiated in cell culture using an optimized, large-scale differentiation procedure. The membrane glycome of the differentiated ADSCs (dADSCs) was compared with mature adipocytes and the progenitor ADSCs. A total of 137 glycan structures were characterized across the three cell types using PGC-LC coupled with negative-ion electrospray ionization mass spectrometry (ESI-MS)/MS. Significantly higher levels of bisecting GlcNAc-type N-glycans were detected in mature adipocytes (32.1% of total glycans) and in in vitro dADSC progeny (1.9% of total glycans) compared to ADSCs. This was further correlated by the mRNA expression of the MGAT3 gene responsible for the enzymatic synthesis of this structural type. The bisecting GlcNAc structures were found on the majority of human native adipocyte membrane proteins, suggesting an important role in human adipocyte biology. Core fucosylation was also significantly increased during in vivo adipogenesis but did not correlate with an increase in Fut8 gene transcript. Unexpectedly, low abundance structures carrying rare β-linked Gal-Gal termini were also detected. Overall, the N-glycan profiles of the in vitro differentiated progeny did not reflect native adipocytes, and the results show that bisecting GlcNAc structures are a characteristic feature of human adipocyte membrane protein N-glycosylation. Raw MS files are available on GlycoPOST (ID: GPST000153 https://glycopost.glycosmos.org/).
Collapse
Affiliation(s)
- Katherine Wongtrakul-Kish
- Biomolecular Discovery Research Centre, Department of Molecular Sciences, Macquarie University, Sydney, NSW 2109, Australia.,ARC Centre for Nanoscale BioPhotonics, Department of Molecular Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Benjamin R Herbert
- Biomolecular Discovery Research Centre, Department of Molecular Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Nicolle H Packer
- Biomolecular Discovery Research Centre, Department of Molecular Sciences, Macquarie University, Sydney, NSW 2109, Australia.,ARC Centre for Nanoscale BioPhotonics, Department of Molecular Sciences, Macquarie University, Sydney, NSW 2109, Australia
| |
Collapse
|
14
|
Pan Y, Xie Z, Cen S, Li M, Liu W, Tang S, Ye G, Li J, Zheng G, Li Z, Yu W, Wang P, Wu Y, Shen H. Long noncoding RNA repressor of adipogenesis negatively regulates the adipogenic differentiation of mesenchymal stem cells through the hnRNP A1-PTX3-ERK axis. Clin Transl Med 2020; 10:e227. [PMID: 33252864 PMCID: PMC7648959 DOI: 10.1002/ctm2.227] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 10/23/2020] [Accepted: 10/25/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) are pluripotent stem cells that can differentiate via osteogenesis and adipogenesis. The mechanism underlying MSC lineage commitment still remains incompletely elucidated. Understanding the regulatory mechanism of MSC differentiation will help researchers induce MSCs toward specific lineages for clinical use. In this research, we intended to figure out the long noncoding RNA (lncRNA) that plays a central role in MSC fate determination and explore its application value in tissue engineering. METHODS The expression pattern of lncRNAs during MSC osteogenesis/adipogenesis was detected by microarray and qRT-PCR. Lentivirus and siRNAs were constructed to regulate the expression of lncRNA repressor of adipogenesis (ROA). MSC osteogenesis/adipogenesis was evaluated by western blot and alizarin red/oil red staining. An adipokine array was used to select the paracrine/autocrine factor PTX3, followed by RNA interference or recombinant human protein stimulation to confirm its function. The activation of signaling pathways was also detected by western blot, and a small molecule inhibitor, SCH772984, was used to inhibit the activation of the ERK pathway. The interaction between ROA and hnRNP A1 was detected by RNA pull-down and RIP assays. Luciferase reporter and chromatin immunoprecipitation assays were used to confirm the binding of hnRNP A1 to the PTX3 promotor. Additionally, an in vivo adipogenesis experiment was conducted to evaluate the regulatory value of ROA in tissue engineering. RESULTS In this study, we demonstrated that MSC adipogenesis is regulated by lncRNA ROA both in vitro and in vivo. Mechanistically, ROA inhibits MSC adipogenesis by downregulating the expression of the key autocrine/paracrine factor PTX3 and the downstream ERK pathway. This downregulation was achieved through transcription inhibition by impeding hnRNP A1 from binding to the promoter of PTX3. CONCLUSIONS ROA negatively regulates MSC adipogenesis through the hnRNP A1-PTX3-ERK axis. ROA may be an effective target for modulating MSCs in tissue engineering.
Collapse
Affiliation(s)
- Yiqian Pan
- Department of OrthopedicsThe Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhenChina
- Department of OrthopedicsSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Zhongyu Xie
- Department of OrthopedicsThe Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhenChina
| | - Shuizhong Cen
- Department of OrthopedicsSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
- Department of OrthopedicsZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Ming Li
- Department of OrthopedicsSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Wenjie Liu
- Department of OrthopedicsThe Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhenChina
| | - Su'an Tang
- Clinical Research CenterZhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Guiwen Ye
- Department of OrthopedicsSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Jinteng Li
- Department of OrthopedicsThe Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhenChina
| | - Guan Zheng
- Department of OrthopedicsThe Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhenChina
| | - Zhaofeng Li
- Department of OrthopedicsSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Wenhui Yu
- Department of OrthopedicsSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Peng Wang
- Department of OrthopedicsThe Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhenChina
| | - Yanfeng Wu
- Center for BiotherapySun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Huiyong Shen
- Department of OrthopedicsThe Eighth Affiliated HospitalSun Yat‐sen UniversityShenzhenChina
- Department of OrthopedicsSun Yat‐sen Memorial HospitalSun Yat‐sen UniversityGuangzhouChina
| |
Collapse
|
15
|
Song T, Yang Y, Jiang S, Peng J. Novel Insights into Adipogenesis from the Perspective of Transcriptional and RNA N6-Methyladenosine-Mediated Post-Transcriptional Regulation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2001563. [PMID: 33173729 PMCID: PMC7610318 DOI: 10.1002/advs.202001563] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Indexed: 05/09/2023]
Abstract
Obesity is a critical risk factor causing the development of metabolic diseases and cancers. Its increasing prevalence worldwide has aroused great concerns of the researchers on adipose development and metabolic function. During adipose expansion, adipogenesis is a way to store lipids as well as to avoid lipotoxicity in other tissues, and may be an approach to offset the negative metabolic effects of obesity. In this Review, the transcriptional regulation of adipogenesis is outlined to characterize numerous biological processes in research on the determination of adipocyte fate and regulation of adipogenic differentiation. Notably, one of the post-transcriptional modifications of mRNA, namely, N6-methyladenosine (m6A), has been recently found to play a role in adipogenesis. Here, the roles of m6A-related enzymes and proteins in adipogenesis, with a particular focus on how these m6A-related proteins function at different stages of adipogenesis, are mainly discussed. The Review also highlights the coordination role of the transcriptional and post-transcriptional (RNA m6A methylation) regulation in adipogenesis and related biological processes. In this context, a better understanding of adipogenesis at both the transcriptional and post-transcriptional levels may facilitate the development of novel strategies to improve metabolic health in obesity.
Collapse
Affiliation(s)
- Tongxing Song
- Department of Animal Nutrition and Feed ScienceCollege of Animal Science and TechnologyHuazhong Agricultural UniversityWuhan430070China
- The Cooperative Innovation Center for Sustainable Pig ProductionWuhan430070China
| | - Yang Yang
- Department of Animal Nutrition and Feed ScienceCollege of Animal Science and TechnologyHuazhong Agricultural UniversityWuhan430070China
- The Cooperative Innovation Center for Sustainable Pig ProductionWuhan430070China
| | - Siwen Jiang
- The Cooperative Innovation Center for Sustainable Pig ProductionWuhan430070China
- Key Laboratory of Animal GeneticsBreeding and Reproduction Ministry of EducationCollege of Animal Science and TechnologyHuazhong Agricultural UniversityWuhan430070China
| | - Jian Peng
- Department of Animal Nutrition and Feed ScienceCollege of Animal Science and TechnologyHuazhong Agricultural UniversityWuhan430070China
- The Cooperative Innovation Center for Sustainable Pig ProductionWuhan430070China
| |
Collapse
|
16
|
Sutjarit N, Thongon N, Weerachayaphorn J, Piyachaturawat P, Suksamrarn A, Suksen K, Papachristou DJ, Blair HC. Inhibition of Adipogenic Differentiation of Human Bone Marrow-Derived Mesenchymal Stem Cells by a Phytoestrogen Diarylheptanoid from Curcuma comosa. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:9993-10002. [PMID: 32838526 DOI: 10.1021/acs.jafc.0c04063] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
We investigated the effect of a phytoestrogen, (3R)-1,7-diphenyl-(4E,6E)-4,6-heptadien-3-ol (DPHD), from Curcuma comosa Roxb. (Zingiberaceae family) on the adipogenic differentiation of mesenchymal progenitors, human bone marrow-derived mesenchymal stem cells (hBMSCs). DPHD inhibited adipocyte differentiation of hBMSCs by suppressing the expression of genes involved in adipogenesis. DPHD at concentrations of 0.1, 1, and 10 μM significantly decreased triglyceride accumulation in hBMSCs to 7.1 ± 0.2, 6.3 ± 0.4, and 4.9 ± 0.2 mg/dL, respectively, compared to the nontreated control (10.1 ± 0.9 mg/dL) (p < 0.01). Based on gene expression profiling, DPHD increased the expression of several genes involved in the Wnt/β-catenin signaling pathway, a negative regulator of adipocyte differentiation in hBMSCs. DPHD also increased the levels of essential signaling proteins which are extracellular signal-regulated kinases 1 and 2 (ERK1/2) and glycogen synthase kinase 3 beta (GSK-3β) that link estrogen receptor (ER) signaling to Wnt/β-catenin signaling. In conclusion, DPHD exhibited the anti-adipogenic effect in hBMSCs by suppression of adipogenic markers in hBMSCs through the activation of ER and Wnt/β catenin signaling pathways. This finding suggests the potential role of DPHD in preventing bone marrow adiposity which is one of the major factors that exacerbates osteoporosis in postmenopause.
Collapse
Affiliation(s)
- Nareerat Sutjarit
- Toxicology Graduate Program, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Natthakan Thongon
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | | | - Pawinee Piyachaturawat
- Toxicology Graduate Program, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Apichart Suksamrarn
- Department of Chemistry, Faculty of Science, Ramkhamhaeng University, Bangkok 10240, Thailand
| | - Kanoknetr Suksen
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Dionysios J Papachristou
- Laboratory of Bone and Soft Tissue Studies, Department of Anatomy-Histology-Embryology, University Patras Medical School, Patras 26504, Greece
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Harry C Blair
- The Pittsburgh Veterans Affairs Medical Center, Pittsburgh, Pennsylvania 15261, United States
- Department of Pathology, School of Medicine, University of Pittsburgh, 3550 Terrace Street, Pittsburgh, Pennsylvania 15261, United States
| |
Collapse
|
17
|
Wang F, Li X, Li Z, Wang S, Fan J. Functions of Circular RNAs in Regulating Adipogenesis of Mesenchymal Stem Cells. Stem Cells Int 2020; 2020:3763069. [PMID: 32802080 PMCID: PMC7416283 DOI: 10.1155/2020/3763069] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 07/07/2020] [Accepted: 07/08/2020] [Indexed: 12/17/2022] Open
Abstract
The mesenchymal stem cells (MSCs) are known as highly plastic stem cells and can differentiate into specialized tissues such as adipose tissue, osseous tissue, muscle tissue, and nervous tissue. The differentiation of mesenchymal stem cells is very important in regenerative medicine. Their differentiation process is regulated by signaling pathways of epigenetic, transcriptional, and posttranscriptional levels. Circular RNA (circRNA), a class of noncoding RNAs generated from protein-coding genes, plays a pivotal regulatory role in many biological processes. Accumulated studies have demonstrated that several circRNAs participate in the cell differentiation process of mesenchymal stem cells in vitro and in vivo. In the current review, characteristics and functions of circRNAs in stem cell differentiation will be discussed. The mechanism and key role of circRNAs in regulating mesenchymal stem cell differentiation, especially adipogenesis, will be reviewed and discussed. Understanding the roles of these circRNAs will present us with a more comprehensive signal path network of modulating stem cell differentiation and help us discover potential biomarkers and therapeutic targets in clinic.
Collapse
Affiliation(s)
- Fanglin Wang
- Department of Tissue Engineering, School of Fundamental Science, China Medical University, Shenyang, Liaoning 110122, China
| | - Xiang Li
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, And Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Zhiyuan Li
- Department of Tissue Engineering, School of Fundamental Science, China Medical University, Shenyang, Liaoning 110122, China
| | - Shoushuai Wang
- Department of Tissue Engineering, School of Fundamental Science, China Medical University, Shenyang, Liaoning 110122, China
| | - Jun Fan
- Department of Tissue Engineering, School of Fundamental Science, China Medical University, Shenyang, Liaoning 110122, China
| |
Collapse
|
18
|
Ambele MA, Dhanraj P, Giles R, Pepper MS. Adipogenesis: A Complex Interplay of Multiple Molecular Determinants and Pathways. Int J Mol Sci 2020; 21:E4283. [PMID: 32560163 PMCID: PMC7349855 DOI: 10.3390/ijms21124283] [Citation(s) in RCA: 196] [Impact Index Per Article: 39.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 06/07/2020] [Indexed: 11/24/2022] Open
Abstract
The formation of adipocytes during embryogenesis has been largely understudied. However, preadipocytes appear to originate from multipotent mesenchymal stromal/stem cells which migrate from the mesoderm to their anatomical localization. Most studies on adipocyte formation (adipogenesis) have used preadipocytes derived from adult stem/stromal cells. Adipogenesis consists of two phases, namely commitment and terminal differentiation. This review discusses the role of signalling pathways, epigenetic modifiers, and transcription factors in preadipocyte commitment and differentiation into mature adipocytes, as well as limitations in our understanding of these processes. To date, a limited number of transcription factors, genes and signalling pathways have been described to regulate preadipocyte commitment. One reason could be that most studies on adipogenesis have used preadipocytes already committed to the adipogenic lineage, which are therefore not suitable for studying preadipocyte commitment. Conversely, over a dozen molecular players including transcription factors, genes, signalling pathways, epigenetic regulators, and microRNAs have been described to be involved in the differentiation of preadipocytes to adipocytes; however, only peroxisome proliferator-activated receptor gamma has proven to be clinically relevant. A detailed understanding of how the molecular players underpinning adipogenesis relate to adipose tissue function could provide new therapeutic approaches for addressing obesity without compromising adipose tissue function.
Collapse
Affiliation(s)
- Melvin A. Ambele
- Department of Immunology, and SAMRC Extramural Unit for Stem Cell Research and Therapy, Institute for Cellular and Molecular Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (M.A.A.); (P.D.); (R.G.)
- Department of Oral Pathology and Oral Biology, School of Dentistry, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa
| | - Priyanka Dhanraj
- Department of Immunology, and SAMRC Extramural Unit for Stem Cell Research and Therapy, Institute for Cellular and Molecular Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (M.A.A.); (P.D.); (R.G.)
| | - Rachel Giles
- Department of Immunology, and SAMRC Extramural Unit for Stem Cell Research and Therapy, Institute for Cellular and Molecular Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (M.A.A.); (P.D.); (R.G.)
| | - Michael S. Pepper
- Department of Immunology, and SAMRC Extramural Unit for Stem Cell Research and Therapy, Institute for Cellular and Molecular Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria 0001, South Africa; (M.A.A.); (P.D.); (R.G.)
| |
Collapse
|
19
|
Zohora FT, Aldebs AI, Nosoudi N, Singh SP, Ramirez-Vick JE. Gene Expression Profiling of Human Adipose Tissue Stem Cells during 2D versus 3D Adipogenesis. Cells Tissues Organs 2020; 208:113-133. [PMID: 32464628 DOI: 10.1159/000507187] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 03/10/2020] [Indexed: 11/19/2022] Open
Abstract
Much of the current understanding on molecular and cellular events of adipose developmental biology comes from monolayer cell culture models using preadipocyte cell lines, although in vivo adipose tissue consists of a much more complex three-dimensional microenvironment of diverse cell types, extracellular network, and tissue-specific morphological and functional features. Added to this fact, the preadipocytes, on which the adipogenesis mechanisms are mostly explored, possess some serious limitations (e.g., time of initial subculture and adipogenic differentiation time), which, perhaps, can efficiently be replaced with progenitor cells such as adipose tissue-derived stem cells (ASCs). With the objective of developing a better in vitro model for adipose developmental biology, this project involves gene expression profiling of human ASCs (hASCs) during their differentiation to adipocytes in a 2D versus 3D culture model. This transcriptional-level analysis revealed that gene expression patterns of adipogenesis-induced hASCs in a 3D self-assembled polypeptide hydrogel are relatively different from the 2D monolayered cells on plastic hard substrate. Moreover, analysis of adipogenic lineage progression 9 days after adipogenic induction shows earlier differentiation of hASCs in 2D over their 3D counterparts. However, differentiation in 2D shows some unexpected behavior in terms of gene expression, which does not seem to be related to adipogenic lineage specification. Since hASCs are already being used in clinical trials due to their therapeutic potential, it is important to have a clear understanding of the molecular mechanisms in an in vivo model microenvironment like the one presented here.
Collapse
Affiliation(s)
- Fatema Tuj Zohora
- Department of Biomedical, Industrial, and Human Factors Engineering, Wright State University, Dayton, Ohio, USA
| | - Alyaa Isam Aldebs
- Department of Biomedical, Industrial, and Human Factors Engineering, Wright State University, Dayton, Ohio, USA
| | - Nasim Nosoudi
- Biomedical Engineering Program,Marshall University, Huntington, West Virginia, USA
| | - Surinder Pal Singh
- CSIR-National Physical Laboratory, Dr. K.S. Krishanan Marg, New Delhi, India
| | - Jaime Eduardo Ramirez-Vick
- Department of Biomedical, Industrial, and Human Factors Engineering, Wright State University, Dayton, Ohio, USA,
| |
Collapse
|
20
|
Kawagishi-Hotta M, Hasegawa S, Igarashi T, Date Y, Ishii Y, Inoue Y, Hasebe Y, Yamada T, Arima M, Iwata Y, Kobayashi T, Nakata S, Sugiura K, Akamatsu H. Increase of gremlin 2 with age in human adipose-derived stromal/stem cells and its inhibitory effect on adipogenesis. Regen Ther 2019; 11:324-330. [PMID: 31709279 PMCID: PMC6831850 DOI: 10.1016/j.reth.2019.09.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 08/23/2019] [Accepted: 09/20/2019] [Indexed: 02/07/2023] Open
Abstract
Introduction Adipose-derived stromal/stem cells (ASCs) have attracted attention as a promising material for regenerative medicine. Previously, we reported an age-related decrease in the adipogenic potential of ASCs from human subjects and found that the individual difference in this potential increased with age, although the mechanisms remain unclear. Recently, other groups demonstrated that a secreted antagonist of bone morphogenetic protein (BMP) signaling, Gremlin 2 (GREM2), inhibits the differentiation of bone marrow-derived mesenchymal stem cells (BMSCs) into osteoblasts and the adipogenesis of 3T3-L1 cell. Here, we examined the effects of GREM2 on the differentiation of ASCs into adipocytes. Methods To examine changes in GREM2 expression levels with age, immunohistochemistry was performed on subcutaneous adipose tissues from subjects 12–97 years of age. Next, GREM2 gene expression levels in ASCs collected from subjects 5–90 years of age were examined by RT-PCR, and the change with age and correlation between the expression level and the adipogenic potential of ASCs were analyzed. In addition, to assess whether GREM2 affects adipogenesis, ASCs (purchased from a vendor) were cultured to induce adipogenesis with recombinant GREM2 protein, and siRNA-induced GREM2 knockdown experiment was also performed using aged ASCs. Results In adipose tissues, GREM2 expression was observed in cells, including ASCs, but not in mature adipocytes, and the expression level per cell increased with age. GREM2 expression levels in ASCs cultured in vitro also increased with age, and the individual differences in the level increased with age. Of note, partial correlation analysis controlled for age revealed that the adipogenic potential of ASCs and the GREM2 gene expression level were negatively correlated. Furthermore, based on a GREM2 addition experiment, GREM2 has inhibitory effects on the adipogenesis of ASCs through activation of Wnt/β-catenin signaling. On the other hand, GREM2 knockdown in aged ASCs promoted adipogenesis. Conclusions The GREM2 expression level was confirmed to play a role in the age-related decrease in adipogenic potential observed in ASCs isolated from adipose tissues as well as in the enhancement of the individual difference, which increased with age. GREM2 in adipose tissues increased with age, which suggested that GREM2 functions as an inhibitory factor of adipogenesis in ASCs. GREM2 in human adipose tissues increase with age. GREM2 expression in adipose-derived stromal/stem cells (ASCs) increased with age. In ASCs, adipogenic potential and GREM2 expression showed a negative correlation. Recombinant GREM2 inhibited the adipogenesis of ASCs. GREM2 knockdown in aged ASCs restored adipogenesis.
Collapse
Affiliation(s)
- Mika Kawagishi-Hotta
- Research Laboratories, Nippon Menard Cosmetic Co., Ltd, Japan.,Nagoya University-MENARD Collaborative Research Chair, Nagoya University Graduate School of Medicine, Japan.,Department of Applied Cell and Regenerative Medicine, Fujita Health University School of Medicine, Japan
| | - Seiji Hasegawa
- Research Laboratories, Nippon Menard Cosmetic Co., Ltd, Japan.,Nagoya University-MENARD Collaborative Research Chair, Nagoya University Graduate School of Medicine, Japan.,Department of Dermatology, Fujita Health University School of Medicine, Japan
| | - Toshio Igarashi
- Research Laboratories, Nippon Menard Cosmetic Co., Ltd, Japan
| | - Yasushi Date
- Research Laboratories, Nippon Menard Cosmetic Co., Ltd, Japan.,Nagoya University-MENARD Collaborative Research Chair, Nagoya University Graduate School of Medicine, Japan
| | - Yoshie Ishii
- Research Laboratories, Nippon Menard Cosmetic Co., Ltd, Japan.,Department of Applied Cell and Regenerative Medicine, Fujita Health University School of Medicine, Japan
| | - Yu Inoue
- Research Laboratories, Nippon Menard Cosmetic Co., Ltd, Japan.,Nagoya University-MENARD Collaborative Research Chair, Nagoya University Graduate School of Medicine, Japan
| | - Yuichi Hasebe
- Research Laboratories, Nippon Menard Cosmetic Co., Ltd, Japan.,Nagoya University-MENARD Collaborative Research Chair, Nagoya University Graduate School of Medicine, Japan
| | - Takaaki Yamada
- Research Laboratories, Nippon Menard Cosmetic Co., Ltd, Japan.,Department of Applied Cell and Regenerative Medicine, Fujita Health University School of Medicine, Japan.,Department of Dermatology, Fujita Health University School of Medicine, Japan
| | - Masaru Arima
- Department of Dermatology, Fujita Health University School of Medicine, Japan
| | - Yohei Iwata
- Department of Dermatology, Fujita Health University School of Medicine, Japan
| | - Tsukane Kobayashi
- Department of Dermatology, Fujita Health University School of Medicine, Japan
| | - Satoru Nakata
- Research Laboratories, Nippon Menard Cosmetic Co., Ltd, Japan
| | - Kazumitsu Sugiura
- Department of Dermatology, Fujita Health University School of Medicine, Japan
| | - Hirohiko Akamatsu
- Department of Applied Cell and Regenerative Medicine, Fujita Health University School of Medicine, Japan
| |
Collapse
|
21
|
Gao S, Guo X, Zhao S, Jin Y, Zhou F, Yuan P, Cao L, Wang J, Qiu Y, Sun C, Kang Z, Gao F, Xu W, Hu X, Yang D, Qin Y, Ning K, Shaw PJ, Zhong G, Cheng L, Zhu H, Gao Z, Chen X, Xu J. Differentiation of human adipose-derived stem cells into neuron/motoneuron-like cells for cell replacement therapy of spinal cord injury. Cell Death Dis 2019; 10:597. [PMID: 31395857 PMCID: PMC6687731 DOI: 10.1038/s41419-019-1772-1] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 04/24/2019] [Accepted: 05/31/2019] [Indexed: 01/06/2023]
Abstract
Human adipose-derived stem cells (hADSCs) are increasingly presumed to be a prospective stem cell source for cell replacement therapy in various degenerative and/or traumatic diseases. The potential of trans-differentiating hADSCs into motor neuron cells indisputably provides an alternative way for spinal cord injury (SCI) treatment. In the present study, a stepwise and efficient hADSC trans-differentiation protocol with retinoic acid (RA), sonic hedgehog (SHH), and neurotrophic factors were developed. With this protocol hADSCs could be converted into electrophysiologically active motoneuron-like cells (hADSC-MNs), which expressed both a cohort of pan neuronal markers and motor neuron specific markers. Moreover, after being primed for neuronal differentiation with RA/SHH, hADSCs were transplanted into SCI mouse model and they survived, migrated, and integrated into injured site and led to partial functional recovery of SCI mice. When ablating the transplanted hADSC-MNs harboring HSV-TK-mCherry overexpression system with antivirial Ganciclovir (GCV), functional relapse was detected by motor-evoked potential (MEP) and BMS assays, implying that transplanted hADSC-MNs participated in rebuilding the neural circuits, which was further confirmed by retrograde neuronal tracing system (WGA). GFP-labeled hADSC-MNs were subjected to whole-cell patch-clamp recording in acute spinal cord slice preparation and both action potentials and synaptic activities were recorded, which further confirmed that those pre-conditioned hADSCs indeed became functionally active neurons in vivo. As well, transplanted hADSC-MNs largely prevented the formation of injury-induced cavities and exerted obvious immune-suppression effect as revealed by preventing astrocyte reactivation and favoring the secretion of a spectrum of anti-inflammatory cytokines and chemokines. Our work suggests that hADSCs can be readily transformed into MNs in vitro, and stay viable in spinal cord of the SCI mouse and exert multi-therapeutic effects by rebuilding the broken circuitry and optimizing the microenvironment through immunosuppression.
Collapse
Affiliation(s)
- Shane Gao
- East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Xuanxuan Guo
- East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Simeng Zhao
- iHuman Institute, Shanghai Science and Technology University, Shanghai, 201210, China
| | - Yinpeng Jin
- Shanghai Public Health Clinical Center, Fudan University, JinShan, Shanghai, 201508, China
| | - Fei Zhou
- Department of Neurology, Third Affiliated Hospital of Navy Military Medical University, Shanghai, 200438, China
| | - Ping Yuan
- Tongji hospital affiliated to Tongji University, Tongji University School of Medicine, Shanghai, 200065, China
| | - Limei Cao
- Shanghai Eighth People's Hospital Affiliated to Jiangsu University, Shanghai, 200233, China
| | - Jian Wang
- East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Yue Qiu
- East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Chenxi Sun
- East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Zhanrong Kang
- Department of Orthopaedics, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 200137, China
| | - Fengjuan Gao
- Zhoupu hospital, Affiliated to Shanghai University of Medicine & Health Sciences, Shanghai, 201318, China
| | - Wei Xu
- Tongji hospital affiliated to Tongji University, Tongji University School of Medicine, Shanghai, 200065, China
| | - Xiao Hu
- Tongji hospital affiliated to Tongji University, Tongji University School of Medicine, Shanghai, 200065, China
| | - Danjing Yang
- East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Ying Qin
- East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Ke Ning
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385A Glossop Road, Sheffield, S10 2HQ, UK
| | - Pamela J Shaw
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385A Glossop Road, Sheffield, S10 2HQ, UK
| | - Guisheng Zhong
- iHuman Institute, Shanghai Science and Technology University, Shanghai, 201210, China.
| | - Liming Cheng
- Tongji hospital affiliated to Tongji University, Tongji University School of Medicine, Shanghai, 200065, China.
| | - Hongwen Zhu
- Tianjin Hospital, Tianjin, 300211, China. .,BOE Technology Group Co., Ltd., Beijing, 100176, China.
| | - Zhengliang Gao
- Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
| | - Xu Chen
- Shanghai Eighth People's Hospital Affiliated to Jiangsu University, Shanghai, 200233, China.
| | - Jun Xu
- East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| |
Collapse
|
22
|
Zhu Z, Xu L, Cai T, Yuan G, Sun N, Lu C, Qian R. Clock represses preadipocytes adipogenesis via GILZ. J Cell Physiol 2018; 233:6028-6040. [PMID: 29278648 DOI: 10.1002/jcp.26420] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 12/20/2017] [Indexed: 02/06/2023]
Abstract
Adiposity is a worldwide health threat that needs to be prevented. Circadian gene Clock (circadian locomotor output cycles kaput) is closely correlated to adiposity; for example, weight gain, adipocytes size expansion, and serum lipid level rise in ClockΔ19 mice compared to C57BL/6J mice. However, the precise role of Clock during adipogenic differentiation is unknown. Herein, the circadian gene Clock is shown to regulate adipogenesis mediated by GILZ. Clock-mediated attenuation and upregulation influenced lipid synthesis and affected the levels of adipogenic transcriptional factors, C/EBP-β, C/EBP-α, PPAR-γ, and FABP4, both in vivo and in vitro (primary adipose-derived stromal cells and 3T3-L1 cells). Chromatin immunoprecipitation (ChIP) assay, reporter gene assay, and serum shock assay found that Clock transcriptionally regulated the glucocorticoid-induced leucine zipper (GILZ). Furthermore, GILZ attenuation could relieve the inhibitory effect of Clock on lipid synthesis and GILZ overexpression also reduced the promotion role of Clock attenuation in adipogenesis suggesting that Clock inhibits adipogenic differentiation of preadipocytes via GILZ. The current results demonstrate how circadian genes are likely to regulate adiposity, affecting the adipogenic differentiation process, as well as, increasing the fat cells number. Therefore, this study may provide novel insights into the underlying mechanism explaining the correlation between Clock mutation and adiposity.
Collapse
Affiliation(s)
- Zhu Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China.,Department of BioBank, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lirong Xu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Tingting Cai
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Gongsheng Yuan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ning Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China.,Research Center on Aging and Medicine, Fudan University, Shanghai, China
| | - Chao Lu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China.,Research Center on Aging and Medicine, Fudan University, Shanghai, China
| | - Ruizhe Qian
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China.,Research Center on Aging and Medicine, Fudan University, Shanghai, China
| |
Collapse
|
23
|
Liu W, Yan X, Liu W, Wang Y, Rao Y, Yu H, Cui J, Xie X, Sun M, Yin L, Li H, Chen F. Alterations of protein glycosylation in embryonic stem cells during adipogenesis. Int J Mol Med 2017; 41:293-301. [PMID: 29115405 PMCID: PMC5746299 DOI: 10.3892/ijmm.2017.3240] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 10/25/2017] [Indexed: 11/06/2022] Open
Abstract
The understanding of adipose tissue development is crucial for the treatment of obesity-related diseases. Adipogenesis has been extensively investigated at the gene and protein levels in recent years. However, the alterations in protein glycosylation during this process remains unknown, particularly that of parthenogenetic embryonic stem cells (pESCs), a type of ESCs with low immunogenicity and no ethical concerns regarding their use. Protein glycosylation markedly affects cell growth and development, cell-to-cell communication, tumour growth and metastasis. In the present study, the adipogenic potentials of J1 ESCs and pESCs were first compared and the results demonstrated that pESCs had lower adipogenic potential compared with J1 ESCs. Lectin microarray was then used to screen the alteration of protein glycosylation during adipogenesis. The results revealed that protein modification of GlcNAc and α-1-2-fucosylation increased, whereas α-1-6‑fucosylation, α-2-6-sialylation and α-1-6-mannosylation decreased in J1 ESCs and pESCs during this process. In addition, α-1-3-mannosylation decreased only in pESCs. Lectin histochemistry and quantitative polymerase chain reaction of glycosyltransferase confirmed the results obtained by lectin microarray. Therefore, protein glycosylation of ESCs was significantly altered during adipogenesis, indicating that protein glycosylation analysis is not only helpful for studying the mechanism of adipogenesis, but may also be used as a marker to monitor adipogenic development.
Collapse
Affiliation(s)
- Wenguang Liu
- Faculty of Life Sciences, Northwest University, Xi'an, Shaanxi 710069, P.R. China
| | - Xingrong Yan
- Faculty of Life Sciences, Northwest University, Xi'an, Shaanxi 710069, P.R. China
| | - Wei Liu
- Faculty of Life Sciences, Northwest University, Xi'an, Shaanxi 710069, P.R. China
| | - Yangyang Wang
- Faculty of Life Sciences, Northwest University, Xi'an, Shaanxi 710069, P.R. China
| | - Yang Rao
- Faculty of Life Sciences, Northwest University, Xi'an, Shaanxi 710069, P.R. China
| | - Hanjie Yu
- Faculty of Life Sciences, Northwest University, Xi'an, Shaanxi 710069, P.R. China
| | - Jihong Cui
- Faculty of Life Sciences, Northwest University, Xi'an, Shaanxi 710069, P.R. China
| | - Xin Xie
- Faculty of Life Sciences, Northwest University, Xi'an, Shaanxi 710069, P.R. China
| | - Mei Sun
- Faculty of Life Sciences, Northwest University, Xi'an, Shaanxi 710069, P.R. China
| | - Lu Yin
- Faculty of Life Sciences, Northwest University, Xi'an, Shaanxi 710069, P.R. China
| | - Hongmin Li
- Faculty of Life Sciences, Northwest University, Xi'an, Shaanxi 710069, P.R. China
| | - Fulin Chen
- Faculty of Life Sciences, Northwest University, Xi'an, Shaanxi 710069, P.R. China
| |
Collapse
|
24
|
Differential expression profile of miRNAs in porcine muscle and adipose tissue during development. Gene 2017; 618:49-56. [DOI: 10.1016/j.gene.2017.04.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 03/20/2017] [Accepted: 04/07/2017] [Indexed: 12/25/2022]
|
25
|
Gao Y, Li J, Xu X, Wang S, Yang Y, Zhou J, Zhang L, Zheng F, Li X, Wang B. Embelin attenuates adipogenesis and lipogenesis through activating canonical Wnt signaling and inhibits high-fat diet-induced obesity. Int J Obes (Lond) 2017; 41:729-738. [PMID: 28163317 DOI: 10.1038/ijo.2017.35] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 01/25/2017] [Accepted: 01/29/2017] [Indexed: 12/17/2022]
Abstract
BACKGROUND Recent studies suggest that Embelin, a natural plant extract might have the potential to prevent body weight gain in rats. However, the mechanisms involved remain to be elucidated. METHODS Effects of Embelin on adipocyte differentiation and lipogenesis were studied in murine ST2 stromal cells and C3H10T1/2 mesenchymal cells. The mechanisms through which Embelin regulates adipogenic differentiation and lipogenesis were explored. The in vivo anti-obesity effects of Embelin in high-fat diet (HFD)-induced obesity mice and possible transcriptional impact were investigated. RESULTS Embelin treatment suppressed ST2 and C3H10T1/2 cells to proliferate, and differentiate into mature adipocytes, along with the inhibition of adipogenic factors peroxisome proliferator-activated receptor γ, CCAAT/enhancer binding protein-α, adipocyte protein 2 and adipsin. Embelin treatment also decreased the expression levels of lipogenic factors sterol regulatory element-binding protein 1, fatty acid synthase, acetyl-CoA carboxylase 1 and stearoyl-Coenzyme A desaturase 1. Embelin promoted the translocation of β-catenin from the cytoplasm into the nucleus in C3H10T1/2. The nuclear protein levels of β-catenin and TCF-4 were increased following Embelin treatment. Furthermore, Dickkopf-1 (Dkk1) expression was downregulated by Embelin, and overexpression of Dkk1 in C3H10T1/2 reversed the inhibition of adipogenesis and lipogenesis by Embelin. In vivo studies showed that Embelin treatment reduced the gain of body weight and fat, decreased the serum level of triglycerides, free fatty acid and total cholesterol, and improved glucose tolerance and insulin resistance in HFD-fed mice. Moreover, Embelin blocked induction of adipogenic and lipogenic factors and Dkk1 in adipose tissue in HFD-fed mice. CONCLUSIONS The present work provides evidences that Embelin is effective in inhibiting adipogenesis and lipogenesis in vitro and the mechanisms may involve canonical Wnt signaling. Embelin has the potential to prevent body weight gain and fat accumulation, and to improve obesity-related glucose tolerance impairment and insulin resistance in the HFD-fed mice.
Collapse
Affiliation(s)
- Y Gao
- Key Lab of Hormones and Development (Ministry of Health), Tianjin Key Lab of Metabolic Diseases, Metabolic Diseases Hospital and Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - J Li
- Key Lab of Hormones and Development (Ministry of Health), Tianjin Key Lab of Metabolic Diseases, Metabolic Diseases Hospital and Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - X Xu
- Key Lab of Hormones and Development (Ministry of Health), Tianjin Key Lab of Metabolic Diseases, Metabolic Diseases Hospital and Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - S Wang
- Department of Microbiology, Tianjin Medical University School of Basic Medical Sciences, Tianjin, China
| | - Y Yang
- Key Lab of Hormones and Development (Ministry of Health), Tianjin Key Lab of Metabolic Diseases, Metabolic Diseases Hospital and Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - J Zhou
- Key Lab of Hormones and Development (Ministry of Health), Tianjin Key Lab of Metabolic Diseases, Metabolic Diseases Hospital and Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - L Zhang
- Department of Microbiology, Tianjin Medical University School of Basic Medical Sciences, Tianjin, China
| | - F Zheng
- Department of Biochemistry, Tianjin University of Traditional Chinese Medicine School of Integrative Medicine, Tianjin, China
| | - X Li
- Department of Microbiology, Tianjin Medical University School of Basic Medical Sciences, Tianjin, China
| | - B Wang
- Key Lab of Hormones and Development (Ministry of Health), Tianjin Key Lab of Metabolic Diseases, Metabolic Diseases Hospital and Institute of Endocrinology, Tianjin Medical University, Tianjin, China.,2011 Collaborative Innovation Center for Metabolic Diseases, Metabolic Diseases Hospital, Tianjin Medical University, Tianjin, China
| |
Collapse
|
26
|
Heljasvaara R, Aikio M, Ruotsalainen H, Pihlajaniemi T. Collagen XVIII in tissue homeostasis and dysregulation - Lessons learned from model organisms and human patients. Matrix Biol 2016; 57-58:55-75. [PMID: 27746220 DOI: 10.1016/j.matbio.2016.10.002] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 09/12/2016] [Accepted: 10/10/2016] [Indexed: 12/13/2022]
Abstract
Collagen XVIII is a ubiquitous basement membrane (BM) proteoglycan produced in three tissue-specific isoforms that differ in their N-terminal non-collagenous sequences, but share collagenous and C-terminal non-collagenous domains. The collagenous domain provides flexibility to the large collagen XVIII molecules on account of multiple interruptions in collagenous sequences. Each isoform has a complex multi-domain structure that endows it with an ability to perform various biological functions. The long isoform contains a frizzled-like (Fz) domain with Wnt-inhibiting activity and a unique domain of unknown function (DUF959), which is also present in the medium isoform. All three isoforms share an N-terminal laminin-G-like/thrombospondin-1 sequence whose specific functions still remain unconfirmed. The proteoglycan nature of the isoforms further increases the functional diversity of collagen XVIII. An anti-angiogenic domain termed endostatin resides in the C-terminus of collagen XVIII and is proteolytically cleaved from the parental molecule during the BM breakdown for example in the process of tumour progression. Recombinant endostatin can efficiently reduce tumour angiogenesis and growth in experimental models by inhibiting endothelial cell migration and proliferation or by inducing their death, but its efficacy against human cancers is still a subject of debate. Mutations in the COL18A1 gene result in Knobloch syndrome, a genetic disorder characterised mainly by severe eye defects and encephalocele and, occasionally, other symptoms. Studies with gene-modified mice have elucidated some aspects of this rare disease, highlighting in particular the importance of collagen XVIII in the development of the eye. Research with model organisms have also helped in determining other structural and biological functions of collagen XVIII, such as its requirement in the maintenance of BM integrity and its emerging roles in regulating cell survival, stem or progenitor cell maintenance and differentiation and inflammation. In this review, we summarise current knowledge on the properties and endogenous functions of collagen XVIII in normal situations and tissue dysregulation. When data is available, we discuss the functions of the distinct isoforms and their specific domains.
Collapse
Affiliation(s)
- Ritva Heljasvaara
- Oulu Center for Cell-Matrix Research, Biocenter Oulu, Faculty of Biochemistry and Molecular Medicine, University of Oulu, FIN-90014 Oulu, Finland; Centre for Cancer Biomarkers CCBIO, Department of Biomedicine, University of Bergen, N-5009 Bergen, Norway.
| | - Mari Aikio
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Heli Ruotsalainen
- Oulu Center for Cell-Matrix Research, Biocenter Oulu, Faculty of Biochemistry and Molecular Medicine, University of Oulu, FIN-90014 Oulu, Finland
| | - Taina Pihlajaniemi
- Oulu Center for Cell-Matrix Research, Biocenter Oulu, Faculty of Biochemistry and Molecular Medicine, University of Oulu, FIN-90014 Oulu, Finland
| |
Collapse
|
27
|
Chen Y, Kim J, Zhang R, Yang X, Zhang Y, Fang J, Chen Z, Teng L, Chen X, Ge H, Atadja P, Li E, Chen T, Qi W. Histone Demethylase LSD1 Promotes Adipocyte Differentiation through Repressing Wnt Signaling. Cell Chem Biol 2016; 23:1228-1240. [DOI: 10.1016/j.chembiol.2016.08.010] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 07/26/2016] [Accepted: 08/10/2016] [Indexed: 02/06/2023]
|
28
|
Han M, Evsikov AV, Zhang L, Lastra-Vicente R, Linask KK. Embryonic exposures of lithium and homocysteine and folate protection affect lipid metabolism during mouse cardiogenesis and placentation. Reprod Toxicol 2016; 61:82-96. [DOI: 10.1016/j.reprotox.2016.03.039] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Revised: 03/10/2016] [Accepted: 03/11/2016] [Indexed: 02/09/2023]
|
29
|
Trans10, cis12 conjugated linoleic acid inhibits 3T3-L1 adipocyte adipogenesis by elevating β-catenin levels. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:363-70. [DOI: 10.1016/j.bbalip.2016.01.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 12/22/2015] [Accepted: 01/08/2016] [Indexed: 12/30/2022]
|
30
|
Ferrer-Lorente R, Bejar MT, Badimon L. Notch signaling pathway activation in normal and hyperglycemic rats differs in the stem cells of visceral and subcutaneous adipose tissue. Stem Cells Dev 2015; 23:3034-48. [PMID: 25035907 DOI: 10.1089/scd.2014.0070] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The precise mechanisms underlying the differential function and cardiometabolic risk of white adipose tissue (WAT) remain unclear. Visceral adipose tissue (VWAT) and subcutaneous adipose tissue (SCWAT) have different metabolic functions that seem to be ascribed to their different intrinsic expansion capacities. Here we have hypothesized that the WAT characteristics are determined by the resident adipose-derived stem cells (ASCs) found in the different WAT depots. Therefore, our objective has been to investigate adipogenesis in anatomically distinct fat depots. ASCs from five different WAT depots were characterized in both healthy lean and diabetic obese rats, showing significant differences in expression of some of genes governing the stemness and the earlier adipogenic differentiation steps. Notch-target genes [Hes (hairy and enhancer of split) and Hey (hairy/enhancer of split related with YRPW motif) families] were upregulated in ASCs derived from visceral depots. Upon adipogenic differentiation, adipocyte cell markers were downregulated in ASCs from VWAT in comparison to ASCs from SCWAT, revealing a lower adipogenic capacity in ASCs of visceral origin than in those of SCWAT in accordance with the differential activation of Notch signaling. Notch upregulation by its activator phenethyl isothiocyanate attenuated the adipogenic differentiation of ASCs from SCWAT whereas Notch inhibition by N-[N-(3,5-difluorophenacetyl-L-alanyl)]-S-phenylglycine t-butyl ester (DAPT) increased the adipogenic differentiation of ASCs from visceral origin. In conclusion, the differential activation of Notch in ASCs is the origin of the different intrinsic WAT expansion capacities that contribute to the regional variations in WAT homeostasis and to its associated cardiometabolic risk.
Collapse
Affiliation(s)
- Raquel Ferrer-Lorente
- 1 Cardiovascular Research Center, CSIC-ICCC , Hospital de la Santa Creu i Sant Pau (UAB), Barcelona, Spain
| | | | | |
Collapse
|
31
|
Lu H, Li X, Mu P, Qian B, Jiang W, Zeng L. Dickkopf-1 promotes the differentiation and adipocytokines secretion via canonical Wnt signaling pathway in primary cultured human preadipocytes. Obes Res Clin Pract 2015; 10:454-64. [PMID: 26383960 DOI: 10.1016/j.orcp.2015.08.016] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2015] [Revised: 08/24/2015] [Accepted: 08/29/2015] [Indexed: 10/23/2022]
Abstract
OBJECTIVE Dickkopf-1, a newly recognized antagonist for canonical Wnt signaling, is secreted in the early stage of human adipose-derived stem cells (ASCs) adipogenic differentiation. This study was aimed to investigate whether human recombinant DKK-1 (rhDKK-1) could affect the differentiation and metabolism as well as adipocytokines secretion in primary cultured human ASCs. METHODS Human ASCs were isolated from omental adipose tissue and induced to adipogenic differentiation in the absence or presence of Wnt signaling antagonist rhDKK-1 and agonist SB216763, respectively. mRNA and protein expression profiles of adipogenic factors during the differentiation process were analyzed using quantitative RT-PCR and Western blotting. Adipocytokines secretion levels in the culture medium were measured by ELISA method. RESULTS Our results showed that DKK-1 was already expressed during the early stage of adipogenesis and reached the peak on the 9th day. Exogenous rhDKK-1 exposure accelerated the differentiation by up-regulating PPAR-γ and C/EBP-α, down-regulating Wnt3a, Wnt10b and β-catenin, without affecting non-canonical Wnt signaling marker (Wnt5a). In addition, rhDKK-1 treatment increased the secretion of leptin, RBP4, TNF-α and adiponectin during differentiation. rhDKK-1 treatment also significantly increased the intracellular accumulation of lipids and lipolysis. Thus, Wnt signal pathway agonist SB216763 down-regulated DKK-1 transcriptional and secretion levels during adipogenic process. CONCLUSIONS Our results suggest that rhDKK-1 could promote ASCs differentiation and increase adipocytokines secretion via canonical Wnt signaling pathway.
Collapse
Affiliation(s)
- Hongyun Lu
- Department of Endocrinology & Metabolism, the Third Affiliated Hospital of Sun Yat-sen University, Guangdong, Guangzhou 510630, China; Department of Endocrinology & Metabolism, the Fifth Affiliated Hospital of Sun Yat-sen University, Guangdong, Zhuhai 519000, China
| | - Xiaofeng Li
- Department of Endocrinology & Metabolism, the Third Affiliated Hospital of Sun Yat-sen University, Guangdong, Guangzhou 510630, China
| | - Panwei Mu
- Department of Endocrinology & Metabolism, the Third Affiliated Hospital of Sun Yat-sen University, Guangdong, Guangzhou 510630, China
| | - Baiying Qian
- Department of Nephrology, the Fifth Affiliated Hospital of Sun Yat-sen University, Guangdong, Zhuhai 519000, China
| | - Wei Jiang
- Department of Endocrinology & Metabolism, the Third Affiliated Hospital of Sun Yat-sen University, Guangdong, Guangzhou 510630, China
| | - Longyi Zeng
- Department of Endocrinology & Metabolism, the Third Affiliated Hospital of Sun Yat-sen University, Guangdong, Guangzhou 510630, China.
| |
Collapse
|
32
|
Song K, Wang S, Mani M, Mani A. Wnt signaling, de novo lipogenesis, adipogenesis and ectopic fat. Oncotarget 2015; 5:11000-3. [PMID: 25526027 PMCID: PMC4294374 DOI: 10.18632/oncotarget.2769] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 11/14/2014] [Indexed: 11/25/2022] Open
Abstract
Wnt signaling is as a major regulator of adipogenesis. It differentially regulates the fate of mesenchymal stem cells (MSC) by promoting osteogenesis and myogenesis, and inhibiting adipogenesis[1]. Its loss of function has been associated with impaired osteogenesis[2] and diverse congenital and adult cardiovascular disorders[3,4]. Our group has identified loss of function mutations in Wnt coreceptor LRP6 that underlie autosomal dominant early onset coronary artery (CAD), osteoporosis and most features of the metabolic syndrome, including high plasma triglyceride and LDL-C, diabetes, hypertension, hyperuricemia and fatty liver disease (unpublished data). Following we will describe our most pertinent findings related to Wnt/LRP6 regulation of de novo lipogenesis and adipogenesis and the role of impaired Wnt signaling in generation of ectopic fat, insulin resistance, elevated plasma lipids and non-alcoholic fatty liver disease (NAFLD).
Collapse
Affiliation(s)
- Kangxing Song
- Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT
| | - Shuxia Wang
- Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT
| | | | - Arya Mani
- Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT. Department of Genetics, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
33
|
Developmental Stage-Dependent Effects of Leukemia Inhibitory Factor on Adipocyte Differentiation of Murine Bone Marrow Stromal Cells. Cell Biochem Biophys 2015; 74:11-7. [DOI: 10.1007/s12013-015-0703-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
34
|
In silico identification of conserved microRNAs and their targets in bovine fat tissue. Gene 2015; 559:119-28. [PMID: 25626178 DOI: 10.1016/j.gene.2015.01.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 01/09/2015] [Accepted: 01/11/2015] [Indexed: 12/22/2022]
Abstract
MicroRNAs (miRNAs) represent a newly identified class of non-protein-coding ~22 nt small RNA which plays important roles in multiple biological processes by degrading targeted mRNA or repressing mRNA translation. Here we present EST (expressed sequence tags)-based and GSS (Genomic Survey Sequences)-based combined approach for the detection of conserved miRNAs of cattle. A total of 20 conserved miRNAs that belong to 18 miRNA families were detected following a range of filtering criteria; their functions were further predicted and analyzed. To confirm our prediction, a miRNA-detecting microarray was designed with probes complementary to previously known mature miRNA sequences from 131 organisms. After hybridizing with small RNAs extracted from beef cattle subcutaneous fat tissue, 219 (32.30%) miRNAs were detected in the 679 known Bos taurus miRNAs and all the miRNAs predicted above were also detected. Conformation of 22 most abundant miRNA expression by qRT-PCR indicated that they were highly accumulated not only in subcutaneous fat tissue but also in intramuscular fat tissue. Bioinformatics of KEGG pathway analysis suggested that 4 differential expression miRNAs (miR-143, miR-145, miR-2325c and miR-2361) involved in different pathways and target genes may regulate the fat deposition differently. Taken together, our results expand the number of known bovine miRNAs and provide a thorough account of the miRNA transcriptome in bovine adipose tissue.
Collapse
|
35
|
Retraction. Prenatal PFOA exposure alters gene expression pathways in murine mammary gland. Toxicol Sci 2014; 145:211. [PMID: 25490953 DOI: 10.1093/toxsci/kfu253] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
36
|
Popov B, Petrov N. pRb-E2F signaling in life of mesenchymal stem cells: Cell cycle, cell fate, and cell differentiation. Genes Dis 2014; 1:174-187. [PMID: 30258863 PMCID: PMC6150080 DOI: 10.1016/j.gendis.2014.09.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 09/14/2014] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent cells that can differentiate into various mesodermal lines forming fat, muscle, bone, and other lineages of connective tissue. MSCs possess plasticity and under special metabolic conditions may transform into cells of unusual phenotypes originating from ecto- and endoderm. After transplantation, MSCs release the humoral factors promoting regeneration of the damaged tissue. During last five years, the numbers of registered clinical trials of MSCs have increased about 10 folds. This gives evidence that MSCs present a new promising resource for cell therapy of the most dangerous diseases. The efficacy of the MSCs therapy is limited by low possibilities to regulate their conversion into cells of damaged tissues that is implemented by the pRb-E2F signaling. The widely accepted viewpoint addresses pRb as ubiquitous regulator of cell cycle and tumor suppressor. However, current publications suggest that basic function of the pRb-E2F signaling in development is to regulate cell fate and differentiation. Through facultative and constitutive chromatin modifications, pRb-E2F signaling promotes transient and stable cells quiescence, cell fate choice to differentiate, to senesce, or to die. Loss of pRb is associated with cancer cell fate. pRb regulates cell fate by retaining quiescence of one cell population in favor of commitment of another or by suppression of genes of different cell phenotype. pRb is the founder member of the "pocket protein" family possessing functional redundancy. Critical increase in the efficacy of the MSCs based cell therapy will depend on precise understanding of various aspects of the pRb-E2F signaling.
Collapse
Affiliation(s)
- Boris Popov
- Institute of Cytology, Russian Academy of Sciences, St.Petersburg, 4, Tikhoretsky Av., 194064, Russia
| | | |
Collapse
|
37
|
MYC is an early response regulator of human adipogenesis in adipose stem cells. PLoS One 2014; 9:e114133. [PMID: 25437437 PMCID: PMC4250176 DOI: 10.1371/journal.pone.0114133] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 11/02/2014] [Indexed: 12/13/2022] Open
Abstract
Adipose stem cell (ASC) differentiation is necessary for the proper maintenance and function of adipose tissue. The procurement and characterization of multipotent ASCs has enabled investigation into the molecular determinants driving human adipogenesis. Here, the transcription factor MYC was identified as a significant regulator of ASC differentiation. Expression of MYC transcript and protein was found to accumulate during the initial course of differentiation. Loss-of-function analysis using siRNA mediated knockdown of MYC demonstrated inhibition of hormonally stimulated adipogenesis. MYC exhibited an early and sustained expression pattern that preceded down regulation of key suppressor genes, as well as induction of transcriptional and functional effectors. Glucocorticoid stimulation was identified as a necessary component for MYC induction and was found to impact adipogenesis in a concentration-dependent manner. Global gene expression analysis of MYC knockdown in ASC enriched for functional pathways related to cell adhesion, cytoskeletal remodeling, and transcriptional components of adipogenesis. These results identify a functional role for MYC in promotion of multipotent ASC to the adipogenic lineage.
Collapse
|
38
|
Kamata M, Okitsu Y, Fujiwara T, Kanehira M, Nakajima S, Takahashi T, Inoue A, Fukuhara N, Onishi Y, Ishizawa K, Shimizu R, Yamamoto M, Harigae H. GATA2 regulates differentiation of bone marrow-derived mesenchymal stem cells. Haematologica 2014; 99:1686-96. [PMID: 25150255 DOI: 10.3324/haematol.2014.105692] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The bone marrow microenvironment comprises multiple cell niches derived from bone marrow mesenchymal stem cells. However, the molecular mechanism of bone marrow mesenchymal stem cell differentiation is poorly understood. The transcription factor GATA2 is indispensable for hematopoietic stem cell function as well as other hematopoietic lineages, suggesting that it may maintain bone marrow mesenchymal stem cells in an immature state and also contribute to their differentiation. To explore this possibility, we established bone marrow mesenchymal stem cells from GATA2 conditional knockout mice. Differentiation of GATA2-deficient bone marrow mesenchymal stem cells into adipocytes induced accelerated oil-drop formation. Further, GATA2 loss- and gain-of-function analyses based on human bone marrow mesenchymal stem cells confirmed that decreased and increased GATA2 expression accelerated and suppressed bone marrow mesenchymal stem cell differentiation to adipocytes, respectively. Microarray analysis of GATA2 knockdowned human bone marrow mesenchymal stem cells revealed that 90 and 189 genes were upregulated or downregulated by a factor of 2, respectively. Moreover, gene ontology analysis revealed significant enrichment of genes involved in cell cycle regulation, and the number of G1/G0 cells increased after GATA2 knockdown. Concomitantly, cell proliferation was decreased by GATA2 knockdown. When GATA2 knockdowned bone marrow mesenchymal stem cells as well as adipocytes were cocultured with CD34-positive cells, hematopoietic stem cell frequency and colony formation decreased. We confirmed the existence of pathological signals that decrease and increase hematopoietic cell and adipocyte numbers, respectively, characteristic of aplastic anemia, and that suppress GATA2 expression in hematopoietic stem cells and bone marrow mesenchymal stem cells.
Collapse
Affiliation(s)
- Mayumi Kamata
- Departments of Hematology and Rheumatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yoko Okitsu
- Departments of Hematology and Rheumatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tohru Fujiwara
- Departments of Hematology and Rheumatology, Tohoku University Graduate School of Medicine, Sendai, Japan Molecular Hematology/Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masahiko Kanehira
- Departments of Hematology and Rheumatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shinji Nakajima
- Departments of Hematology and Rheumatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Taro Takahashi
- Departments of Hematology and Rheumatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Ai Inoue
- Departments of Hematology and Rheumatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Noriko Fukuhara
- Departments of Hematology and Rheumatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yasushi Onishi
- Departments of Hematology and Rheumatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kenichi Ishizawa
- Departments of Hematology and Rheumatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Ritsuko Shimizu
- Molecular Hematology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masayuki Yamamoto
- Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hideo Harigae
- Departments of Hematology and Rheumatology, Tohoku University Graduate School of Medicine, Sendai, Japan Molecular Hematology/Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
39
|
Saben J, Thakali KM, Lindsey FE, Zhong Y, Badger TM, Andres A, Shankar K. Distinct adipogenic differentiation phenotypes of human umbilical cord mesenchymal cells dependent on adipogenic conditions. Exp Biol Med (Maywood) 2014; 239:1340-51. [PMID: 24951473 DOI: 10.1177/1535370214539225] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The umbilical cord (UC) matrix is a source of multipotent mesenchymal stem cells (MSCs) that have adipogenic potential and thus can be a model to study adipogenesis. However, existing variability in adipocytic differentiation outcomes may be due to discrepancies in methods utilized for adipogenic differentiation. Additionally, functional characterization of UCMSCs as adipocytes has not been described. We tested the potential of three well-established adipogenic cocktails containing IBMX, dexamethasone, and insulin (MDI) plus indomethacin (MDI-I) or rosiglitazone (MDI-R) to stimulate adipocyte differentiation in UCMSCs. MDI, MDI-I, and MDI-R treatment significantly increased peroxisome proliferator-activated receptor gamma (PPARγ) and CCAAT-enhancer binding protein alpha (C/EBPα) mRNA and induced lipid droplet formation. However, MDI-I had the greatest impact on mRNA expression of PPARγ, C/EBPα, FABP4, GPD1, PLIN1, PLIN2, and ADIPOQ and lipid accumulation, whereas MDI showed the least. Interestingly, there were no treatment group differences in the amount of PPARγ protein. However, MDI-I treated cells had significantly more C/EBPα protein compared to MDI or MDI-R, suggesting that indomethacin-dependent increased C/EBPα may contribute to the adipogenesis-inducing potency of MDI-I. Additionally, bone morphogenetic protein 4 (BMP4) treatment of UCMSCs did not enhance responsiveness to MDI-induced differentiation. Finally to characterize adipocyte function, differentiated UCMSCs were stimulated with insulin and downstream signaling was assessed. Differentiated UCMSCs were responsive to insulin at two weeks but showed decreased sensitivity by five weeks following differentiation, suggesting that long-term differentiation may induce insulin resistance. Together, these data indicate that UCMSCs undergo adipogenesis when differentiated in MDI, MDI-I, and MDI-R, however the presence of indomethacin greatly enhances their adipogenic potential beyond that of rosiglitazone. Furthermore, our results suggest that insulin signaling pathways of differentiated UCMSCs are functionally similar to adipocytes.
Collapse
Affiliation(s)
- Jessica Saben
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA
| | - Keshari M Thakali
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA
| | - Forrest E Lindsey
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA
| | - Ying Zhong
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA
| | - Thomas M Badger
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA
| | - Aline Andres
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA
| | - Kartik Shankar
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA
| |
Collapse
|
40
|
Abstract
Adipocyte differentiation, termed adipogenesis, is a complicated process in which pluripotent mesenchymal stem cells differentiate into mature adipocytes. The process of adipocyte differentiation is tightly regulated by a number of transcription factors, hormones and signaling pathway molecules. Recent studies have demonstrated that microRNAs, which belong to small noncoding RNA species, are also involved in adipocyte differentiation. In vivo and in vitro studies have revealed that various microRNAs affect adipogenesis by targeting several adipogenic transcription factors and key signaling molecules. In this review, we will summarize the roles of microRNAs in adipogenesis and their target genes associated with each stage of adipocyte differentiation.
Collapse
Affiliation(s)
- You Hwa Son
- Seoul National University School of Biological Sciences, Seoul National University, Seoul, Korea
| | - Sojeong Ka
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Korea
| | - A Young Kim
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Korea
| | - Jae Bum Kim
- Seoul National University School of Biological Sciences, Seoul National University, Seoul, Korea
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Korea
| |
Collapse
|
41
|
Lafontan M. Adipose tissue and adipocyte dysregulation. DIABETES & METABOLISM 2014; 40:16-28. [DOI: 10.1016/j.diabet.2013.08.002] [Citation(s) in RCA: 115] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Accepted: 08/25/2013] [Indexed: 12/19/2022]
|
42
|
Klein SM, Vykoukal J, Prantl L, Dolderer JH. Tissue Engineering of Vascularized Adipose Tissue for Soft Tissue Reconstruction. STEM CELLS IN AESTHETIC PROCEDURES 2014:23-40. [DOI: 10.1007/978-3-642-45207-9_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
43
|
Murdolo G, Bartolini D, Tortoioli C, Piroddi M, Iuliano L, Galli F. Lipokines and oxysterols: novel adipose-derived lipid hormones linking adipose dysfunction and insulin resistance. Free Radic Biol Med 2013; 65:811-820. [PMID: 23954331 DOI: 10.1016/j.freeradbiomed.2013.08.007] [Citation(s) in RCA: 45] [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: 06/11/2013] [Revised: 08/07/2013] [Accepted: 08/08/2013] [Indexed: 02/07/2023]
Abstract
The expansion of adipose tissue (AT) is, by definition, a hallmark of obesity. However, not all increases in fat mass are associated with pathophysiological cues. Indeed, whereas a "healthy" fat mass accrual, mainly in the subcutaneous depots, preserves metabolic homeostasis, explaining the occurrence of the metabolically healthy obese phenotype, "unhealthy" AT expansion is importantly associated with insulin resistance/type 2 diabetes and the metabolic syndrome. The development of a dysfunctional adipose organ may find mechanistic explanation in a reduced ability to recruit new and functional (pre)adipocytes from undifferentiated precursor cells. Such a failure of the adipogenic process underlies the "AT expandability" paradigm. The inability of AT to expand further to store excess nutrients, rather than obesity per se, induces a diabetogenic milieu by promoting the overflow and the ectopic deposition of fatty acids in insulin-dependent organs (i.e., lipotoxicity), the secretion of various metabolically detrimental adipose-derived hormones (i.e., adipokines and lipokines), and the occurrence of local and systemic inflammation and oxidative stress. Hitherto, fatty acids (i.e., lipokines) and the oxidation by-products of cholesterol and polyunsaturated fatty acids, such as nonenzymatic oxysterols and reactive aldehyde species, respectively, emerge as key modulators of (pre)adipocyte signaling through Wnt/β-catenin and MAPK pathways and potential regulators of glucose homeostasis. These and other mechanistic insights linking adipose dysfunction, oxidative stress, and impairment of glucose homeostasis are discussed in this review article, which focuses on adipose peroxidation as a potential instigator of, and a putative therapeutic target for, obesity-associated metabolic dysfunctions.
Collapse
Affiliation(s)
- Giuseppe Murdolo
- Department of Internal Medicine, Assisi Hospital, I-06081 Assisi, Perugia, Italy; Section of Internal Medicine, Endocrine, and Metabolic Sciences, Italy.
| | - Desirée Bartolini
- Section of Applied Biochemistry and Nutritional Sciences, Department of Internal Medicine, Perugia University, Perugia, Italy
| | | | - Marta Piroddi
- Section of Applied Biochemistry and Nutritional Sciences, Department of Internal Medicine, Perugia University, Perugia, Italy
| | - Luigi Iuliano
- Unit of Vascular Medicine, Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Francesco Galli
- Section of Applied Biochemistry and Nutritional Sciences, Department of Internal Medicine, Perugia University, Perugia, Italy
| |
Collapse
|
44
|
Bethel M, Chitteti BR, Srour EF, Kacena MA. The changing balance between osteoblastogenesis and adipogenesis in aging and its impact on hematopoiesis. Curr Osteoporos Rep 2013; 11:99-106. [PMID: 23423562 PMCID: PMC3643998 DOI: 10.1007/s11914-013-0135-6] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Osteoblasts (OBs) and adipocytes (APs) share a common mesenchymal ancestor. It is now clear that mesenchymal stem cell (MSC) maturation along the OB lineage comes at the expense of adipogenesis and vice versa. During aging, this balance increasingly favors the formation of APs. Hematopoiesis also slowly declines during the aging process. The role of OB lineage cells in hematopoiesis has been studied, but less is known about how APs regulate hematopoiesis. A few studies have demonstrated a negative relationship between APs and hematopoiesis; however, there is also evidence that brown adipose tissue (BAT) may promote hematopoiesis. This review will examine the current knowledge of how adipogenesis and osteogenesis change with aging and the implications of this changing environment on hematopoeisis.
Collapse
Affiliation(s)
- Monique Bethel
- Postdoctoral Fellow, Department of Orthopaedic Surgery, Indiana University School of Medicine, 1120 South Drive, FH 115, Indianapolis, IN 46202, 317-278-2804 (phone), 317-278-9568 (fax),
| | - Brahmananda R. Chitteti
- Postdoctoral Fellow, Department of Medicine. Indiana University School of Medicine, 980 W. Walnut Street, R3-C356, Indianapolis, IN 46202, 317-274-0352 (phone), 317-274-0396 (fax),
| | - Edward F. Srour
- Robert J. and Annie S. Rohn Professor of Leukemia Research, Departments of Medicine, Pediatrics, Microbiology and Immunology. Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, 980 W. Walnut Street, R3-C312, Indianapolis, IN 46202, 317-274-0343 (phone), 317-274-0396 (fax),
| | - Melissa A. Kacena
- Assistant Professor, Department of Orthopaedic Surgery, Indiana University School of Medicine, 1120 South Drive, FH 115H, Indianapolis, IN 46202, 317-2783482 (phone), 317-278-9568 (Fax),
| |
Collapse
|
45
|
Wang H, Zheng Y, Wang G, Li H. Identification of microRNA and bioinformatics target gene analysis in beef cattle intramuscular fat and subcutaneous fat. MOLECULAR BIOSYSTEMS 2013; 9:2154-62. [PMID: 23728155 DOI: 10.1039/c3mb70084d] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
MicroRNA (miRNA) is endogenous non-coding RNA that has been proposed to play an important role in the formation of body fat. However, the differential expression of miRNA and the role of these miRNAs in bovine intramuscular and subcutaneous fat tissue are still unknown. In this study, the profile of differentially expressed miRNA and the target gene analysis in intramuscular adipose and subcutaneous adipose of adult beef cattle were investigated by microarray and bioinformatics. The data identified 88 differentially expressed miRNAs in 213 miRNAs which were detected on the microarray, and 30 miRNAs among these 88 miRNAs were changed significantly between intramuscular and subcutaneous fat (fold change >1, P < 0.001). miR-143, miR-145, miR-26a, miR-2373-5p and miR-23b-3p were highly expressed in intramuscular fat, whilst miR-26a, miR-2373-5p, miR-2325c, miR-3613 and miR-2361 showed highest abundance in subcutaneous fat. Bioinformatics of KEGG pathway analysis and GO term enrichment suggested that these differentially expressed miRNAs involved in different pathways and target genes may regulate differently the fat deposition. Taken together, our study provides the first evidence for better understanding the differential expression and mechanisms of miRNA in bovine fat deposition, and provides thinking to improve the quality of beef by reducing subcutaneous fat and increasing intramuscular fat of beef cattle.
Collapse
Affiliation(s)
- HaiYang Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | | | | | | |
Collapse
|
46
|
Hoffman MD, Benoit DSW. Agonism of Wnt-β-catenin signalling promotes mesenchymal stem cell (MSC) expansion. J Tissue Eng Regen Med 2013; 9:E13-26. [PMID: 23554411 DOI: 10.1002/term.1736] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Revised: 01/09/2013] [Accepted: 01/30/2013] [Indexed: 12/17/2022]
Abstract
Promoting mesenchymal stem cell (MSC) proliferation has numerous applications in stem cell therapies, particularly in the area of regenerative medicine. In order for cell-based regenerative approaches to be realized, MSC proliferation must be achieved in a controlled manner without compromising stem cell differentiation capacities. Here we demonstrate that 6-bromoindirubin-3'-oxime (BIO) increases MSC β-catenin activity 106-fold and stem cell-associated gene expression ~33-fold, respectively, over untreated controls. Subsequently, BIO treatment increases MSC populations 1.8-fold in typical 2D culture conditions, as well as 1.3-fold when encapsulated within hydrogels compared to untreated cells. Furthermore, we demonstrate that BIO treatment does not reduce MSC multipotency where MSCs maintain their ability to differentiate into osteoblasts, chondrocytes and adipocytes using standard conditions. Taken together, our results demonstrate BIO's potential utility as a proliferative agent for cell transplantation and tissue regeneration.
Collapse
Affiliation(s)
- Michael D Hoffman
- Departments of Biomedical Engineering, Chemical Engineering and Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Danielle S W Benoit
- Departments of Biomedical Engineering, Chemical Engineering and Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
47
|
Zhou H, Kaplan T, Li Y, Grubisic I, Zhang Z, Wang PJ, Eisen MB, Tjian R. Dual functions of TAF7L in adipocyte differentiation. eLife 2013; 2:e00170. [PMID: 23326641 PMCID: PMC3539393 DOI: 10.7554/elife.00170] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2012] [Accepted: 11/09/2012] [Indexed: 12/22/2022] Open
Abstract
The diverse transcriptional mechanisms governing cellular differentiation and development of mammalian tissue remains poorly understood. Here we report that TAF7L, a paralogue of TFIID subunit TAF7, is enriched in adipocytes and white fat tissue (WAT) in mouse. Depletion of TAF7L reduced adipocyte-specific gene expression, compromised adipocyte differentiation, and WAT development as well. Ectopic expression of TAF7L in myoblasts reprograms these muscle precursors into adipocytes upon induction. Genome-wide mRNA-seq expression profiling and ChIP-seq binding studies confirmed that TAF7L is required for activating adipocyte-specific genes via a dual mechanism wherein it interacts with PPARγ at enhancers and TBP/Pol II at core promoters. In vitro binding studies confirmed that TAF7L forms complexes with both TBP and PPARγ. These findings suggest that TAF7L plays an integral role in adipocyte gene expression by targeting enhancers as a cofactor for PPARγ and promoters as a component of the core transcriptional machinery.DOI:http://dx.doi.org/10.7554/eLife.00170.001.
Collapse
Affiliation(s)
- Haiying Zhou
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, United States
- Li Ka Shing Center For Biomedical and Health Sciences, CIRM Center of Excellence, University of California, Berkeley, Berkeley, United States
| | - Tommy Kaplan
- Department of Molecular and Cell Biology, California Institute of Quantitative Biosciences, University of California, Berkeley, Berkeley, United States
- School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yan Li
- Janelia Farm Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Ivan Grubisic
- Li Ka Shing Center For Biomedical and Health Sciences, CIRM Center of Excellence, University of California, Berkeley, Berkeley, United States
- UC Berkeley-UCSF Graduate Program in Bioengineering, Department of Molecular and Cell Biology, California Institute of Quantitative Biosciences, University of California, Berkeley, Berkeley, United States
| | - Zhengjian Zhang
- Janelia Farm Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - P Jeremy Wang
- Department of Animal Biology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, United States
| | - Michael B Eisen
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, United States
- Department of Molecular and Cell Biology, California Institute of Quantitative Biosciences, University of California, Berkeley, Berkeley, United States
| | - Robert Tjian
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, United States
- Li Ka Shing Center For Biomedical and Health Sciences, CIRM Center of Excellence, University of California, Berkeley, Berkeley, United States
| |
Collapse
|
48
|
Fujikura J, Hosoda K, Nakao K. Cell transplantation therapy for diabetes mellitus: endocrine pancreas and adipocyte. Endocr J 2013; 60:697-708. [PMID: 23719783 DOI: 10.1507/endocrj.ej13-0162] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Experimental transplantation of endocrine tissues has led to significant advances in our understanding of endocrinology and metabolism. Endocrine cell transplantation therapy is expected to be applied to the treatment of metabolic endocriopathies. Restoration of functional pancreatic beta-cell mass or of functional adipose mass are reasonable treatment approaches for patients with diabetes or lipodystrophy, respectively. Human induced pluripotent stem (iPS) cell research is having a great impact on life sciences. Doctors Takahashi and Yamanaka discovered that the forced expression of a set of genes can convert mouse and human somatic cells into a pluripotent state [1, 2]. These iPS cells can differentiate into a variety of cell types. Therefore, iPS cells from patients may be a potential cell source for autologous cell replacement therapy. This review briefly summarizes the current knowledge about transplantation therapy for diabetes mellitus, the development of the endocrine pancreas and adipocytes, and endocrine-metabolic disease-specific iPS cells.
Collapse
Affiliation(s)
- Junji Fujikura
- Division of Endocrinology and Metabolism, Kyoto University Hospital, Kyoto 606-8507, Japan.
| | | | | |
Collapse
|
49
|
Murdolo G, Piroddi M, Luchetti F, Tortoioli C, Canonico B, Zerbinati C, Galli F, Iuliano L. Oxidative stress and lipid peroxidation by-products at the crossroad between adipose organ dysregulation and obesity-linked insulin resistance. Biochimie 2012; 95:585-94. [PMID: 23274128 DOI: 10.1016/j.biochi.2012.12.014] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Accepted: 12/13/2012] [Indexed: 12/11/2022]
Abstract
Obesity has been proposed as an energy balance disorder in which the expansion of adipose tissue (AT) leads to unfavorable health outcomes. Even though adiposity represents the most powerful driving force for the development of insulin resistance (IR) and type 2 diabetes, mounting evidence points to "adipose dysregulation", rather than fat mass accrual per se, as a key pathophysiological trigger of the obesity-linked metabolic complications. The dysfunctional fat, besides hypertrophic adipose cells and inflammatory cues, displays a reduced ability to form new adipocytes from the undifferentiated precursor cells (ie, the preadipocytes). The failure of adipogenesis poses a "diabetogenic" milieu either by promoting the ectopic overflow/deposition of lipids in non-adipose targets (lipotoxicity) or by inducing a dysregulated secretion of different adipose-derived hormones (ie, adipokines and lipokines). This novel and provocative paradigm ("expandability hypothesis") further extends current "adipocentric view" implicating a reduced adipogenic capacity as a missing link between "unhealthy" fat expansion and impairment of metabolic homeostasis. Hitherto, reactive oxygen species have been implicated in multiple forms of IR. However, the effects of stress on adipogenesis remain controversial. Compelling circumstantial data indicate that lipid peroxidation by-products (ie, oxysterols and 4-hydrononenal) may detrimentally affect adipose homeostasis partly by impairing (pre)adipocyte differentiation. In this scenario, it is tempting to speculate that a fine tuning of the adipose redox status may provide new mechanistic insights at the interface between fat dysregulation and development of metabolic dysfunctions. Yet, in humans, the molecular "signatures" of oxidative stress in the dysregulated fat as well as the pathophysiological effects of adipose (per)oxidation on glucose homeostasis remain poorly investigated. In this review we will summarize the potential mechanisms by which increased oxidative stress in fat may impair (pre)adipocyte differentiation and promote the adipose dysfunction. We will also attempt to highlight the conundrum with the adipose redox changes and the regulation of glucose homeostasis. Finally, we will briefly discuss the scientific rationale for proposing the adipose redox state as a potential target for novel therapeutic strategies to curb/prevent adiposity-linked insulin resistance.
Collapse
Affiliation(s)
- Giuseppe Murdolo
- Department of Internal Medicine, Assisi Hospital, Via Valentin Muller 1, Assisi, Perugia, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Pelled G, Snedeker JG, Ben-Arav A, Rigozzi S, Zilberman Y, Kimelman-Bleich N, Gazit Z, Müller R, Gazit D. Smad8/BMP2-engineered mesenchymal stem cells induce accelerated recovery of the biomechanical properties of the Achilles tendon. J Orthop Res 2012; 30:1932-9. [PMID: 22696396 PMCID: PMC3479351 DOI: 10.1002/jor.22167] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2011] [Accepted: 05/21/2012] [Indexed: 02/04/2023]
Abstract
Tendon tissue regeneration is an important goal for orthopedic medicine. We hypothesized that implantation of Smad8/BMP2-engineered MSCs in a full-thickness defect of the Achilles tendon (AT) would induce regeneration of tissue with improved biomechanical properties. A 2 mm defect was created in the distal region of murine ATs. The injured tendons were then sutured together or given implants of genetically engineered MSCs (GE group), non-engineered MSCs (CH3 group), or fibrin gel containing no cells (FG group). Three weeks later the mice were killed, and their healing tendons were excised and processed for histological or biomechanical analysis. A biomechanical analysis showed that tendons that received implants of genetically engineered MSCs had the highest effective stiffness (>70% greater than natural healing, p < 0.001) and elastic modulus. There were no significant differences in either ultimate load or maximum stress among the treatment groups. Histological analysis revealed a tendon-like structure with elongated cells mainly in the GE group. ATs that had been implanted with Smad8/BMP2-engineered stem cells displayed a better material distribution and functional recovery than control groups. While additional study is required to determine long-term effects of GE MSCs on tendon healing, we conclude that genetically engineered MSCs may be a promising therapeutic tool for accelerating short-term functional recovery in the treatment of tendon injuries.
Collapse
Affiliation(s)
- Gadi Pelled
- Skeletal Biotech Lab, Faculty of Dental Medicine, Hebrew University of Jerusalem, Jerusalem, Israel,Skeletal Regeneration and Stem Cell Therapy Laboratory, Department of Surgery and Cedars-Sinai Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Jess G. Snedeker
- Laboratory for Orthopedic Research, Department of Orthopedics, University of Zurich, Balgrist, Zurich, Switzerland,Institute for Biomechanics, Department of Mechanical Engineering, Swiss Federal Institute of Technology (ETH) Zürich, Switzerland
| | - Ayelet Ben-Arav
- Skeletal Biotech Lab, Faculty of Dental Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Samuela Rigozzi
- Institute for Biomechanics, Department of Mechanical Engineering, Swiss Federal Institute of Technology (ETH) Zürich, Switzerland
| | - Yoram Zilberman
- Skeletal Biotech Lab, Faculty of Dental Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Nadav Kimelman-Bleich
- Skeletal Biotech Lab, Faculty of Dental Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Zulma Gazit
- Skeletal Biotech Lab, Faculty of Dental Medicine, Hebrew University of Jerusalem, Jerusalem, Israel,Skeletal Regeneration and Stem Cell Therapy Laboratory, Department of Surgery and Cedars-Sinai Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Ralph Müller
- Institute for Biomechanics, Department of Mechanical Engineering, Swiss Federal Institute of Technology (ETH) Zürich, Switzerland
| | - Dan Gazit
- Skeletal Biotech Lab, Faculty of Dental Medicine, Hebrew University of Jerusalem, Jerusalem, Israel,Skeletal Regeneration and Stem Cell Therapy Laboratory, Department of Surgery and Cedars-Sinai Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California
| |
Collapse
|