1
|
Yamada K, Kubota Y, Kosaka K, Yamaji Y, Akita S, Kuroda M, Mitsukawa N. Emergence of regulatory and matrix-producing stromal cells during adipogenic induction in human ceiling culture-derived preadipocytes. Biochem Biophys Res Commun 2025; 771:152012. [PMID: 40409116 DOI: 10.1016/j.bbrc.2025.152012] [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: 05/04/2025] [Accepted: 05/13/2025] [Indexed: 05/25/2025]
Abstract
Ceiling culture-derived preadipocytes (ccdPAs) are fibroblast-like cells believed to originate from mature adipocytes after the loss of lipid droplets. Unlike adipose-derived stem cells (ASCs), which are heterogeneous, ccdPAs are considered a more homogeneous population. However, their response to adipogenic differentiation stimuli at the single-cell level has not been fully characterized. In this study, we performed single-cell RNA sequencing (scRNA-seq) and epigenetic analyses to investigate early transcriptomic changes following adipogenic induction in ASCs and ccdPAs. scRNA-seq revealed that, in addition to preadipocytes/adipocytes, a substantial population of regulatory or structural cells, characterized by the expression of genes associated with extracellular matrix organization, structural support, and cell-cell interactions, such as F3 and MGP, emerged in both ASCs and ccdPAs. Notably, ATOH8, a transcription factor with limited prior characterization in adipogenesis, showed markedly higher expression in ccdPAs than in ASCs. This was supported by epigenetic analyses demonstrating lower CpG methylation and higher H3K4me3 levels at the ATOH8 locus in ccdPAs. Our findings suggest that adipogenic induction of ccdPAs generates diverse cell populations. This study provides the first single-cell level insight into the adipogenic response of primary cultured human ccdPAs.
Collapse
Affiliation(s)
- Kahoko Yamada
- Department of Plastic Surgery, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba-city, Chiba, 260-8670, Japan
| | - Yoshitaka Kubota
- Department of Plastic Surgery, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba-city, Chiba, 260-8670, Japan.
| | - Kentaro Kosaka
- Department of Plastic Surgery, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba-city, Chiba, 260-8670, Japan
| | - Yoshihisa Yamaji
- Department of Plastic Surgery, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba-city, Chiba, 260-8670, Japan
| | - Shinsuke Akita
- Department of Plastic Surgery, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba-city, Chiba, 260-8670, Japan
| | - Masayuki Kuroda
- Center for Advanced Medicine, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba-city, Chiba, 260-8670, Japan
| | - Nobuyuki Mitsukawa
- Department of Plastic Surgery, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba-city, Chiba, 260-8670, Japan
| |
Collapse
|
2
|
Wang X, Wang J, Zhao X, Zhang J, Zhang Y. The adipokines in oral cancer pathogenesis and its potential as a new therapeutic approach. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03939-w. [PMID: 40056203 DOI: 10.1007/s00210-025-03939-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 02/16/2025] [Indexed: 03/10/2025]
Abstract
The involvement of adipose tissue in the development of cancer is currently the subject of an increasing number of research due to the growing relevance of lipid metabolism in tumor growth. Obesity influences the tumor immune microenvironment (TME) in oral cancer. Visceral white adipose tissue (WAT) consists of adipocytes, connective tissue, immune cells, and stromovascular cells. The metabolic processes of immune cells within the adipose tissue of individuals with obesity predominantly depend on oxidative phosphorylation (intrinsically) and are characterized by elevated levels of M2 macrophages, Treg cells, Th2 cells, and eosinophils from an extrinsic perspective. The adipokines secreted by adipocytes facilitate communication with adjacent tissues to regulate glucose and lipid metabolism. Obesity influences cancer progression through the dysregulation of adipocytokines, characterized by an augmented synthesis of the oncogenic adipokine leptin, coupled with a reduced secretion of adiponectin. Under standard physiological settings, these adipokines fulfill essential roles in sustaining homeostasis. This review analyzed the influence of adipocytes on oral cancer by detailing the mediators released by adipocytes. Comprehending the molecular foundations of the protumor roles of adipokines in oral cancers might provide novel treatment targets.
Collapse
Affiliation(s)
- Xue Wang
- Department of Stomatology, Jilin Province FAW General Hospital, Jilin, 130000, China
| | - Jiapeng Wang
- Department of Orthopedics, Jilin Province FAW General Hospital, Jilin, 130000, China.
| | - Xuemei Zhao
- Department of Stomatology, Jilin Province FAW General Hospital, Jilin, 130000, China
| | - Jiayin Zhang
- Department of Stomatology, Jilin Province FAW General Hospital, Jilin, 130000, China
| | - Yan Zhang
- Medical Department, Changchun Sci-Tech University, Changchun, 130000, China
| |
Collapse
|
3
|
Wang YW, Luo CW. Unveiling the signal valve specifically tuning the TGF-β1 suppression of osteogenesis: mediation through a SMAD1-SMAD2 complex. Cell Commun Signal 2025; 23:38. [PMID: 39844165 PMCID: PMC11752969 DOI: 10.1186/s12964-025-02051-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 01/16/2025] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND TGF-β1 is the most abundant cytokine in bone, in which it serves as a vital factor to interdict adipogenesis and osteogenesis of bone marrow-derived mesenchymal stem cells (BM-MSCs). However, how TGF-β1 concurrently manipulates differentiation into these two distinct lineages remains elusive. METHODS Treatments with ligands or inhibitors followed by biochemical characterization, reporter assay, quantitative PCR and induced differentiation were applied to MSC line or primary BM-MSCs for signaling dissection. In vivo adipogenesis and ex vivo culture of bone explants were used to verify the functions of different SMAD complexes. Ingenuity Pathway Analysis, and analysis of transcriptomic datasets from human BM-MSCs in combination with hierarchical clustering and STRING assay were used to decipher the interplaying co-repressors. Mouse models of chronic and acute bone loss followed by biochemical assays and micro-computed tomography demonstrated the bone effects when functionally blocking the critical co-repressor HDAC1. RESULTS Distinct from the TGF-β1 inhibition on adipogenesis through canonical SMAD2/3 signaling, we clarified that TGF-β1 suppresses osteogenesis by inducing the formation of previously unidentified mixed SMADs mainly composed of SMAD1 and SMAD2, in which SMAD2 recruits more TGF-β1-induced co-repressors including HDAC1, TGIF1 and ATF3, whereas SMAD1 allows directing the whole transcriptional suppression complex to the cis-elements of osteogenic genes. Depletion of the cross-activation to the mixed SMADs dismantled specifically the TGF-β1 suppression on osteogenesis without affecting its inhibition on adipogenesis. Such phenomena can be reproduced via knockdown of co-repressors such as Hdac1 or addition of HDAC1 inhibitors in TGF-β1-treated MSCs. In either the chronic or the acute bone loss model, we demonstrated that the TGF-β signaling was augmented in the bone niche during osteolysis, whereas administration of HDAC1 inhibitors significantly improved bone quality. CONCLUSION This study identifies a new signal valve through which TGF-β1 can inhibit osteogenesis specifically. Functional interruption of this valve can tilt the seesaw balance of BM-MSC differentiation towards osteogenesis, highlighting the interplaying co-repressors, such as HDAC1, as promising therapeutic targets to combat diverse degenerative orthopedic diseases.
Collapse
Affiliation(s)
- Ying-Wen Wang
- Department of Life Sciences, Institute of Genome Sciences, National Yang Ming Chiao Tung University, 155 Li-Nong Street, Section 2, Beitou, Taipei, 112, Taiwan
| | - Ching-Wei Luo
- Department of Life Sciences, Institute of Genome Sciences, National Yang Ming Chiao Tung University, 155 Li-Nong Street, Section 2, Beitou, Taipei, 112, Taiwan.
| |
Collapse
|
4
|
Liu S, Ren J, Hu Y, Zhou F, Zhang L. TGFβ family signaling in human stem cell self-renewal and differentiation. CELL REGENERATION (LONDON, ENGLAND) 2024; 13:26. [PMID: 39604763 PMCID: PMC11602941 DOI: 10.1186/s13619-024-00207-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/16/2024] [Accepted: 11/01/2024] [Indexed: 11/29/2024]
Abstract
Human stem cells are undifferentiated cells with the capacity for self-renewal and differentiation into distinct cell lineages, playing important role in the development and maintenance of diverse tissues and organs. The microenvironment of stem cell provides crucial factors and components that exert significant influence over the determination of cell fate. Among these factors, cytokines from the transforming growth factor β (TGFβ) superfamily, including TGFβ, bone morphogenic protein (BMP), Activin and Nodal, have been identified as important regulators governing stem cell maintenance and differentiation. In this review, we present a comprehensive overview of the pivotal roles played by TGFβ superfamily signaling in governing human embryonic stem cells, somatic stem cells, induced pluripotent stem cells, and cancer stem cells. Furthermore, we summarize the latest research and advancements of TGFβ family in various cancer stem cells and stem cell-based therapy, discussing their potential clinical applications in cancer therapy and regeneration medicine.
Collapse
Affiliation(s)
- Sijia Liu
- International Biomed-X Research Center, Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Jiang Ren
- The First Affiliated Hospital, MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, Institute of Biomedical Innovation, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yanmei Hu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Fangfang Zhou
- The First Affiliated Hospital, the Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, China.
| | - Long Zhang
- International Biomed-X Research Center, Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China.
- The First Affiliated Hospital, MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, Institute of Biomedical Innovation, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China.
- MOE Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
5
|
Guo G, Wang W, Tu M, Zhao B, Han J, Li J, Pan Y, Zhou J, Ma W, Liu Y, Sun T, Han X, An Y. Deciphering adipose development: Function, differentiation and regulation. Dev Dyn 2024; 253:956-997. [PMID: 38516819 DOI: 10.1002/dvdy.708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 03/02/2024] [Accepted: 03/10/2024] [Indexed: 03/23/2024] Open
Abstract
The overdevelopment of adipose tissues, accompanied by excess lipid accumulation and energy storage, leads to adipose deposition and obesity. With the increasing incidence of obesity in recent years, obesity is becoming a major risk factor for human health, causing various relevant diseases (including hypertension, diabetes, osteoarthritis and cancers). Therefore, it is of significance to antagonize obesity to reduce the risk of obesity-related diseases. Excess lipid accumulation in adipose tissues is mediated by adipocyte hypertrophy (expansion of pre-existing adipocytes) or hyperplasia (increase of newly-formed adipocytes). It is necessary to prevent excessive accumulation of adipose tissues by controlling adipose development. Adipogenesis is exquisitely regulated by many factors in vivo and in vitro, including hormones, cytokines, gender and dietary components. The present review has concluded a comprehensive understanding of adipose development including its origin, classification, distribution, function, differentiation and molecular mechanisms underlying adipogenesis, which may provide potential therapeutic strategies for harnessing obesity without impairing adipose tissue function.
Collapse
Affiliation(s)
- Ge Guo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Wanli Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Mengjie Tu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Binbin Zhao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Jiayang Han
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Jiali Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Yanbing Pan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Jie Zhou
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Wen Ma
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Yi Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Tiantian Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Xu Han
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Yang An
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| |
Collapse
|
6
|
Umetsu A, Watanabe M, Sato T, Higashide M, Nishikiori N, Furuhashi M, Ohguro H. TGF-β effects on adipogenesis of 3T3-L1 cells differ in 2D and 3D cell culture conditions. FEBS Open Bio 2024. [PMID: 39380256 DOI: 10.1002/2211-5463.13890] [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: 02/06/2024] [Revised: 04/04/2024] [Accepted: 08/23/2024] [Indexed: 10/10/2024] Open
Abstract
The TGF-β superfamily plays a pivotal role in the regulation of adipogenesis, but little is known about the potential differential role of the three isoforms of TGF-β, TGF-β-1~3. To further elucidate their role, two-dimensionally (2D) and three-dimensionally (3D) cultured 3T3-L1 mouse preadipocytes were subjected to the following analyses: (a) qPCR analysis of adipogenesis-related factors and major extracellular matrix protein (2D and /or 3D), (b) lipid staining by Oil Red O (2D) or BODIPY (3D), (c) Seahorse cellular metabolic measurement (2D), and (d) size and stiffness measurements of 3D 3T3-L1 spheroids. In the 2D cultured 3T3-L1 cells, mRNA expression levels of adipogenesis-related genes and Oil Red O lipid staining intensity were significantly increased by adipogenesis and they were substantially decreased following treatment with 0.1 nm TGF-β isoforms, with TGF-β2 having the greater effects. Consistent with these results, treatment with TGF-β2 resulted in suppression of mitochondrial and glycolytic functions in 2D cultured 3T3-L1 cells. However, the inhibitory effect of TGF-β on adipogenesis decreased under 3D spheroid culture conditions and TGF-β isoforms did not affect adipogenesis-induced (a) enlargement and downsizing of 3T3-L1 spheroids, (b) increase in BODIPY lipid staining intensity, and (c) up-regulation of the mRNA expression of adipogenesis-related genes. The findings presented herein suggest that the three TGF-β isoforms have different suppressive effects on adipogenesis-related cellular properties of 2D cultured 3T3-L1 cells and that their effects decrease under 3D spheroid culture conditions.
Collapse
Affiliation(s)
- Araya Umetsu
- Department of Ophthalmology, Sapporo Medical University School of Medicine, Japan
| | - Megumi Watanabe
- Department of Ophthalmology, Sapporo Medical University School of Medicine, Japan
| | - Tatsuya Sato
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Japan
- Department of Cellular Physiology and Signal Transduction, Sapporo Medical University School of Medicine, Japan
| | - Megumi Higashide
- Department of Ophthalmology, Sapporo Medical University School of Medicine, Japan
| | - Nami Nishikiori
- Department of Ophthalmology, Sapporo Medical University School of Medicine, Japan
| | - Masato Furuhashi
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University School of Medicine, Japan
| | - Hiroshi Ohguro
- Department of Ophthalmology, Sapporo Medical University School of Medicine, Japan
| |
Collapse
|
7
|
Ford HR, Mitchell TM, Scull T, Benitez OJ, Strieder-Barboza C. The Effect of Subclinical Ketosis on the Peripheral Blood Mononuclear Cell Inflammatory Response and Its Crosstalk with Depot-Specific Preadipocyte Function in Dairy Cows. Animals (Basel) 2024; 14:1995. [PMID: 38998107 PMCID: PMC11240650 DOI: 10.3390/ani14131995] [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: 05/03/2024] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 07/14/2024] Open
Abstract
During the periparturient period, cows undergo heightened energy demands at lactation onset, paired with reduced dry matter intake, leading to negative energy balance (NEB). Excessive lipolysis-driven adipose tissue remodeling, triggered by NEB, significantly contributes to ketosis in periparturient dairy cows. However, the role of peripheral blood mononuclear cells (PBMCs) in the pathogenesis of ketosis and in modulating adipose tissue function remains poorly understood. Here, we investigated how ketosis affects the transcriptional profile and secretome of PBMCs and its influence on preadipocyte function in visceral adipose tissue (VAT) and subcutaneous adipose tissue (SAT). Twenty-one postpartum Holstein dairy cows were categorized as either subclinical ketosis (SCK; BHB ≥ 1.0 mM) or control (CON; BHB < 0.8 mM) based on blood beta-hydroxybutyrate (BHB) concentration screening. Blood samples were collected intravenously for the isolation of PBMCs and serum metabolic profiling. Ketosis elevated circulating NEFA and BHB levels but reduced total WBC and neutrophil counts. Isolated PBMCs were evaluated for gene expression and used to produce conditioned media (PBMC-CM), during which PBMCs were stimulated with 10 ng/mL LPS. The overall phenotype of PBMCs was largely consistent between SCK and CON cows, with minimal differences detected in immunomodulatory cytokine expression and PBMC-CM composition following stimulation. Preadipocytes isolated from non-ketotic cows were treated with PBMC-CM to assess the effect of PBMC secretomes on adipose cell function. Preadipocytes treated with SCK PBMC-CM showed reduced lipid accumulation compared to those treated with CON PBMC-CM regardless of the depot. SAT preadipocytes had heightened expression of lipid metabolism-related genes, including DGAT1, LIPE, and FASN, compared to VAT when treated with SCK PBMC-CM. Preadipocytes treated with CM from PBMC stimulated by LPS exhibited upregulation in IL1B and IL6 regardless of the depot or source of PBMCs. Together, these results indicate that although PBMC profiles showed minimal differences, preadipocytes treated with PBMC-CM may be influenced by additional factors, leading to altered preadipocyte function and gene expression that may contribute to adipose cellular dysfunction.
Collapse
Affiliation(s)
- Hunter R. Ford
- Department of Veterinary Sciences, Davis College of Agricultural Sciences and Natural Resources, Texas Tech University, Lubbock, TX 79409, USA; (H.R.F.); (T.M.M.); (T.S.); (O.J.B.)
| | - Ty M. Mitchell
- Department of Veterinary Sciences, Davis College of Agricultural Sciences and Natural Resources, Texas Tech University, Lubbock, TX 79409, USA; (H.R.F.); (T.M.M.); (T.S.); (O.J.B.)
| | - Tanner Scull
- Department of Veterinary Sciences, Davis College of Agricultural Sciences and Natural Resources, Texas Tech University, Lubbock, TX 79409, USA; (H.R.F.); (T.M.M.); (T.S.); (O.J.B.)
| | - Oscar J. Benitez
- Department of Veterinary Sciences, Davis College of Agricultural Sciences and Natural Resources, Texas Tech University, Lubbock, TX 79409, USA; (H.R.F.); (T.M.M.); (T.S.); (O.J.B.)
- School of Veterinary Medicine, Texas Tech University, Amarillo, TX 79106, USA
| | - Clarissa Strieder-Barboza
- Department of Veterinary Sciences, Davis College of Agricultural Sciences and Natural Resources, Texas Tech University, Lubbock, TX 79409, USA; (H.R.F.); (T.M.M.); (T.S.); (O.J.B.)
- School of Veterinary Medicine, Texas Tech University, Amarillo, TX 79106, USA
| |
Collapse
|
8
|
Huang M, Wang X, Chen Y, Pessoa MT, Terrell KC, Zhang J, Tian J, Xie Z, Pierre SV, Cai L. Role of Na/K-ATPase α1 caveolin-binding motif in adipogenesis. Am J Physiol Cell Physiol 2024; 327:C48-C64. [PMID: 38708522 PMCID: PMC11371328 DOI: 10.1152/ajpcell.00168.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/28/2024] [Accepted: 04/29/2024] [Indexed: 05/07/2024]
Abstract
Deficiencies in mice and in humans have brought to the fore the importance of the caveolar network in key aspects of adipocyte biology. The conserved N-terminal caveolin-binding motif (CBM) of the ubiquitous Na/K-ATPase (NKA) α1 isoform, which allows NKA/caveolin-1 (Cav1) interaction, influences NKA signaling and caveolar distribution. It has been shown to be critical for animal development and ontogenesis, as well as lineage-specific differentiation of human induced pluripotent stem cells (hiPSCs). However, its role in postnatal adipogenesis has not been fully examined. Using a genetic approach to alter CBM in hiPSC-derived adipocytes (iAdi-mCBM) and in mice (mCBM), we investigated the regulatory function of NKA CBM signaling in adipogenesis. Seahorse XF cell metabolism analyses revealed impaired glycolysis and decreased ATP synthesis-coupled respiration in iAdi-mCBM. These metabolic dysfunctions were accompanied by evidence of extensive remodeling of the extracellular matrix (ECM), including increased collagen staining, overexpression of ECM marker genes, and heightened TGF-β signaling uncovered by RNAseq analysis. Rescue of mCBM by lentiviral delivery of WT NKA α1 or treatment of mCBM hiPSCs with the TGF-β inhibitor SB431542 normalized ECM, suggesting that NKA CBM signaling integrity is required for adequate control of TGF-β signaling and ECM stiffness during adipogenesis. The physiological impact was revealed in mCBM male mice with reduced fat mass accompanied by histological and transcriptional evidence of elevated adipose fibrosis and decreased adipocyte size. Based on these findings, we propose that the genetic alteration of the NKA/Cav1 regulatory path uncovered in human iAdi leads to lipodystrophy in mice.NEW & NOTEWORTHY A Na/K-ATPase α1 caveolin-binding motif regulates adipogenesis. Mutation of this binding motif in the mouse leads to reduced fat with increased extracellular matrix production and inflammation. RNA-seq analysis and pharmacological interventions in human iPSC-derived adipocytes revealed that TGF-β signal, rather than Na/K-ATPase-mediated ion transport, is a key mediator of NKA regulation of adipogenesis.
Collapse
Affiliation(s)
- Minqi Huang
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia, United States
| | - Xiaoliang Wang
- Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia, United States
| | - Yiliang Chen
- Versiti Blood Research Institute, Milwaukee, West Virginia, United States
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Marco T Pessoa
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia, United States
| | - Kayleigh C Terrell
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia, United States
| | - Jue Zhang
- Versiti Blood Research Institute, Milwaukee, West Virginia, United States
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Jiang Tian
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia, United States
- Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia, United States
| | - Zijian Xie
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia, United States
| | - Sandrine V Pierre
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia, United States
| | - Liquan Cai
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia, United States
| |
Collapse
|
9
|
Chang YH, Tseng YH, Wang JM, Tsai YS, Huang HS. TG-interacting factor 1 regulates mitotic clonal expansion during adipocyte differentiation. Biochim Biophys Acta Mol Cell Biol Lipids 2024; 1869:159492. [PMID: 38575107 DOI: 10.1016/j.bbalip.2024.159492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 03/01/2024] [Accepted: 03/28/2024] [Indexed: 04/06/2024]
Abstract
Obesity is one of the significant health challenges in the world and is highly associated with abnormal adipogenesis. TG-interacting factor 1 (TGIF1) is essential for differentiating murine adipocytes and human adipose tissue-derived stem cells. However, the mode of action needs to be better elucidated. To investigate the roles of TGIF1 in differentiation in-depth, CRISPR/Cas9 knockout technology was performed to generate TGIF1-silenced preadipocytes. The absence of TGIF1 in 3 T3-F442A preadipocytes abolished lipid accumulation throughout the differentiation using Oil Red O staining. Conversely, we established 3 T3-F442A preadipocytes stably expressing TGIF1 and doxycycline-inducible TGIF1 in TGIF1-silenced 3 T3-F442A preadipocytes. Remarkably, the induction of TGIF1 by doxycycline during the initial differentiation phase successfully promoted lipid accumulation in TGIF1-silenced 3 T3-F442A cells. We further explored the mechanisms of TGIF1 in early differentiation. We demonstrated that TGIF1 promoted the mitotic clonal expansion via upregulation of CCAAT/enhancer-binding proteins β expression, interruption with peroxisome proliferators activated receptor γ downstream regulation, and inhibition of p27kip1 expression. In conclusion, we strengthen the pivotal roles of TGIF1 in early differentiation, which might contribute to resolving obesity-associated metabolic syndromes.
Collapse
Affiliation(s)
- Yu-Hao Chang
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Hua Tseng
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA.
| | - Ju-Ming Wang
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan.
| | - Yau-Sheng Tsai
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Huei-Sheng Huang
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
10
|
Yu B, Cai Z, Liu J, Zhang T, Feng X, Wang C, Li J, Gu Y, Zhang J. Identification of key differentially methylated genes in regulating muscle development and intramuscular fat deposition in chickens. Int J Biol Macromol 2024; 264:130737. [PMID: 38460642 DOI: 10.1016/j.ijbiomac.2024.130737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 02/26/2024] [Accepted: 03/06/2024] [Indexed: 03/11/2024]
Abstract
Muscle development and intramuscular fat (IMF) deposition are intricate physiological processes characterized by multiple gene expressions and interactions. In this research, the phenotypic variations in the breast muscle of Jingyuan chickens were examined at three different time points: 42, 126, and 180 days old. Differential expression analysis and weighted gene co-expression network analysis (WGCNA) were performed to identify differentially methylated genes (DMGs) responsible for regulating muscle development and IMF deposition. The findings indicate a significant increase in breast muscle weight (BMW), myofiber diameter, and cross-sectional area, as well as IMF content, in correlation with the progressive number of growing days in Jingyuan chickens. The findings also revealed that 380 hypo-methylated and 253 hyper-methylated DMGs were identified between the three groups of breast muscle. Module gene and DMG association analysis identified m6A methylation-mediated multiple DMGs associated with muscle development and fat metabolism. In vitro cell modeling analysis reveals stage-specific differences in the expression of CUBN, MEGF10, BOP1, and BMPR2 during the differentiation of myoblasts and intramuscular preadipocytes. Cycloleucine treatment significantly inhibited the expression levels of CUBN, BOP1, and BMPR2, and promoted the expression of MEGF10. These results suggest that m6A methylation-mediated CUBN, MEGF10, BOP1, and BMPR2 can serve as potential candidate genes for regulating muscle development and IMF deposition, and provide an important theoretical basis for further investigation of the functional mechanism of m6A modification involved in adipogenesis.
Collapse
Affiliation(s)
- Baojun Yu
- College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China
| | - Zhengyun Cai
- College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China
| | - Jiamin Liu
- College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China
| | - Tong Zhang
- College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China
| | - Xiaofang Feng
- College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China
| | - Chuanchuan Wang
- College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China
| | - Jiwei Li
- College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China
| | - Yaling Gu
- College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China
| | - Juan Zhang
- College of Animal Science and Technology, Ningxia University, Yinchuan 750021, China.
| |
Collapse
|
11
|
Lim NK, Jeon HB, Kim S. The transdifferentiation of human dedifferentiated fat cells into fibroblasts: An in vitro experimental pilot study. Medicine (Baltimore) 2024; 103:e37595. [PMID: 38552064 PMCID: PMC10977558 DOI: 10.1097/md.0000000000037595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 02/22/2024] [Indexed: 04/02/2024] Open
Abstract
BACKGROUND Skin grafting is a common method of treating damaged skin; however, surgical complications may arise in patients with poor health. Currently, no effective conservative treatment is available for extensive skin loss. Mature adipocytes, which constitute a substantial portion of adipose tissue, have recently emerged as a potential source of stemness. When de-lipidated, these cells exhibit fibroblast-like characteristics and the ability to redifferentiate, offering homogeneity and research utility as "dedifferentiated fat cells." METHODS AND RESULTS We conducted an in vitro study to induce fibroblast-like traits in the adipose tissue by transdifferentiating mature adipocytes for skin regeneration. Human subcutaneous fat tissues were isolated and purified from mature adipocytes that underwent a transformation process over 14 days of cultivation. Microscopic analysis revealed lipid degradation over time, ultimately transforming cells into fibroblast-like forms. Flow cytometry was used to verify their characteristics, highlighting markers such as CD90 and CD105 (mesenchymal stem cell markers) and CD56 and CD106 (for detecting fibroblast characteristics). Administering dedifferentiated fat cells with transforming growth factor-β at the identified optimal differentiation concentration of 5 ng/mL for a span of 14 days led to heightened expression of alpha smooth muscle actin and fibronectin, as evidenced by RNA and protein analysis. Meanwhile, functional validation through cell sorting demonstrated limited fibroblast marker expression in both treated and untreated cells after transdifferentiation by transforming growth factor-β. CONCLUSION Although challenges remain in achieving more effective transformation and definitive fibroblast differentiation, our trial could pave the way for a novel skin regeneration treatment strategy.
Collapse
Affiliation(s)
- Nam Kyu Lim
- Department of Plastic and Reconstructive Surgery, Dankook University College of Medicine, Cheonan, Chungcheongnamdo, Republic of Korea
- Dankook Physician Scientist Research Center (DPSRC), Dankook University Hospital, Cheonan, Chungcheongnamdo, Republic of Korea
| | - Hong Bae Jeon
- Department of Plastic and Reconstructive Surgery, Dankook University College of Medicine, Cheonan, Chungcheongnamdo, Republic of Korea
- Dankook Physician Scientist Research Center (DPSRC), Dankook University Hospital, Cheonan, Chungcheongnamdo, Republic of Korea
| | - Sungyeon Kim
- Department of Plastic and Reconstructive Surgery, Dankook University College of Medicine, Cheonan, Chungcheongnamdo, Republic of Korea
| |
Collapse
|
12
|
Deng Z, Fan T, Xiao C, Tian H, Zheng Y, Li C, He J. TGF-β signaling in health, disease, and therapeutics. Signal Transduct Target Ther 2024; 9:61. [PMID: 38514615 PMCID: PMC10958066 DOI: 10.1038/s41392-024-01764-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 08/31/2023] [Accepted: 01/31/2024] [Indexed: 03/23/2024] Open
Abstract
Transforming growth factor (TGF)-β is a multifunctional cytokine expressed by almost every tissue and cell type. The signal transduction of TGF-β can stimulate diverse cellular responses and is particularly critical to embryonic development, wound healing, tissue homeostasis, and immune homeostasis in health. The dysfunction of TGF-β can play key roles in many diseases, and numerous targeted therapies have been developed to rectify its pathogenic activity. In the past decades, a large number of studies on TGF-β signaling have been carried out, covering a broad spectrum of topics in health, disease, and therapeutics. Thus, a comprehensive overview of TGF-β signaling is required for a general picture of the studies in this field. In this review, we retrace the research history of TGF-β and introduce the molecular mechanisms regarding its biosynthesis, activation, and signal transduction. We also provide deep insights into the functions of TGF-β signaling in physiological conditions as well as in pathological processes. TGF-β-targeting therapies which have brought fresh hope to the treatment of relevant diseases are highlighted. Through the summary of previous knowledge and recent updates, this review aims to provide a systematic understanding of TGF-β signaling and to attract more attention and interest to this research area.
Collapse
Affiliation(s)
- Ziqin Deng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Tao Fan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chu Xiao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - He Tian
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yujia Zheng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chunxiang Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
13
|
Eisinger K, Girke P, Buechler C, Krautbauer S. Adipose tissue depot specific expression and regulation of fibrosis-related genes and proteins in experimental obesity. Mamm Genome 2024; 35:13-30. [PMID: 37884762 PMCID: PMC10884164 DOI: 10.1007/s00335-023-10022-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 10/02/2023] [Indexed: 10/28/2023]
Abstract
Transforming growth factor beta (Tgfb) is a well-studied pro-fibrotic cytokine, which upregulates cellular communication network factor 2 (Ccn2), collagen, and actin alpha 2, smooth muscle (Acta2) expression. Obesity induces adipose tissue fibrosis, which contributes to metabolic diseases. This work aimed to analyze the expression of Tgfb, Ccn2, collagen1a1 (Col1a1), Acta2 and BMP and activin membrane-bound inhibitor (Bambi), which is a negative regulator of Tgfb signaling, in different adipose tissue depots of mice fed a standard chow, mice fed a high fat diet (HFD) and ob/ob mice. Principally, these genes were low expressed in brown adipose tissues and this difference was less evident for the ob/ob mice. Ccn2 and Bambi protein as well as mRNA expression, and collagen1a1 mRNA were not induced in the adipose tissues upon HFD feeding whereas Tgfb and Acta2 mRNA increased in the white fat depots. Immunoblot analysis showed that Acta2 protein was higher in subcutaneous and perirenal fat of these mice. In the ob/ob mice, Ccn2 mRNA and Ccn2 protein were upregulated in the fat depots. Here, Tgfb, Acta2 and Col1a1 mRNA levels and serum Tgfb protein were increased. Acta2 protein was, however, not higher in subcutaneous and perirenal fat of these mice. Col6a1 mRNA was shown before to be higher in obese fat tissues. Current analysis proved the Col6a1 protein was induced in subcutaneous fat of HFD fed mice. Notably, Col6a1 was reduced in perirenal fat of ob/ob mice in comparison to the respective controls. 3T3-L1 cells express Ccn2 and Bambi protein, whose levels were not changed by fatty acids, leptin, lipopolysaccharide, tumor necrosis factor and interleukin-6. All of these factors led to higher Tgfb in 3T3-L1 adipocyte media but did not increase its mRNA levels. Free fatty acids induced necrosis whereas apoptosis did not occur in any of the in vitro incubations excluding cell death as a main reason for higher Tgfb in cell media. In summary, Tgfb mRNA is consistently induced in white fat tissues in obesity but this is not paralleled by a clear increase of its target genes. Moreover, discrepancies between mRNA and protein expression of Acta2 were observed. Adipocytes seemingly do not contribute to higher Tgfb mRNA levels in obesity. These cells release more Tgfb protein when challenged with obesity-related metabolites connecting metabolic dysfunction and fibrosis.
Collapse
Affiliation(s)
- Kristina Eisinger
- Department of Internal Medicine I, Regensburg University Hospital, 93053, Regensburg, Germany
| | - Philipp Girke
- Department of Genetics, University of Regensburg, 93040, Regensburg, Germany
| | - Christa Buechler
- Department of Internal Medicine I, Regensburg University Hospital, 93053, Regensburg, Germany.
| | - Sabrina Krautbauer
- Department of Internal Medicine I, Regensburg University Hospital, 93053, Regensburg, Germany
| |
Collapse
|
14
|
Engin AB, Engin A. MicroRNAs as Epigenetic Regulators of Obesity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:595-627. [PMID: 39287866 DOI: 10.1007/978-3-031-63657-8_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
In obesity, the process of adipogenesis largely determines the number of adipocytes in body fat depots. Adipogenesis is regulated by several adipocyte-selective micro-ribonucleic acids (miRNAs) and transcription factors that modulate adipocyte proliferation and differentiation. However, some miRNAs block the expression of master regulators of adipogenesis. Since the specific miRNAs display different expressions during adipogenesis, in mature adipocytes and permanent obesity, their use as biomarkers or therapeutic targets is feasible. Upregulated miRNAs in persistent obesity are downregulated during adipogenesis. Moreover, some of the downregulated miRNAs in obese individuals are upregulated in mature adipocytes. Induction of adipocyte stress and hypertrophy leads to the release of adipocyte-derived exosomes (AdEXs) that contain the cargo molecules, miRNAs. miRNAs are important messengers for intercellular communication involved in metabolic responses and have very specific signatures that direct the metabolic activity of target cells. While each miRNA targets multiple messenger RNAs (mRNAs), which may coordinate or antagonize each other's functions, several miRNAs are dysregulated in other tissues during obesity-related comorbidities. Deletion of the miRNA-processing enzyme DICER in pro-opiomelanocortin-expressing cells results in obesity, which is characterized by hyperphagia, increased adiposity, hyperleptinemia, defective glucose metabolism, and alterations in the pituitary-adrenal axis. In recent years, RNA-based therapeutical approaches have entered clinical trials as novel therapies against overweight and its complications. Development of lipid droplets, macrophage accumulation, macrophage polarization, tumor necrosis factor receptor-associated factor 6 activity, lipolysis, lipotoxicity, and insulin resistance are effectively controlled by miRNAs. Thereby, miRNAs as epigenetic regulators are used to determine the new gene transcripts and therapeutic targets.
Collapse
Affiliation(s)
- Ayse Basak Engin
- Faculty of Pharmacy, Department of Toxicology, Gazi University, Hipodrom, Ankara, Turkey.
| | - Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey
| |
Collapse
|
15
|
Liu G, Wang Y, Pan Y, Tian L, Choi MH, Wang L, Kim JY, Zhang J, Cheng SH, Zhang L. Hypertonicity induces mitochondrial extracellular vesicles (MEVs) that activate TNF-α and β-catenin signaling to promote adipocyte dedifferentiation. Stem Cell Res Ther 2023; 14:333. [PMID: 38115136 PMCID: PMC10731851 DOI: 10.1186/s13287-023-03558-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 11/02/2023] [Indexed: 12/21/2023] Open
Abstract
BACKGROUND Recent studies demonstrated that elevated osmolarity could induce adipocyte dedifferentiation, representing an appealing procedure to generate multipotent stem cells. Here we aim to elucidate the molecular mechanisms that underlie osmotic induction of adipocyte reprogramming. METHODS To induce dedifferentiation, the 3T3-L1 or SVF adipocytes were cultured under the hypertonic pressure in 2% PEG 300 medium. Adipocyte dedifferentiation was monitored by aspect ratio measurement, Oil Red staining and qPCR to examine the morphology, lipid droplets, and specific genes of adipocytes, respectively. The osteogenic and chondrogenic re-differentiation capacities of dedifferentiated adipocytes were also examined. To investigate the mechanisms of the osmotic stress-induced dedifferentiation, extracellular vesicles (EVs) were collected from the reprograming cells, followed by proteomic and functional analyses. In addition, qPCR, ELISA, and TNF-α neutralizing antibody (20 ng/ml) was applied to examine the activation and effects of the TNF-α signaling. Furthermore, we also analyzed the Wnt signaling by assessing the activation of β-catenin and applying BML-284, an agonist of β-catenin. RESULTS Hypertonic treatment induced dedifferentiation of both 3T3-L1 and the primary stromal vascular fraction (SVF) adipocytes, characterized by morphological and functional changes. Proteomic profiling revealed that hypertonicity induced extracellular vesicles (EVs) containing mitochondrial molecules including NDUFA9 and VDAC. Functionally, the mitochondrial EVs (MEVs) stimulated TNF-α signaling that activates Wnt-β-catenin signaling and adipocyte dedifferentiation. Neutralizing TNF-α inhibited hypertonic dedifferentiation of adipocytes. In addition, direct activation of Wnt-β-catenin signaling using BML-284 could efficiently induce adipocyte dedifferentiation while circumventing the apoptotic effect of the hypertonic treatment. CONCLUSIONS Hypertonicity prompts the adipocytes to release MEVs, which in turn enhances the secretion of TNF-α as a pro-inflammatory cytokine during the stress response. Importantly, TNF-α is essential for the activation of the Wnt/β-catenin signaling that drives adipocyte dedifferentiation. A caveat of the hypertonic treatment is apoptosis, which could be circumvented by direct activation of the Wnt/β-catenin signaling using BML-284.
Collapse
Affiliation(s)
- Guopan Liu
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, China
- Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, 518057, China
| | - Ying Wang
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, China
- Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, 518057, China
| | - Yilin Pan
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, China
- Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, 518057, China
| | - Li Tian
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, China
- Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, 518057, China
| | - Ming Ho Choi
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, China
| | - Li Wang
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, China
| | - Jin Young Kim
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, China
| | - Jian Zhang
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200025, China
| | - Shuk Han Cheng
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, China
| | - Liang Zhang
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, China.
- Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, 518057, China.
| |
Collapse
|
16
|
Amiri MA, Farshidfar N, Miron RJ, Dziedzic A, Hamedani S, Daneshi S, Tayebi L. The Potential Therapeutic Effects of Platelet-Derived Biomaterials on Osteoporosis: A Comprehensive Review of Current Evidence. Int J Biomater 2023; 2023:9980349. [PMID: 38098766 PMCID: PMC10721351 DOI: 10.1155/2023/9980349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 11/09/2023] [Accepted: 11/24/2023] [Indexed: 12/17/2023] Open
Abstract
Osteoporosis is a chronic multifactorial condition that affects the skeletal system, leading to the deterioration of bone microstructure and an increased risk of bone fracture. Platelet-derived biomaterials (PDBs), so-called platelet concentrates, such as platelet-rich plasma (PRP) and platelet-rich fibrin (PRF), have shown potential for improving bone healing by addressing microstructural impairment. While the administration of platelet concentrates has yielded positive results in bone regeneration, the optimal method for its administration in the clinical setting is still debatable. This comprehensive review aims to explore the systemic and local use of PRP/PRF for treating various bone defects and acute fractures in patients with osteoporosis. Furthermore, combining PRP/PRF with stem cells or osteoinductive and osteoconductive biomaterials has shown promise in restoring bone microstructural properties, treating bony defects, and improving implant osseointegration in osteoporotic animal models. Here, reviewing the results of in vitro and in vivo studies, this comprehensive evaluation provides a detailed mechanism for how platelet concentrates may support the healing process of osteoporotic bone fractures.
Collapse
Affiliation(s)
- Mohammad Amin Amiri
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nima Farshidfar
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Richard J. Miron
- Department of Periodontology, University of Bern, Bern, Switzerland
| | - Arkadiusz Dziedzic
- Department of Conservative Dentistry with Endodontics, Medical University of Silesia, Katowice, Poland
| | - Shahram Hamedani
- Oral and Dental Disease Research Center, School of Dentistry, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sajad Daneshi
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Lobat Tayebi
- Marquette University School of Dentistry, Milwaukee, WI 53233, USA
| |
Collapse
|
17
|
Huna A, Flaman JM, Lodillinsky C, Zhu K, Makulyte G, Pakulska V, Coute Y, Ruisseaux C, Saintigny P, Hernandez-Vargas H, Defossez PA, Boissan M, Martin N, Bernard D. RSK3 switches cell fate: from stress-induced senescence to malignant progression. J Exp Clin Cancer Res 2023; 42:318. [PMID: 38008756 PMCID: PMC10680185 DOI: 10.1186/s13046-023-02909-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 11/19/2023] [Indexed: 11/28/2023] Open
Abstract
BACKGROUND TGFβ induces several cell phenotypes including senescence, a stable cell cycle arrest accompanied by a secretory program, and epithelial-mesenchymal transition (EMT) in normal epithelial cells. During carcinogenesis cells lose the ability to undergo senescence in response to TGFβ but they maintain an EMT, which can contribute to tumor progression. Our aim was to identify mechanisms promoting TGFβ-induced senescence escape. METHODS In vitro experiments were performed with primary human mammary epithelial cells (HMEC) immortalized by hTert. For kinase library screen and modulation of gene expression retroviral transduction was used. To characterize gene expression, RNA microarray with GSEA analysis and RT-qPCR were used. For protein level and localization, Western blot and immunofluorescence were performed. For senescence characterization crystal violet assay, Senescence Associated-β-Galactosidase activity, EdU staining were conducted. To determine RSK3 partners FLAG-baited immunoprecipitation and mass spectrometry-based proteomic analyses were performed. Proteosome activity and proteasome enrichment assays were performed. To validate the role of RSK3 in human breast cancer, analysis of METABRIC database was performed. Murine intraductal xenografts using MCF10DCIS.com cells were carried out, with histological and immunofluorescence analysis of mouse tissue sections. RESULTS A screen with active kinases in HMECs upon TGFβ treatment identified that the serine threonine kinase RSK3, or RPS6KA2, a kinase mainly known to regulate cancer cell death including in breast cancer, reverted TGFβ-induced senescence. Interestingly, RSK3 expression decreased in response to TGFβ in a SMAD3-dependent manner, and its constitutive expression rescued SMAD3-induced senescence, indicating that a decrease in RSK3 itself contributes to TGFβ-induced senescence. Using transcriptomic analyses and affinity purification coupled to mass spectrometry-based proteomics, we unveiled that RSK3 regulates senescence by inhibiting the NF-κΒ pathway through the decrease in proteasome-mediated IκBα degradation. Strikingly, senescent TGFβ-treated HMECs display features of epithelial to mesenchymal transition (EMT) and during RSK3-induced senescence escaped HMECs conserve EMT features. Importantly, RSK3 expression is correlated with EMT and invasion, and inversely correlated with senescence and NF-κΒ in human claudin-low breast tumors and its expression enhances the formation of breast invasive tumors in the mouse mammary gland. CONCLUSIONS We conclude that RSK3 switches cell fate from senescence to malignancy in response to TGFβ signaling.
Collapse
Affiliation(s)
- Anda Huna
- Cancer Research Center of Lyon, Inserm U1052, CNRS UMR 5286, Université de Lyon, Centre Léon Bérard, 69373, Lyon, France
- Equipe Labellisée La Ligue Contre Le Cancer, Lyon, France
| | - Jean-Michel Flaman
- Cancer Research Center of Lyon, Inserm U1052, CNRS UMR 5286, Université de Lyon, Centre Léon Bérard, 69373, Lyon, France
- Equipe Labellisée La Ligue Contre Le Cancer, Lyon, France
| | - Catalina Lodillinsky
- INSERM UMR_S 938, Saint-Antoine Research Center, CRSA, University Sorbonne, Paris, France
- Research Area, Instituto de Oncología Ángel H. Roffo, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Kexin Zhu
- Cancer Research Center of Lyon, Inserm U1052, CNRS UMR 5286, Université de Lyon, Centre Léon Bérard, 69373, Lyon, France
- Equipe Labellisée La Ligue Contre Le Cancer, Lyon, France
| | - Gabriela Makulyte
- Cancer Research Center of Lyon, Inserm U1052, CNRS UMR 5286, Université de Lyon, Centre Léon Bérard, 69373, Lyon, France
- Equipe Labellisée La Ligue Contre Le Cancer, Lyon, France
| | - Victoria Pakulska
- Université Grenoble Alpes, Inserm, UA13 BGE, CNRS, CEA, FR2048, 38000, Grenoble, France
| | - Yohann Coute
- Université Grenoble Alpes, Inserm, UA13 BGE, CNRS, CEA, FR2048, 38000, Grenoble, France
| | - Clémence Ruisseaux
- Cancer Research Center of Lyon, Inserm U1052, CNRS UMR 5286, Université de Lyon, Centre Léon Bérard, 69373, Lyon, France
| | - Pierre Saintigny
- Cancer Research Center of Lyon, Inserm U1052, CNRS UMR 5286, Université de Lyon, Centre Léon Bérard, 69373, Lyon, France
| | - Hector Hernandez-Vargas
- Cancer Research Center of Lyon, Inserm U1052, CNRS UMR 5286, Université de Lyon, Centre Léon Bérard, 69373, Lyon, France
| | | | - Mathieu Boissan
- INSERM UMR_S 938, Saint-Antoine Research Center, CRSA, University Sorbonne, Paris, France
| | - Nadine Martin
- Cancer Research Center of Lyon, Inserm U1052, CNRS UMR 5286, Université de Lyon, Centre Léon Bérard, 69373, Lyon, France
- Equipe Labellisée La Ligue Contre Le Cancer, Lyon, France
| | - David Bernard
- Cancer Research Center of Lyon, Inserm U1052, CNRS UMR 5286, Université de Lyon, Centre Léon Bérard, 69373, Lyon, France.
- Equipe Labellisée La Ligue Contre Le Cancer, Lyon, France.
| |
Collapse
|
18
|
Gupta A, Efthymiou V, Kodani SD, Shamsi F, Patti ME, Tseng YH, Streets A. Mapping the transcriptional landscape of human white and brown adipogenesis using single-nuclei RNA-seq. Mol Metab 2023; 74:101746. [PMID: 37286033 PMCID: PMC10338377 DOI: 10.1016/j.molmet.2023.101746] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 05/24/2023] [Accepted: 05/30/2023] [Indexed: 06/09/2023] Open
Abstract
Adipogenesis is key to maintaining organism-wide energy balance and healthy metabolic phenotype, making it critical to thoroughly comprehend its molecular regulation in humans. By single-nuclei RNA-sequencing (snRNA-seq) of over 20,000 differentiating white and brown preadipocytes, we constructed a high-resolution temporal transcriptional landscape of human white and brown adipogenesis. White and brown preadipocytes were isolated from a single individual's neck region, thereby eliminating inter-subject variability across two distinct lineages. These preadipocytes were also immortalized to allow for controlled, in vitro differentiation, allowing sampling of distinct cellular states across the spectrum of adipogenic progression. Pseudotemporal cellular ordering revealed the dynamics of ECM remodeling during early adipogenesis, and lipogenic/thermogenic response during late white/brown adipogenesis. Comparison with adipogenic regulation in murine models Identified several novel transcription factors as potential targets for adipogenic/thermogenic drivers in humans. Among these novel candidates, we explored the role of TRPS1 in adipocyte differentiation and showed that its knockdown impairs white adipogenesis in vitro. Key adipogenic and lipogenic markers revealed in our analysis were applied to analyze publicly available scRNA-seq datasets; these confirmed unique cell maturation features in recently discovered murine preadipocytes, and revealed inhibition of adipogenic expansion in humans with obesity. Overall, our study presents a comprehensive molecular description of both white and brown adipogenesis in humans and provides an important resource for future studies of adipose tissue development and function in both health and metabolic disease state.
Collapse
Affiliation(s)
- Anushka Gupta
- University of California at Berkeley, University of California at San Francisco Graduate Program in Bioengineering, Berkeley, CA 94720, USA
| | - Vissarion Efthymiou
- Department of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02115, USA
| | - Sean D Kodani
- Department of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02115, USA
| | - Farnaz Shamsi
- Department of Molecular Pathobiology, New York University, New York, NY 10010, USA
| | - Mary Elizabeth Patti
- Department of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02115, USA
| | - Yu-Hua Tseng
- Department of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Aaron Streets
- University of California at Berkeley, University of California at San Francisco Graduate Program in Bioengineering, Berkeley, CA 94720, USA; Biophysics Graduate Group, University of California at Berkeley, Berkeley, CA 94720, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA.
| |
Collapse
|
19
|
Zhai B, Li H, Li S, Gu J, Zhang H, Zhang Y, Li H, Tian Y, Li G, Wang Y. Transcriptome analysis reveals FABP5 as a key player in the development of chicken abdominal fat, regulated by miR-122-5p targeting. BMC Genomics 2023; 24:386. [PMID: 37430185 PMCID: PMC10331962 DOI: 10.1186/s12864-023-09476-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 06/21/2023] [Indexed: 07/12/2023] Open
Abstract
BACKGROUND The development of abdominal fat and meat quality are closely related and can impact economic efficiency. In this study, we conducted transcriptome sequencing of the abdominal fat tissue of Gushi chickens at 6, 14, 22, and 30 weeks, and selected key miRNA-mRNA regulatory networks related to abdominal fat development through correlation analysis. RESULTS A total of 1893 differentially expressed genes were identified. Time series analysis indicated that at around 6 weeks, the development of chicken abdominal fat was extensively regulated by the TGF-β signaling pathway, Wnt signaling pathway, and PPAR signaling pathway. However, at 30 weeks of age, the apoptosis signaling pathway was the most significant, and correlation analysis revealed several genes highly correlated with abdominal fat development, including Fatty Acid Binding Protein 5 (FABP5). Based on miRNA transcriptome data, it was discovered that miR-122-5p is a potential target miRNA for FABP5. Cell experiments showed that miR-122-5p can directly target FABP5 to promote the differentiation of preadipocytes. CONCLUSION The present study confirms that the key gene FABP5 and its target gene miR-122-5p are critical regulatory factors in the development of chicken abdominal fat. These results provide new insights into the molecular regulatory mechanisms associated with the development of abdomen-al fat in chickens.
Collapse
Affiliation(s)
- Bin Zhai
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450046, China
| | - Hongtai Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450046, China
| | - Shuaihao Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450046, China
| | - Jinxing Gu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450046, China
| | - Hongyuan Zhang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450046, China
| | - Yanhua Zhang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450046, China
- Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou, 450046, P. R. China
| | - Hong Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450046, China
- Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou, 450046, P. R. China
| | - Yadong Tian
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450046, China
- Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou, 450046, P. R. China
| | - Guoxi Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450046, China.
- The Shennong Laboratory, Zhengzhou, 450046, China.
- Henan Key Laboratory for Innovation and Utilization of Chicken Germplasm Resources, Zhengzhou, 450046, P. R. China.
| | - Yongcai Wang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450046, China.
| |
Collapse
|
20
|
Kiran S, Mandal M, Rakib A, Bajwa A, Singh UP. miR-10a-3p modulates adiposity and suppresses adipose inflammation through TGF-β1/Smad3 signaling pathway. Front Immunol 2023; 14:1213415. [PMID: 37334370 PMCID: PMC10272755 DOI: 10.3389/fimmu.2023.1213415] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 05/19/2023] [Indexed: 06/20/2023] Open
Abstract
Background Obesity is a multifactorial disease characterized by an enhanced amount of fat and energy storage in adipose tissue (AT). Obesity appears to promote and maintain low-grade chronic inflammation by activating a subset of inflammatory T cells, macrophages, and other immune cells that infiltrate the AT. Maintenance of AT inflammation during obesity involves regulation by microRNAs (miRs), which also regulate the expression of genes implicated in adipocyte differentiation. This study aims to use ex vivo and in vitro approaches to evaluate the role and mechanism of miR-10a-3p in adipose inflammation and adipogenesis. Methods Wild-type BL/6 mice were placed on normal (ND) and high-fat diet (HFD) for 12 weeks and their obesity phenotype, inflammatory genes, and miRs expression were examined in the AT. We also used differentiated 3T3-L1 adipocytes for mechanistic in vitro studies. Results Microarray analysis allowed us to identify an altered set of miRs in the AT immune cells and Ingenuity pathway analysis (IPA) prediction demonstrated that miR-10a-3p expression was downregulated in AT immune cells in the HFD group as compared to ND. A molecular mimic of miR-10a-3p reduced expression of inflammatory M1 macrophages, cytokines, and chemokines, including transforming growth factor-beta 1 (TGF-β1), transcription factor Krüppel-like factor 4 (KLF4), and interleukin 17F (IL-17F) and induced expression of forkhead box P3 (FoxP3) in the immune cells isolated from AT of HFD-fed mice as compared to ND. In differentiated 3T3-L1 adipocytes, the miR-10a-3p mimics also reduced expression of proinflammatory genes and lipid accumulation, which plays a role in the dysregulation of AT function. In these cells, overexpression of miR-10a-3p reduced the expression of TGF-β1, Smad3, CHOP-10, and fatty acid synthase (FASN), relative to the control scramble miRs. Conclusion Our findings suggest that miR-10a-3p mimic mediates the TGF-β1/Smad3 signaling to improve metabolic markers and adipose inflammation. This study provides a new opportunity for the development of miR-10a-3p as a novel therapeutic for adipose inflammation, and its associated metabolic disorders.
Collapse
Affiliation(s)
- Sonia Kiran
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Mousumi Mandal
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Ahmed Rakib
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Amandeep Bajwa
- Department of Surgery, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Udai P. Singh
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
21
|
Expression and Function of BMP and Activin Membrane-Bound Inhibitor (BAMBI) in Chronic Liver Diseases and Hepatocellular Carcinoma. Int J Mol Sci 2023; 24:ijms24043473. [PMID: 36834884 PMCID: PMC9964332 DOI: 10.3390/ijms24043473] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 02/06/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
BAMBI (bone morphogenetic protein and activin membrane-bound inhibitor) is a transmembrane pseudoreceptor structurally related to transforming growth factor (TGF)-β type 1 receptors (TGF-β1Rs). BAMBI lacks a kinase domain and functions as a TGF-β1R antagonist. Essential processes such as cell differentiation and proliferation are regulated by TGF-β1R signaling. TGF-β is the best-studied ligand of TGF-βRs and has an eminent role in inflammation and fibrogenesis. Liver fibrosis is the end stage of almost all chronic liver diseases, such as non-alcoholic fatty liver disease, and at the moment, there is no effective anti-fibrotic therapy available. Hepatic BAMBI is downregulated in rodent models of liver injury and in the fibrotic liver of patients, suggesting that low BAMBI has a role in liver fibrosis. Experimental evidence convincingly demonstrated that BAMBI overexpression is able to protect against liver fibrosis. Chronic liver diseases have a high risk of hepatocellular carcinoma (HCC), and BAMBI was shown to exert tumor-promoting as well as tumor-protective functions. This review article aims to summarize relevant studies on hepatic BAMBI expression and its role in chronic liver diseases and HCC.
Collapse
|
22
|
Johnston EK, Abbott RD. Adipose Tissue Paracrine-, Autocrine-, and Matrix-Dependent Signaling during the Development and Progression of Obesity. Cells 2023; 12:407. [PMID: 36766750 PMCID: PMC9913478 DOI: 10.3390/cells12030407] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/19/2023] [Accepted: 01/23/2023] [Indexed: 01/27/2023] Open
Abstract
Obesity is an ever-increasing phenomenon, with 42% of Americans being considered obese (BMI ≥ 30) and 9.2% being considered morbidly obese (BMI ≥ 40) as of 2016. With obesity being characterized by an abundance of adipose tissue expansion, abnormal tissue remodeling is a typical consequence. Importantly, this pathological tissue expansion is associated with many alterations in the cellular populations and phenotypes within the tissue, lending to cellular, paracrine, mechanical, and metabolic alterations that have local and systemic effects, including diabetes and cardiovascular disease. In particular, vascular dynamics shift during the progression of obesity, providing signaling cues that drive metabolic dysfunction. In this review, paracrine-, autocrine-, and matrix-dependent signaling between adipocytes and endothelial cells is discussed in the context of the development and progression of obesity and its consequential diseases, including adipose fibrosis, diabetes, and cardiovascular disease.
Collapse
Affiliation(s)
| | - Rosalyn D. Abbott
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| |
Collapse
|
23
|
Maharjan BR, McLennan SV, Twigg SM, Williams PF. The Effect of TGFβ1 in Adipocyte on Inflammatory and Fibrotic Markers at Different Stages of Adipocyte Differentiation. PATHOPHYSIOLOGY 2022; 29:640-649. [PMID: 36548206 PMCID: PMC9788619 DOI: 10.3390/pathophysiology29040050] [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: 10/20/2022] [Revised: 11/18/2022] [Accepted: 11/21/2022] [Indexed: 11/24/2022] Open
Abstract
Transforming growth factor beta (TGFβ) is a versatile cytokine. Although a profibrotic role of TGFβ is well established, its effect on tissue inhibitor of metalloproteinase (TIMPs) and inflammatory mediators are incompletely described. This study investigates the profibrotic and pro-inflammatory role of TGFβ1 during adipocyte differentiation. NIH3T3L1 cells were used for the in vitro study and were differentiated by adding a standard differentiation mix either with rosiglitazone (R-Diff) or without (S-Diff). Recombinant TGFβ1 (2 ng/mL) was added to the undifferentiated preadipocyte during the commitment stage and at the terminal differentiation stage. TGFβ1 treatment significantly decreased adiponectin mRNA at both early commitment (>300 fold) and terminal differentiated cells [S-Diff (~33%) or R-Diff (~20%)]. TGFβ1 upregulated collagen VI mRNA and its regulators connective tissue growth factor (CCN2/CTGF), TIMP1 and TIMP3 mRNA levels in undifferentiated preadipocytes and adipocytes at commitment stage. But in the terminal differentiated adipocytes, changes in mRNA and protein of collagen VI and TIMP3 mRNA were not observed despite an increase in CCN2/CTGF, TIMP1 mRNA. Although TGFβ1 upregulated interleukin-6 (IL6) and monocyte chemoattractant protein-1 (MCP1) mRNA at all stages of differentiation, decreased tumor necrosis factor-α (TNFα) mRNA was observed early in adipocyte differentiation. This study highlights the complex role of TGFβ1 on extracellular matrix (ECM) remodeling and inflammatory markers in stimulating both synthetic and inhibitory markers of fibrosis at different stages of adipocyte differentiation.
Collapse
Affiliation(s)
- Babu Raja Maharjan
- Greg Brown Diabetes & Endocrinology Laboratory, Sydney Medical School, University of Sydney, Sydney, NSW 2006, Australia
- School of Medicine, Department of Biochemistry, Patan Academy of Health Sciences, Lalitpur 44700, Nepal
- Correspondence: (B.R.M.); (P.F.W.); Tel.: +61-2-8627-1889 (B.R.M. & P.F.W.)
| | - Susan V. McLennan
- Greg Brown Diabetes & Endocrinology Laboratory, Sydney Medical School, University of Sydney, Sydney, NSW 2006, Australia
- New South Wales Health Pathology, Sydney, NSW 2050, Australia
| | - Stephen M. Twigg
- Greg Brown Diabetes & Endocrinology Laboratory, Sydney Medical School, University of Sydney, Sydney, NSW 2006, Australia
- Department of Endocrinology, Royal Prince Alfred Hospital, Sydney, NSW 2006, Australia
| | - Paul F. Williams
- Greg Brown Diabetes & Endocrinology Laboratory, Sydney Medical School, University of Sydney, Sydney, NSW 2006, Australia
- Correspondence: (B.R.M.); (P.F.W.); Tel.: +61-2-8627-1889 (B.R.M. & P.F.W.)
| |
Collapse
|
24
|
Lee ES, Guo T, Srivastava RK, Shabbir A, Ibáñez CF. Activin receptor ALK4 promotes adipose tissue hyperplasia by suppressing differentiation of adipocyte precursors. J Biol Chem 2022; 299:102716. [PMID: 36403856 PMCID: PMC9758429 DOI: 10.1016/j.jbc.2022.102716] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/09/2022] [Accepted: 11/10/2022] [Indexed: 11/19/2022] Open
Abstract
Adipocyte hyperplasia and hypertrophy are the two main processes contributing to adipose tissue expansion, yet the mechanisms that regulate and balance their involvement in obesity are incompletely understood. Activin B/GDF-3 receptor ALK7 is expressed in mature adipocytes and promotes adipocyte hypertrophy upon nutrient overload by suppressing adrenergic signaling and lipolysis. In contrast, the role of ALK4, the canonical pan-activin receptor, in adipose tissue is unknown. Here, we report that, unlike ALK7, ALK4 is preferentially expressed in adipocyte precursors, where it suppresses differentiation, allowing proliferation and adipose tissue expansion. ALK4 expression in adipose tissue increases upon nutrient overload and positively correlates with fat depot mass and body weight, suggesting a role in adipose tissue hyperplasia during obesity. Mechanistically, ALK4 signaling suppresses expression of CEBPα and PPARγ, two master regulators of adipocyte differentiation. Conversely, ALK4 deletion enhances CEBPα/PPARγ expression and induces premature adipocyte differentiation, which can be rescued by CEBPα knockdown. These results clarify the function of ALK4 in adipose tissue and highlight the contrasting roles of the two activin receptors in the regulation of adipocyte hyperplasia and hypertrophy during obesity.
Collapse
Affiliation(s)
- Ee-Soo Lee
- Department of Physiology and Life Sciences Institute, National University of, Singapore, Singapore
| | - Tingqing Guo
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Raj Kamal Srivastava
- Department of Physiology and Life Sciences Institute, National University of, Singapore, Singapore
| | - Assim Shabbir
- Division of General Surgery, University Surgical Cluster, National University, Health System, Singapore
| | - Carlos F Ibáñez
- Department of Physiology and Life Sciences Institute, National University of, Singapore, Singapore; Department of Neuroscience, Karolinska Institute, Stockholm, Sweden; Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University School of Life Sciences, Beijing, China; Chinese Institute for Brain Research, Life Science Park, Beijing, China; Stellenbosch Institute for Advanced Study, Wallenberg Research Centre at Stellenbosch University, Stellenbosch, South Africa.
| |
Collapse
|
25
|
Yang L, Wang H, Hao W, Li T, Fang H, Bai H, Yan P, Wei S. TGFβ3 regulates adipogenesis of bovine subcutaneous preadipocytes via typical Smad and atypical MAPK signaling pathways. ELECTRON J BIOTECHN 2022. [DOI: 10.1016/j.ejbt.2022.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
26
|
Singh K, Sachan N, Ene T, Dabovic B, Rifkin D. Latent Transforming Growth Factor β Binding Protein 3 Controls Adipogenesis. Matrix Biol 2022; 112:155-170. [PMID: 35933071 DOI: 10.1016/j.matbio.2022.08.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 07/29/2022] [Accepted: 08/02/2022] [Indexed: 11/24/2022]
Abstract
Transforming growth factor-beta (TGFβ) is released from cells as part of a trimeric latent complex consisting of TGFβ, the TGFβ propeptides, and either a latent TGFβ binding protein (LTBP) or glycoprotein-A repetitions predominant (GARP) protein. LTBP1 and 3 modulate latent TGFβ function with respect to secretion, matrix localization, and activation and, therefore, are vital for the proper function of the cytokine in a number of tissues. TGFβ modulates stem cell differentiation into adipocytes (adipogenesis), but the potential role of LTBPs in this process has not been studied. We observed that 72 h post adipogenesis initiation Ltbp1, 2, and 4 expression levels decrease by 74-84%, whereas Ltbp3 expression levels remain constant during adipogenesis. We found that LTBP3 silencing in C3H/10T1/2 cells reduced adipogenesis, as measured by the percentage of cells with lipid vesicles and the expression of the transcription factor peroxisome proliferator-activated receptor gamma (PPARγ). Lentiviral mediated expression of an Ltbp3 mRNA resistant to siRNA targeting rescued the phenotype, validating siRNA specificity. Knockdown (KD) of Ltbp3 expression in 3T3-L1, M2, and primary bone marrow stromal cells (BMSC) indicated a similar requirement for Ltbp3. Epididymal and inguinal white adipose tissue fat pad weights of Ltbp3-/- mice were reduced by 62% and 57%, respectively, compared to wild-type mice. Inhibition of adipogenic differentiation upon LTBP3 loss is mediated by TGFβ, as TGFβ neutralizing antibody and TGFβ receptor I kinase blockade rescue the LTBP3 KD phenotype. These results indicate that LTBP3 has a TGFβ-dependent function in adipogenesis both in vitro and possibly in vivo.
Collapse
Affiliation(s)
- Karan Singh
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, USA
| | - Nalani Sachan
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, USA
| | - Taylor Ene
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, USA
| | - Branka Dabovic
- Division of Advanced Research Technologies, New York University Grossman School of Medicine, New York, NY, USA
| | - Daniel Rifkin
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, USA; Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
27
|
Duch P, Díaz-Valdivia N, Ikemori R, Gabasa M, Radisky ES, Arshakyan M, Gea-Sorlí S, Mateu-Bosch A, Bragado P, Carrasco JL, Mori H, Ramírez J, Teixidó C, Reguart N, Fillat C, Radisky DC, Alcaraz J. Aberrant TIMP-1 overexpression in tumor-associated fibroblasts drives tumor progression through CD63 in lung adenocarcinoma. Matrix Biol 2022; 111:207-225. [PMID: 35787446 PMCID: PMC9667815 DOI: 10.1016/j.matbio.2022.06.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 06/10/2022] [Accepted: 06/29/2022] [Indexed: 12/29/2022]
Abstract
Tissue inhibitor of metalloproteinase-1 (TIMP-1) is an important regulator of extracellular matrix turnover that has been traditionally regarded as a potential tumor suppressor owing to its inhibitory effects of matrix metalloproteinases. Intriguingly, this interpretation has been challenged by the consistent observation that increased expression of TIMP-1 is associated with poor prognosis in virtually all cancer types including lung cancer, supporting a tumor-promoting function. However, how TIMP-1 is dysregulated within the tumor microenvironment and how it drives tumor progression in lung cancer is poorly understood. We analyzed the expression of TIMP-1 and its cell surface receptor CD63 in two major lung cancer subtypes: lung adenocarcinoma (ADC) and squamous cell carcinoma (SCC), and defined the tumor-promoting effects of their interaction. We found that TIMP-1 is aberrantly overexpressed in tumor-associated fibroblasts (TAFs) in ADC compared to SCC. Mechanistically, TIMP-1 overexpression was mediated by the selective hyperactivity of the pro-fibrotic TGF-β1/SMAD3 pathway in ADC-TAFs. Likewise, CD63 was upregulated in ADC compared to SCC cells. Genetic analyses revealed that TIMP-1 secreted by TGF-β1-activated ADC-TAFs is both necessary and sufficient to enhance growth and invasion of ADC cancer cells in culture, and that tumor cell expression of CD63 was required for these effects. Consistently, in vivo analyses revealed that ADC cells co-injected with fibroblasts with reduced SMAD3 or TIMP-1 expression into immunocompromised mice attenuated tumor aggressiveness compared to tumors bearing parental fibroblasts. We also found that high TIMP1 and CD63 mRNA levels combined define a stronger prognostic biomarker than TIMP1 alone. Our results identify an excessive stromal TIMP-1 within the tumor microenvironment selectively in lung ADC, and implicate it in a novel tumor-promoting TAF-carcinoma crosstalk, thereby pointing to TIMP-1/CD63 interaction as a novel therapeutic target in lung cancer.
Collapse
Affiliation(s)
- Paula Duch
- Unit of Biophysics and Bioengineering, Department of Biomedicine, School of Medicine and Health Sciences, Universitat de Barcelona, Barcelona 08036, Spain
| | - Natalia Díaz-Valdivia
- Unit of Biophysics and Bioengineering, Department of Biomedicine, School of Medicine and Health Sciences, Universitat de Barcelona, Barcelona 08036, Spain
| | - Rafael Ikemori
- Unit of Biophysics and Bioengineering, Department of Biomedicine, School of Medicine and Health Sciences, Universitat de Barcelona, Barcelona 08036, Spain
| | - Marta Gabasa
- Unit of Biophysics and Bioengineering, Department of Biomedicine, School of Medicine and Health Sciences, Universitat de Barcelona, Barcelona 08036, Spain; Thoracic Oncology Unit, Hospital Clinic Barcelona, Barcelona 08036, Spain
| | - Evette S Radisky
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, United States
| | - Marselina Arshakyan
- Unit of Biophysics and Bioengineering, Department of Biomedicine, School of Medicine and Health Sciences, Universitat de Barcelona, Barcelona 08036, Spain
| | - Sabrina Gea-Sorlí
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona 08036, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid 08029, Spain
| | - Anna Mateu-Bosch
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona 08036, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid 08029, Spain
| | - Paloma Bragado
- Instituto de Investigaciones Sanitarias San Carlos (IdISSC), Madrid, 28040, Spain; Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad Complutense, 28040 Madrid, Spain
| | - Josep Lluís Carrasco
- Unit of Biostatistics, Department of Basic Clinical Practice, School of Medicine and Health Sciences, Universitat de Barcelona, Barcelona, Spain
| | - Hidetoshi Mori
- Center for Immunology and Infectious Diseases, University of California Davis, Davis, CA 95616, United States
| | - Josep Ramírez
- Thoracic Oncology Unit, Hospital Clinic Barcelona, Barcelona 08036, Spain; Pathology Service, Hospital Clínic de Barcelona, Barcelona 08036, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Cristina Teixidó
- Thoracic Oncology Unit, Hospital Clinic Barcelona, Barcelona 08036, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona 08036, Spain; Pathology Service, Hospital Clínic de Barcelona, Barcelona 08036, Spain
| | - Noemí Reguart
- Thoracic Oncology Unit, Hospital Clinic Barcelona, Barcelona 08036, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona 08036, Spain
| | - Cristina Fillat
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona 08036, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid 08029, Spain; Department of Medicine, School of Medicine and Health Sciences, Universitat de Barcelona, Barcelona, Spain
| | - Derek C Radisky
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, United States
| | - Jordi Alcaraz
- Unit of Biophysics and Bioengineering, Department of Biomedicine, School of Medicine and Health Sciences, Universitat de Barcelona, Barcelona 08036, Spain; Thoracic Oncology Unit, Hospital Clinic Barcelona, Barcelona 08036, Spain; Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona 08028, Spain.
| |
Collapse
|
28
|
Targeting of the Peritumoral Adipose Tissue Microenvironment as an Innovative Antitumor Therapeutic Strategy. Biomolecules 2022; 12:biom12050702. [PMID: 35625629 PMCID: PMC9138344 DOI: 10.3390/biom12050702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/09/2022] [Accepted: 05/11/2022] [Indexed: 12/03/2022] Open
Abstract
The tumor microenvironment (TME) plays a key role in promoting and sustaining cancer growth. Adipose tissue (AT), due to its anatomical distribution, is a prevalent component of TME, and contributes to cancer development and progression. Cancer-associated adipocytes (CAAs), reprogrammed by cancer stem cells (CSCs), drive cancer progression by releasing metabolites and inflammatory adipokines. In this review, we highlight the mechanisms underlying the bidirectional crosstalk among CAAs, CSCs, and stromal cells. Moreover, we focus on the recent advances in the therapeutic targeting of adipocyte-released factors as an innovative strategy to counteract cancer progression.
Collapse
|
29
|
Huang CJ, Choo KB, Chen CF. The MicroRNA-Signaling-Peroxisome Proliferator-Activated Receptor Gamma Connection in the Modulation of Adipogenesis: Bioinformatics Projection on Chicken. Poult Sci 2022; 101:101950. [PMID: 35689996 PMCID: PMC9192975 DOI: 10.1016/j.psj.2022.101950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 03/19/2022] [Accepted: 04/15/2022] [Indexed: 10/29/2022] Open
|
30
|
Cho JH, Lee JH, Lee KM, Lee CK, Shin DM. BMP-2 Induced Signaling Pathways and Phenotypes: Comparisons Between Senescent and Non-senescent Bone Marrow Mesenchymal Stem Cells. Calcif Tissue Int 2022; 110:489-503. [PMID: 34714366 DOI: 10.1007/s00223-021-00923-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 10/04/2021] [Indexed: 10/20/2022]
Abstract
The use of BMP-2 in orthopedic surgery is limited by uncertainty surrounding its effects on the differentiation of mesenchymal stem cells (MSCs) and how this is affected by cellular aging. This study compared the effects of recombinant human BMP-2 (rhBMP-2) on osteogenic and adipogenic differentiation between senescent and non-senescent MSCs. Senescent and non-senescent MSCs were cultured in osteogenic and adipogenic differentiation medium containing various concentrations of rhBMP-2. The phenotypes of these cells were compared by performing a calcium assay, adipogenesis assay, staining, real-time PCR, western blotting, and microarray analysis. rhBMP-2 induced osteogenic differentiation to a lesser extent (P < 0.001 and P = 0.005 for alkaline phosphatase activity and Ca2+ release) in senescent MSCs regardless of dose-dependent increase in both cells. However, the induction of adipogenic differentiation by rhBMP-2 was comparable between them. There was no difference between these two groups of cells in the adipogenesis assay (P = 0.279) and their expression levels of PPARγ were similar. Several genes such as CHRDL1, NOG, SMAD1, SMAD7, and FST encoding transcription factors were proposed to underlie the different responses of senescent and non-senescent MSCs to rhBMP-2 in microarray analyses. Furthermore, inflammatory, adipogenic, or cell death-related signaling pathways such as NF-kB or p38-MAPK pathways were upregulated by BMP-2 in senescent MSCs, whereas bone forming signaling pathways involving BMP, SMAD, and TGF- ß were upregulated in non-senescent MSCs as expected. This phenomenon explains bone forming dominance by non-senescent MSCs and possible frequent complications such as seroma, osteolysis, or neuritis in senescent MSCs during BMP-2 use in orthopedic surgery.
Collapse
Affiliation(s)
- Jae Hwan Cho
- Department of Orthopedic Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jae Hyup Lee
- Department of Orthopedic Surgery, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, Republic of Korea.
- Department of Orthopedic Surgery, SMG-SNU Boramae Medical Center, Boramae-ro 5-gil, Dongjak-Gu, Seoul, Republic of Korea.
| | - Kyung Mee Lee
- Department of Orthopedic Surgery, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, Republic of Korea
| | - Choon-Ki Lee
- Department of Orthopedic Surgery, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, Republic of Korea
| | - Dong-Myung Shin
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
31
|
Tang C, Liang D, Qiu Y, Zhu J, Tang G. Omentin‑1 induces osteoblast viability and differentiation via the TGF‑β/Smad signaling pathway in osteoporosis. Mol Med Rep 2022; 25:132. [PMID: 35179221 PMCID: PMC8867465 DOI: 10.3892/mmr.2022.12648] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 07/13/2021] [Indexed: 11/25/2022] Open
Abstract
Osteoporosis is a bone-related disease that results from impaired bone formation and excessive bone resorption. The potential value of adipokines has been investigated previously, due to their influence on osteogenesis. However, the osteogenic effects induced by omentin-1 remain unclear. The aim of the present study was to determine the regulatory effects of omentin-1 on osteoblast viability and differentiation, as well as to explore the underlying molecular mechanism. The present study investigated the effects of omentin-1 on the viability and differentiation of mouse pre-osteoblast cells (MC3T3-E1) using quantitative and qualitative measures. A Cell Counting Kit-8 assay was used to assess the viability of MC3T3-E1 cells following treatment with different doses of omentin-1. Omentin-1 and bone morphogenetic protein (BMP) inhibitor were added to osteogenic induction mediums in different ways to assess their effect. The alkaline phosphatase (ALP) activity and Alizarin Red S (ARS) staining of MC3T3-E1 cells treated with omentin-1 and/or BMP inhibitor were used to examine the effects of omentin-1 on differentiation and mineralization. Western blotting was used to further explore its potential mechanism, and to study the role of omentin-1 on the viability and differentiation of osteoblasts. The results showed that omentin-1 altered the viability of MC3T3-E1 cells in a dose-dependent manner. Omentin-1 treatment significantly increased the expression of members of the TGF-β/Smad signaling pathway. In the omentin-1 group, the ALP activity of the MC3T3-E1 cells was increased, and the ARS staining area was also increased. The mRNA and protein expression levels of BMP2, Runt-related transcription factor 2, collagen1, osteopontin, osteocalcin and osterix in the omentin-1 group were also significantly upregulated. All these effects were reversed following treatment with SIS3 HCl. These results demonstrated that omentin-1 can significantly promote osteoblast viability and differentiation via the TGF-β/Smad signaling pathway, thereby promoting bone formation and preventing osteoporosis.
Collapse
Affiliation(s)
- Cuisong Tang
- Department of Radiology, Clinical Medical College of Shanghai Tenth People's Hospital of Nanjing Medical University, Shanghai 200072, P.R. China
| | - Dengpan Liang
- Department of Cardiology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Yuyou Qiu
- Department of Radiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Jingqi Zhu
- Department of Radiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, P.R. China
| | - Guangyu Tang
- Department of Radiology, Clinical Medical College of Shanghai Tenth People's Hospital of Nanjing Medical University, Shanghai 200072, P.R. China
| |
Collapse
|
32
|
Rodríguez‐Barrueco R, Latorre J, Devis‐Jáuregui L, Lluch A, Bonifaci N, Llobet FJ, Olivan M, Coll‐Iglesias L, Gassner K, Davis ML, Moreno‐Navarrete JM, Castells‐Nobau A, Plata‐Peña L, Dalmau‐Pastor M, Höring M, Liebisch G, Olkkonen VM, Arnoriaga‐Rodríguez M, Ricart W, Fernández‐Real JM, Silva JM, Ortega FJ, Llobet‐Navas D. A microRNA Cluster Controls Fat Cell Differentiation and Adipose Tissue Expansion By Regulating SNCG. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2104759. [PMID: 34898027 PMCID: PMC8811811 DOI: 10.1002/advs.202104759] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Indexed: 05/08/2023]
Abstract
The H19X-encoded miR-424(322)/503 cluster regulates multiple cellular functions. Here, it is reported for the first time that it is also a critical linchpin of fat mass expansion. Deletion of this miRNA cluster in mice results in obesity, while increasing the pool of early adipocyte progenitors and hypertrophied adipocytes. Complementary loss and gain of function experiments and RNA sequencing demonstrate that miR-424(322)/503 regulates a conserved genetic program involved in the differentiation and commitment of white adipocytes. Mechanistically, it is demonstrated that miR-424(322)/503 targets γ-Synuclein (SNCG), a factor that mediates this program rearrangement by controlling metabolic functions in fat cells, allowing adipocyte differentiation and adipose tissue enlargement. Accordingly, diminished miR-424(322) in mice and obese humans co-segregate with increased SNCG in fat and peripheral blood as mutually exclusive features of obesity, being normalized upon weight loss. The data unveil a previously unknown regulatory mechanism of fat mass expansion tightly controlled by the miR-424(322)/503 through SNCG.
Collapse
Affiliation(s)
- Ruth Rodríguez‐Barrueco
- Molecular Mechanisms and Experimental Therapy in Oncology‐Oncobell ProgramBellvitge Biomedical Research Institute (IDIBELL)L'Hospitalet de Llobregat08908Spain
- Anatomy UnitDepartment of Pathology and Experimental TherapySchool of MedicineUniversity of Barcelona (UB)L'Hospitalet de Llobregat08907Spain
| | - Jessica Latorre
- Department of DiabetesEndocrinology, and Nutrition (UDEN)Institut d'Investigació Biomèdica de Girona (IDIBGI)Salt17190Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y la Nutrición (CIBEROBN)Instituto de Salud Carlos III (ISCIII)Madrid28029Spain
| | - Laura Devis‐Jáuregui
- Molecular Mechanisms and Experimental Therapy in Oncology‐Oncobell ProgramBellvitge Biomedical Research Institute (IDIBELL)L'Hospitalet de Llobregat08908Spain
| | - Aina Lluch
- Molecular Mechanisms and Experimental Therapy in Oncology‐Oncobell ProgramBellvitge Biomedical Research Institute (IDIBELL)L'Hospitalet de Llobregat08908Spain
- Department of DiabetesEndocrinology, and Nutrition (UDEN)Institut d'Investigació Biomèdica de Girona (IDIBGI)Salt17190Spain
| | - Nuria Bonifaci
- Molecular Mechanisms and Experimental Therapy in Oncology‐Oncobell ProgramBellvitge Biomedical Research Institute (IDIBELL)L'Hospitalet de Llobregat08908Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)Instituto de Salud Carlos III, (ISCIII)Madrid28029Spain
| | - Francisco J. Llobet
- Molecular Mechanisms and Experimental Therapy in Oncology‐Oncobell ProgramBellvitge Biomedical Research Institute (IDIBELL)L'Hospitalet de Llobregat08908Spain
| | - Mireia Olivan
- Molecular Mechanisms and Experimental Therapy in Oncology‐Oncobell ProgramBellvitge Biomedical Research Institute (IDIBELL)L'Hospitalet de Llobregat08908Spain
- Anatomy UnitDepartment of Pathology and Experimental TherapySchool of MedicineUniversity of Barcelona (UB)L'Hospitalet de Llobregat08907Spain
| | - Laura Coll‐Iglesias
- Molecular Mechanisms and Experimental Therapy in Oncology‐Oncobell ProgramBellvitge Biomedical Research Institute (IDIBELL)L'Hospitalet de Llobregat08908Spain
| | - Katja Gassner
- Molecular Mechanisms and Experimental Therapy in Oncology‐Oncobell ProgramBellvitge Biomedical Research Institute (IDIBELL)L'Hospitalet de Llobregat08908Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)Instituto de Salud Carlos III, (ISCIII)Madrid28029Spain
| | - Meredith L. Davis
- Molecular Mechanisms and Experimental Therapy in Oncology‐Oncobell ProgramBellvitge Biomedical Research Institute (IDIBELL)L'Hospitalet de Llobregat08908Spain
- Department of PathologyDuke University School of MedicineDurhamNC27710USA
| | - José M. Moreno‐Navarrete
- Department of DiabetesEndocrinology, and Nutrition (UDEN)Institut d'Investigació Biomèdica de Girona (IDIBGI)Salt17190Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y la Nutrición (CIBEROBN)Instituto de Salud Carlos III (ISCIII)Madrid28029Spain
| | - Anna Castells‐Nobau
- Department of DiabetesEndocrinology, and Nutrition (UDEN)Institut d'Investigació Biomèdica de Girona (IDIBGI)Salt17190Spain
| | - Laura Plata‐Peña
- Molecular Mechanisms and Experimental Therapy in Oncology‐Oncobell ProgramBellvitge Biomedical Research Institute (IDIBELL)L'Hospitalet de Llobregat08908Spain
| | - Miki Dalmau‐Pastor
- Anatomy UnitDepartment of Pathology and Experimental TherapySchool of MedicineUniversity of Barcelona (UB)L'Hospitalet de Llobregat08907Spain
- MIFAS by GRECMIP (Minimally Invasive Foot and Ankle Society)Merignac33700France
| | - Marcus Höring
- Institute of Clinical Chemistry and Laboratory MedicineRegensburg University HospitalRegensburg93053Germany
| | - Gerhard Liebisch
- Institute of Clinical Chemistry and Laboratory MedicineRegensburg University HospitalRegensburg93053Germany
| | - Vesa M. Olkkonen
- Minerva Foundation Institute for Medical Research (Biomedicum 2U)and Department of AnatomyFaculty of MedicineUniversity of HelsinkiHelsinki00290Finland
| | - Maria Arnoriaga‐Rodríguez
- Department of DiabetesEndocrinology, and Nutrition (UDEN)Institut d'Investigació Biomèdica de Girona (IDIBGI)Salt17190Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y la Nutrición (CIBEROBN)Instituto de Salud Carlos III (ISCIII)Madrid28029Spain
| | - Wifredo Ricart
- Department of DiabetesEndocrinology, and Nutrition (UDEN)Institut d'Investigació Biomèdica de Girona (IDIBGI)Salt17190Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y la Nutrición (CIBEROBN)Instituto de Salud Carlos III (ISCIII)Madrid28029Spain
| | - José M. Fernández‐Real
- Department of DiabetesEndocrinology, and Nutrition (UDEN)Institut d'Investigació Biomèdica de Girona (IDIBGI)Salt17190Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y la Nutrición (CIBEROBN)Instituto de Salud Carlos III (ISCIII)Madrid28029Spain
| | - José M. Silva
- Department of PathologyIcahn School of Medicine at Mount SinaiNew YorkNY10029USA
| | - Francisco J. Ortega
- Department of DiabetesEndocrinology, and Nutrition (UDEN)Institut d'Investigació Biomèdica de Girona (IDIBGI)Salt17190Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y la Nutrición (CIBEROBN)Instituto de Salud Carlos III (ISCIII)Madrid28029Spain
| | - David Llobet‐Navas
- Molecular Mechanisms and Experimental Therapy in Oncology‐Oncobell ProgramBellvitge Biomedical Research Institute (IDIBELL)L'Hospitalet de Llobregat08908Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)Instituto de Salud Carlos III, (ISCIII)Madrid28029Spain
| |
Collapse
|
33
|
Liu S, Wang L, Ling D, Valencak TG, You W, Shan T. Potential key factors involved in regulating adipocyte dedifferentiation. J Cell Physiol 2021; 237:1639-1647. [PMID: 34796916 DOI: 10.1002/jcp.30637] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 10/26/2021] [Accepted: 11/09/2021] [Indexed: 12/11/2022]
Abstract
Adipocytes are the key constituents of adipose tissue, and their de-differentiation process has been widely observed in physiological and pathological conditions. For obese people, the promotion of adipocyte de-differentiation or maintenance of an undifferentiated state of adipocytes may help to improve their metabolic condition. Thus, understanding the regulatory mechanisms of adipocyte de-differentiation is necessary for treating metabolic diseases. Attractively, in addition to intracellular signals regulating adipocyte de-differentiation, external factors such as temperature and pressure also affect adipocyte de-differentiation. In this review, we summarize the recent progress in the field and discuss the regulatory roles and mechanisms of involved endogenous and exogenous factors during the process of de-differentiation.
Collapse
Affiliation(s)
- Shiqi Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China.,Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, Zhejiang, China.,Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Liyi Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China.,Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, Zhejiang, China.,Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Defeng Ling
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China.,Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, Zhejiang, China.,Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Teresa G Valencak
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Wenjing You
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China.,Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, Zhejiang, China.,Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Tizhong Shan
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang, China.,Key Laboratory of Molecular Animal Nutrition, Ministry of Education, Hangzhou, Zhejiang, China.,Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, Zhejiang, China
| |
Collapse
|
34
|
Kumari R, Irudayam MJ, Al Abdallah Q, Jones TL, Mims TS, Puchowicz MA, Pierre JF, Brown CW. SMAD2 and SMAD3 differentially regulate adiposity and the growth of subcutaneous white adipose tissue. FASEB J 2021; 35:e22018. [PMID: 34731499 DOI: 10.1096/fj.202101244r] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/28/2021] [Accepted: 10/13/2021] [Indexed: 11/11/2022]
Abstract
Adipose tissue is the primary site of energy storage, playing important roles in health. While adipose research largely focuses on obesity, fat also has other critical functions, producing adipocytokines and contributing to normal nutrient metabolism, which in turn play important roles in satiety and total energy homeostasis. SMAD2/3 proteins are downstream mediators of activin signaling, which regulate critical preadipocyte and mature adipocyte functions. Smad2 global knockout mice exhibit embryonic lethality, whereas global loss of Smad3 protects mice against diet-induced obesity. The direct contributions of Smad2 and Smad3 in adipose tissues, however, are unknown. Here, we sought to determine the primary effects of adipocyte-selective reduction of Smad2 or Smad3 on diet-induced adiposity using Smad2 or Smad3 "floxed" mice intercrossed with Adiponectin-Cre mice. Additionally, we examined visceral and subcutaneous preadipocyte differentiation efficiency in vitro. Almost all wild type subcutaneous preadipocytes differentiated into mature adipocytes. In contrast, visceral preadipocytes differentiated poorly. Exogenous activin A suppressed differentiation of preadipocytes from both depots. Smad2 conditional knockout (Smad2cKO) mice did not exhibit significant effects on weight gain, irrespective of diet, whereas Smad3 conditional knockout (Smad3cKO) male mice displayed a trend of reduced body weight on high-fat diet. On both diets, Smad3cKO mice displayed an adipose depot-selective phenotype, with a significant reduction in subcutaneous fat mass but not visceral fat mass. Our data suggest that Smad3 is an important contributor to the maintenance of subcutaneous white adipose tissue in a sex-selective fashion. These findings have implications for understanding SMAD-mediated, depot selective regulation of adipocyte growth and differentiation.
Collapse
Affiliation(s)
- Roshan Kumari
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, USA.,Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Maria Johnson Irudayam
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Qusai Al Abdallah
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Tamekia L Jones
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, USA.,Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA.,Children's Foundation Research Institute, Memphis, Tennessee, USA
| | - Tahliyah S Mims
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Michelle A Puchowicz
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Joseph F Pierre
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Chester W Brown
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, USA.,Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, Tennessee, USA.,Le Bonheur Children's Hospital, Memphis, Tennessee, USA
| |
Collapse
|
35
|
Bio-engineering a prevascularized human tri-layered skin substitute containing a hypodermis. Acta Biomater 2021; 134:215-227. [PMID: 34303011 DOI: 10.1016/j.actbio.2021.07.033] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 07/14/2021] [Accepted: 07/15/2021] [Indexed: 12/17/2022]
Abstract
Severe injuries to skin including hypodermis require full-thickness skin replacement. Here, we bioengineered a tri-layered human skin substitute (TLSS) containing the epidermis, dermis, and hypodermis. The hypodermal layer was generated by differentiation of human adipose stem cells (ASC) in a collagen type I hydrogel and combined with a prevascularized dermis consisting of human dermal microvascular endothelial cells and fibroblasts, which arranged into a dense vascular network. Subsequently, keratinocytes were seeded on top to generate the epidermal layer of the TLSS. The differentiation of ASC into adipocytes was confirmed in vitro on the mRNA level by the presence of adiponectin, as well as by the expression of perilipin and FABP-4 proteins. Moreover, functional characteristics of the hypodermis in vitro and in vivo were evaluated by Oil Red O, BODIPY, and AdipoRed stainings visualizing intracellular lipid droplets. Further, we demonstrated that both undifferentiated ASC and mature adipocytes present in the hypodermis influenced the keratinocyte maturation and homeostasis in the skin substitutes after transplantation. In particular, an enhanced secretion of TGF-β1 by these cells affected the epidermal morphogenesis as assessed by the expression of key proteins involved in the epidermal differentiation including cytokeratin 1, 10, 19 and cornified envelope formation such as involucrin. Here, we propose a novel functional hypodermal-dermo-epidermal tri-layered skin substitute containing blood capillaries that efficiently promote regeneration of skin defects. STATEMENT OF SIGNIFICANCE: The main objective of this study was to develop and assess the usefulness of a tri-layered human prevascularized skin substitute (TLSS) containing an epidermis, dermis, and hypodermis. The bioengineered hypodermis was generated from human adipose mesenchymal stem cells (ASC) and combined with a prevascularized dermis and epidermis. The TLSS represents an exceptional model for studying the role of cell-cell and cell-matrix interactions in vitro and in vivo. In particular, we observed that enhanced secretion of TGF-β1 in the hypodermis exerted a profound impact on fibroblast and keratinocyte differentiation, as well as epidermal barrier formation and homeostasis. Therefore, improved understanding of the cell-cell interactions in such a physiological skin model is essential to gain insights into different aspects of wound healing.
Collapse
|
36
|
Felicidade I, Bocchi M, Ramos MRZ, Carlos LDO, Wagner NRF, Campos ACL, Ribeiro LR, Mantovani MS, Watanabe MAE, Vitiello GAF. Transforming growth factor beta 1 (TGFβ1) plasmatic levels and haplotype structures in obesity: a role for TGFβ1 in steatosis development. Mol Biol Rep 2021; 48:6401-6411. [PMID: 34403036 DOI: 10.1007/s11033-021-06640-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 08/10/2021] [Indexed: 11/25/2022]
Abstract
BACKGROUND Obesity is considered a chronic inflammatory disease and transforming growth factor beta 1 (TGFβ1) might exert important roles in disease pathogenesis regulating adipocyte differentiation and immune-inflammatory environment. However, the role of this cytokine as a biomarker in obesity is poorly addressed. Therefore, the present study aimed to evaluate the impact of TGFB1 polymorphisms and TGFβ1 plasmatic levels in obesity METHODS AND RESULTS: TGFB1 promoter region polymorphisms (rs1800468, G-800A and rs1800469, C-509 T) were evaluated in 75 obese patients and 45 eutrophic patients through PCR-RFLP and plasmatic TGFβ1 was quantified through ELISA from 37 of the obese patients, and correlations with clinical and biochemical parameters were tested. Despite no association was found between TGFB1 polymorphisms and obesity susceptibility, several correlations with clinical data were noted. Among others, AC haplotype negatively correlated with plasmatic TGFβ1, while plasmatic TGFβ1 negatively correlated with C-reactive protein and positively correlated with liver abnormalities on ultrasound and, specifically, with steatosis presence and degree. Conversely, GT haplotype, which associates with higher TGFβ1 production, was also positively correlated with the same parameters of liver abnormalities. Further, plasmatic vitamin D negatively correlated with TGFβ1, while positively correlated with AC haplotype. CONCLUSION Overall, the results indicate that TGFβ1 might exert important roles in obesity pathophysiology and correlate with biochemical and clinical parameters both at systemic protein as well as at genetic level. Importantly, the consistent positive correlation at both levels with steatosis might suggest this cytokine as a biomarker for this hepatic abnormality in obese patients.
Collapse
Affiliation(s)
- Ingrid Felicidade
- Department of General Biology, Biological Sciences Center, Londrina State University (UEL), Londrina, PR, Brazil
- School of Medicine, Department of Pathology, São Paulo State University (UNESP), São Paulo, SP, Brazil
| | - Mayara Bocchi
- Department of Pathological Sciences, Biological Sciences Center, Londrina State University (UEL), Londrina, PR, Brazil
| | | | | | | | | | - Lúcia Regina Ribeiro
- School of Medicine, Department of Pathology, São Paulo State University (UNESP), São Paulo, SP, Brazil
| | - Mário Sérgio Mantovani
- Department of General Biology, Biological Sciences Center, Londrina State University (UEL), Londrina, PR, Brazil
| | - Maria Angelica Ehara Watanabe
- Department of Pathological Sciences, Biological Sciences Center, Londrina State University (UEL), Londrina, PR, Brazil
| | - Glauco Akelinghton Freire Vitiello
- Department of Pathological Sciences, Biological Sciences Center, Londrina State University (UEL), Londrina, PR, Brazil.
- Laboratory of DNA Polymorphisms and Immunology, Department of Pathological Sciences, Biological Sciences Center, State University of Londrina, PR445, Km 380 Celso Garcia Cid highway, Londrina, PR, 86057-970, Brazil.
| |
Collapse
|
37
|
Smad2/3 Activation Regulates Smad1/5/8 Signaling via a Negative Feedback Loop to Inhibit 3T3-L1 Adipogenesis. Int J Mol Sci 2021; 22:ijms22168472. [PMID: 34445177 PMCID: PMC8395197 DOI: 10.3390/ijms22168472] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/30/2021] [Accepted: 08/01/2021] [Indexed: 12/22/2022] Open
Abstract
Adipose tissues (AT) expand in response to energy surplus through adipocyte hypertrophy and hyperplasia. The latter, also known as adipogenesis, is a process by which multipotent precursors differentiate to form mature adipocytes. This process is directed by developmental cues that include members of the TGF-β family. Our goal here was to elucidate, using the 3T3-L1 adipogenesis model, how TGF-β family growth factors and inhibitors regulate adipocyte development. We show that ligands of the Activin and TGF-β families, several ligand traps, and the SMAD1/5/8 signaling inhibitor LDN-193189 profoundly suppressed 3T3-L1 adipogenesis. Strikingly, anti-adipogenic traps and ligands engaged the same mechanism of action involving the simultaneous activation of SMAD2/3 and inhibition of SMAD1/5/8 signaling. This effect was rescued by the SMAD2/3 signaling inhibitor SB-431542. By contrast, although LDN-193189 also suppressed SMAD1/5/8 signaling and adipogenesis, its effect could not be rescued by SB-431542. Collectively, these findings reveal the fundamental role of SMAD1/5/8 for 3T3-L1 adipogenesis, and potentially identify a negative feedback loop that links SMAD2/3 activation with SMAD1/5/8 inhibition in adipogenic precursors.
Collapse
|
38
|
Srivastava RK, Lee ES, Sim E, Sheng NC, Ibáñez CF. Sustained anti-obesity effects of life-style change and anti-inflammatory interventions after conditional inactivation of the activin receptor ALK7. FASEB J 2021; 35:e21759. [PMID: 34245608 DOI: 10.1096/fj.202002785rr] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 06/08/2021] [Accepted: 06/11/2021] [Indexed: 01/11/2023]
Abstract
Life-style change and anti-inflammatory interventions have only transient effects in obesity. It is not clear how benefits obtained by these treatments can be maintained longer term, especially during sustained high caloric intake. Constitutive ablation of the activin receptor ALK7 in adipose tissue enhances catecholamine signaling and lipolysis in adipocytes, and protects mice from diet-induced obesity. Here, we investigated the consequences of conditional ALK7 ablation in adipocytes of adult mice with pre-existing obesity. Although ALK7 deletion had little effect on its own, it synergized strongly with a transient switch to low-fat diet (life-style change) or anti-inflammatory treatment (Na-salicylate), resulting in enhanced lipolysis, increased energy expenditure, and reduced adipose tissue mass and body weight gain, even under sustained high caloric intake. By themselves, diet-switch and salicylate had only a temporary effect on weight gain. Mechanistically, combination of ALK7 ablation with either treatment strongly enhanced the levels of β3-AR, the main adrenergic receptor for catecholamine stimulation of lipolysis, and C/EBPα, an upstream regulator of β3-AR expression. These results suggest that inhibition of ALK7 can be combined with simple interventions to produce longer-lasting benefits in obesity.
Collapse
Affiliation(s)
- Raj Kamal Srivastava
- Department of Physiology and Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Ee-Soo Lee
- Department of Physiology and Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Eunice Sim
- Department of Physiology and Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - New Chih Sheng
- Department of Physiology and Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Carlos F Ibáñez
- Department of Physiology and Life Sciences Institute, National University of Singapore, Singapore, Singapore.,Peking-Tsinghua Center for Life Sciences, Peking University School of Life Sciences, Beijing, China.,PKU-IDG/McGovern Institute for Brain Research, Peking University School of Life Sciences, Beijing, China.,Chinese Institute for Brain Research, Beijing, China.,Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
39
|
Matsuzawa T, Morita M, Shimane A, Otsuka R, Mei Y, Irie F, Yamaguchi Y, Yanai K, Yoshikawa T. Heparan sulfate promotes differentiation of white adipocytes to maintain insulin sensitivity and glucose homeostasis. J Biol Chem 2021; 297:101006. [PMID: 34310946 PMCID: PMC8379462 DOI: 10.1016/j.jbc.2021.101006] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 07/16/2021] [Accepted: 07/22/2021] [Indexed: 11/29/2022] Open
Abstract
Heparan sulfate (HS), a highly sulfated linear polysaccharide, is involved in diverse biological functions in various tissues. Although previous studies have suggested a possible contribution of HS to the differentiation of white adipocytes, there has been no direct evidence supporting this. Here, we inhibited the synthesis of HS chains in 3T3-L1 cells using CRISPR–Cas9 technology, resulting in impaired differentiation of adipocytes with attenuated bone morphogenetic protein 4 (BMP4)–fibroblast growth factor 1 (FGF1) signaling pathways. HS reduction resulted in reduced glucose uptake and decreased insulin-dependent intracellular signaling. We then made heterozygous mutant mice for the Ext1 gene, which encodes an enzyme essential for the HS biosynthesis, specifically in the visceral white adipose tissue (Fabp4-Cre+::Ext1flox/WT mice, hereafter called Ext1Δ/WT) to confirm the importance of HS in vivo. The expression levels of transcription factors that control adipocyte differentiation, such as peroxisome proliferator–activated receptor gamma, were reduced in Ext1Δ/WT adipocytes, which contained smaller, unilocular lipid droplets, reduced levels of enzymes involved in lipid synthesis, and altered expression of BMP4–FGF1 signaling molecules. Furthermore, we examined the impact of HS reduction in visceral white adipose tissue on systemic glucose homeostasis. We observed that Ext1Δ/WT mice showed glucose intolerance because of insulin resistance. Our results demonstrate that HS plays a crucial role in the differentiation of white adipocytes through BMP4–FGF1 signaling pathways, thereby contributing to insulin sensitivity and glucose homeostasis.
Collapse
Affiliation(s)
- Takuro Matsuzawa
- Department of Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masanobu Morita
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Ai Shimane
- Department of Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Rina Otsuka
- Department of Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yu Mei
- Department of Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Fumitoshi Irie
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Yu Yamaguchi
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Kazuhiko Yanai
- Department of Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takeo Yoshikawa
- Department of Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan.
| |
Collapse
|
40
|
Cancer-Associated Adipocytes in Breast Cancer: Causes and Consequences. Int J Mol Sci 2021; 22:ijms22073775. [PMID: 33917351 PMCID: PMC8038661 DOI: 10.3390/ijms22073775] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/01/2021] [Accepted: 04/02/2021] [Indexed: 02/07/2023] Open
Abstract
Breast cancer progression is highly dependent on the heterotypic interaction between tumor cells and stromal cells of the tumor microenvironment. Cancer-associated adipocytes (CAAs) are emerging as breast cancer cell partners favoring proliferation, invasion, and metastasis. This article discussed the intersection between extracellular signals and the transcriptional cascade that regulates adipocyte differentiation in order to appreciate the molecular pathways that have been described to drive adipocyte dedifferentiation. Moreover, recent studies on the mechanisms through which CAAs affect the progression of breast cancer were reviewed, including adipokine regulation, metabolic reprogramming, extracellular matrix remodeling, and immune cell modulation. An in-depth understanding of the complex vicious cycle between CAAs and breast cancer cells is crucial for designing novel strategies for new therapeutic interventions.
Collapse
|
41
|
The effect of TGFβ1 on thermogenic markers is dependent on the degree of adipocyte differentiation. Biosci Rep 2021; 40:223097. [PMID: 32352511 PMCID: PMC7225410 DOI: 10.1042/bsr20194262] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 04/15/2020] [Accepted: 04/27/2020] [Indexed: 12/02/2022] Open
Abstract
Transforming growth factor β (TGFβ) a multifunctional cytokine is known to regulate cell proliferation, differentiation, migration and survival. Although there is variable expression of modulators of TGFβ action during differentiation, a differential effect on fat cell metabolism at the different stages of adipocyte differentiation was unclear. In the present study, 3T3L1 cells were used as an in vitro model to study the effect of TGFβ on adipogenic and thermogenic markers at various stages of preadipocyte to mature adipocyte differentiation. As in our earlier studies on the effect of TGFβ on CEBP’s, we used a standard differentiation mix, and one with the addition of rosiglitazone. RhTGFβ1 was added to undifferentiated adipocytes (preadipocytes) and to adipocytes at day 0 (commitment stage) as well as day 10 (terminal differentiation). Cellular responses in terms of Pref1, PPARγ, TLE3, PGC1α, PRDM16, UCP1 and UCP2 mRNA levels and selected protein products, were determined. Increases in PPARγ, PRDM16, UCP1 and UCP2 mRNA and decreases in Pref1 are good indicators of successful differentiation. The early addition of rhTGFβ1 during commitment stage decreased PPARγ, PRDM16, TLE3, UCP1 and UCP2 mRNA and decreased PRDM16 protein consistent with our earlier report on the inhibition of CEBP’s by TGFβ and CCN2. The addition of rhTGFβ1 to mature adipocyte at day 10 increased UCP1 mRNA and increased PRDM16 and UCP1 proteins. In the present study, our results suggest that TGFβ1 added late enhances the thermogenic potential of mature cells and causes 3T3L1 cells to differentiate to resemble brown or beige rather than white adipose tissue.
Collapse
|
42
|
Sowa Y, Kishida T, Louis F, Sawai S, Seki M, Numajiri T, Takahashi K, Mazda O. Direct Conversion of Human Fibroblasts into Adipocytes Using a Novel Small Molecular Compound: Implications for Regenerative Therapy for Adipose Tissue Defects. Cells 2021; 10:cells10030605. [PMID: 33803331 PMCID: PMC8000077 DOI: 10.3390/cells10030605] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 03/02/2021] [Accepted: 03/08/2021] [Indexed: 02/07/2023] Open
Abstract
There is a need in plastic surgery to prepare autologous adipocytes that can be transplanted in patients to reconstruct soft tissue defects caused by tumor resection, including breast cancer, and by trauma and other diseases. Direct conversion of somatic cells into adipocytes may allow sufficient functional adipocytes to be obtained for use in regeneration therapy. Chemical libraries of 10,800 molecules were screened for the ability to induce lipid accumulation in human dermal fibroblasts (HDFs) in culture. Chemical compound-mediated directly converted adipocytes (CCCAs) were characterized by lipid staining, immunostaining, and qRT-PCR, and were also tested for adipokine secretion and glucose uptake. CCCAs were also implanted into mice to examine their distribution in vivo. STK287794 was identified as a small molecule that induced the accumulation of lipid droplets in HDFs. CCCAs expressed adipocyte-related genes, secreted adiponectin and leptin, and abundantly incorporated glucose. After implantation in mice, CCCAs resided in granulation tissue and remained adipose-like. HDFs were successfully converted into adipocytes by adding a single chemical compound, STK287794. C/EBPα and PPARγ were upregulated in STK287794-treated cells, which strongly suggests involvement of these adipocyte-related transcription factors in the chemical direct conversion. Our method may be useful for the preparation of autogenous adipocytes for transplantation therapy for soft tissue defects and fat tissue atrophy.
Collapse
Affiliation(s)
- Yoshihiro Sowa
- Departments of Plastic and Reconstructive Surgery, Graduate School of Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan;
- Correspondence: ; Tel.: +81-75-251-5730; Fax: +81-75-251-5732
| | - Tsunao Kishida
- Immunology, Graduate School of Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (T.K.); (O.M.)
| | - Fiona Louis
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Suita, Osaka 565-0871, Japan;
| | - Seiji Sawai
- Orthopaedics Graduate School of Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (S.S.); (K.T.)
| | - Makoto Seki
- CellAxia Inc, Nihonbashi, Tokyo 103-0012, Japan;
| | - Toshiaki Numajiri
- Departments of Plastic and Reconstructive Surgery, Graduate School of Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan;
| | - Kenji Takahashi
- Orthopaedics Graduate School of Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (S.S.); (K.T.)
| | - Osam Mazda
- Immunology, Graduate School of Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (T.K.); (O.M.)
| |
Collapse
|
43
|
Lamarche É, AlSudais H, Rajgara R, Fu D, Omaiche S, Wiper-Bergeron N. SMAD2 promotes myogenin expression and terminal myogenic differentiation. Development 2021; 148:dev.195495. [PMID: 33462116 DOI: 10.1242/dev.195495] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 12/24/2020] [Indexed: 11/20/2022]
Abstract
SMAD2 is a transcription factor, the activity of which is regulated by members of the transforming growth factor β (TGFβ) superfamily. Although activation of SMAD2 and SMAD3 downstream of TGFβ or myostatin signaling is known to inhibit myogenesis, we found that SMAD2 in the absence of TGFβ signaling promotes terminal myogenic differentiation. We found that, during myogenic differentiation, SMAD2 expression is induced. Knockout of SMAD2 expression in primary myoblasts did not affect the efficiency of myogenic differentiation but produced smaller myotubes with reduced expression of the terminal differentiation marker myogenin. Conversely, overexpression of SMAD2 stimulated myogenin expression, and enhanced both differentiation and fusion, and these effects were independent of classical activation by the TGFβ receptor complex. Loss of Smad2 in muscle satellite cells in vivo resulted in decreased muscle fiber caliber and impaired regeneration after acute injury. Taken together, we demonstrate that SMAD2 is an important positive regulator of myogenic differentiation, in part through the regulation of Myog.
Collapse
Affiliation(s)
- Émilie Lamarche
- Graduate Program in Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Rm 3106Q, Ottawa, Ontario K1H 8M5, Canada
| | - Hamood AlSudais
- Graduate Program in Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Rm 3106Q, Ottawa, Ontario K1H 8M5, Canada
| | - Rashida Rajgara
- Graduate Program in Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Rm 3106Q, Ottawa, Ontario K1H 8M5, Canada
| | - Dechen Fu
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Rm 3106Q, Ottawa, Ontario K1H 8M5, Canada
| | - Saadeddine Omaiche
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Rm 3106Q, Ottawa, Ontario K1H 8M5, Canada
| | - Nadine Wiper-Bergeron
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Rm 3106Q, Ottawa, Ontario K1H 8M5, Canada
| |
Collapse
|
44
|
Woo J, Koziol-White C, Panettieri R, Jude J. TGF-β: The missing link in obesity-associated airway diseases? CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2021; 2:100016. [PMID: 34909651 PMCID: PMC8663968 DOI: 10.1016/j.crphar.2021.100016] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 01/15/2021] [Accepted: 01/20/2021] [Indexed: 01/19/2023] Open
Abstract
Obesity is emerging as a global public health epidemic. The co-morbidities associated with obesity significantly contribute to reduced quality of life, mortality, and global healthcare burden. Compared to other asthma comorbidities, obesity prominently engenders susceptibility to inflammatory airway diseases such as asthma and chronic obstructive pulmonary disease (COPD), contributes to greater disease severity and evokes insensitivity to current therapies. Unlike in other metabolic diseases associated with obesity, the mechanistic link between obesity and airway diseases is only poorly defined. Transforming growth factor-β (TGF-β) is a pleiotropic inflammatory cytokine belonging to a family of growth factors with pivotal roles in asthma. In this review, we summarize the role of TGF-β in major obesity-associated co-morbidities to shed light on mechanisms of the diseases. Literature evidence shows that TGF-β mechanistically links many co-morbidities with obesity through its profibrotic, remodeling, and proinflammatory functions. We posit that TGF-β plays a similar mechanistic role in obesity-associated inflammatory airway diseases such as asthma and COPD. Concerning the role of TGF-β on metabolic effects of obesity, we posit that TGF-β has a similar mechanistic role in obesity-associated inflammatory airway diseases in interplay with different comorbidities such as hypertension, metabolic diseases like type 2 diabetes, and cardiomyopathies. Future studies in TGF-β-dependent mechanisms in obesity-associated inflammatory airway diseases will advance our understanding of obesity-induced asthma and help find novel therapeutic targets for prevention and treatment.
Collapse
Affiliation(s)
- Joanna Woo
- Rutgers Institute for Translational Medicine & Science, The State University of New Jersey, 89 French Street, Rutgers, 160 Frelinghuysen Road, Piscataway, NJ08854, United States
- Ernest Mario School of Pharmacy, The State University of New Jersey, 89 French Street, Rutgers, 160 Frelinghuysen Road, Piscataway, NJ08854, United States
| | - Cynthia Koziol-White
- Rutgers Institute for Translational Medicine & Science, The State University of New Jersey, 89 French Street, Rutgers, 160 Frelinghuysen Road, Piscataway, NJ08854, United States
- Robert Wood Johnson Medical School, The State University of New Jersey, 89 French Street, Rutgers, 160 Frelinghuysen Road, Piscataway, NJ08854, United States
| | - Reynold Panettieri
- Rutgers Institute for Translational Medicine & Science, The State University of New Jersey, 89 French Street, Rutgers, 160 Frelinghuysen Road, Piscataway, NJ08854, United States
- Robert Wood Johnson Medical School, The State University of New Jersey, 89 French Street, Rutgers, 160 Frelinghuysen Road, Piscataway, NJ08854, United States
- Ernest Mario School of Pharmacy, The State University of New Jersey, 89 French Street, Rutgers, 160 Frelinghuysen Road, Piscataway, NJ08854, United States
| | - Joseph Jude
- Rutgers Institute for Translational Medicine & Science, The State University of New Jersey, 89 French Street, Rutgers, 160 Frelinghuysen Road, Piscataway, NJ08854, United States
- Robert Wood Johnson Medical School, The State University of New Jersey, 89 French Street, Rutgers, 160 Frelinghuysen Road, Piscataway, NJ08854, United States
- Ernest Mario School of Pharmacy, The State University of New Jersey, 89 French Street, Rutgers, 160 Frelinghuysen Road, Piscataway, NJ08854, United States
| |
Collapse
|
45
|
Kandhaya-Pillai R, Hou D, Zhang J, Yang X, Compoginis G, Mori T, Tchkonia T, Martin GM, Hisama FM, Kirkland JL, Oshima J. SMAD4 mutations and cross-talk between TGF-β/IFNγ signaling accelerate rates of DNA damage and cellular senescence, resulting in a segmental progeroid syndrome-the Myhre syndrome. GeroScience 2021; 43:1481-1496. [PMID: 33428109 DOI: 10.1007/s11357-020-00318-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 12/14/2020] [Indexed: 12/13/2022] Open
Abstract
SMAD4 encodes a member of the SMAD family of proteins involved in the TGF-β signaling pathway. Potentially heritable, autosomal dominant, gain-of-function heterozygous variants of SMAD4 cause a rare developmental disorder, the Myhre syndrome, which is associated with a wide range of developmental and post-developmental phenotypes that we now characterize as a novel segmental progeroid syndrome. Whole-exome sequencing of a patient referred to our International Registry of Werner Syndrome revealed a heterozygous p.Arg496Cys variant of the SMAD4 gene. To investigate the role of SMAD4 mutations in accelerated senescence, we generated cellular models overexpressing either wild-type SMAD4 or mutant SMAD4-R496C in normal skin fibroblasts. We found that cells expressing the SMAD4-R496C mutant exhibited decreased proliferation and elevated expression of cellular senescence and inflammatory markers, including IL-6, IFNγ, and a TGF-β target gene, PAI-1. Here we show that transient exposure to TGF-β, an inflammatory cytokine, followed by chronic IFNγ stimulation, accelerated rates of senescence that were associated with increased DNA damage foci and SMAD4 expression. TGF-β, IFNγ, or combinations of both were not sufficient to reduce proliferation rates of fibroblasts. In contrast, TGF-β alone was able to induce preadipocyte senescence via induction of the mTOR protein. The mTOR inhibitor rapamycin mitigated TGF-β-induced expression of p21, p16, and DNA damage foci and improved replicative potential of preadipocytes, supporting the cell-specific response to this cytokine. These findings collectively suggest that persistent DNA damage and cross-talk between TGF-β/IFNγ pathways contribute to a series of molecular events leading to cellular senescence and a segmental progeroid syndrome.
Collapse
Affiliation(s)
- Renuka Kandhaya-Pillai
- Department of Pathology, University of Washington, Box 357470, HSB, Seattle, WA, K-543, USA
| | - Deyin Hou
- Department of Pathology, University of Washington, Box 357470, HSB, Seattle, WA, K-543, USA
| | - Jiaming Zhang
- Department of Pathology, University of Washington, Box 357470, HSB, Seattle, WA, K-543, USA
| | - Xiaomeng Yang
- Department of Pathology, University of Washington, Box 357470, HSB, Seattle, WA, K-543, USA
| | - Goli Compoginis
- Department of Dermatology, University of Southern California, Los Angeles, CA, USA
| | - Takayasu Mori
- Department of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tamara Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - George M Martin
- Department of Pathology, University of Washington, Box 357470, HSB, Seattle, WA, K-543, USA
| | - Fuki M Hisama
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA, USA
| | - James L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Junko Oshima
- Department of Pathology, University of Washington, Box 357470, HSB, Seattle, WA, K-543, USA.
| |
Collapse
|
46
|
Rodrigues LM, Schoonmaker JP, Resende FD, Siqueira GR, Rodrigues Machado Neto O, Gionbelli MP, Ramalho Santos Gionbelli T, Ladeira MM. Effects of protein supplementation on Nellore cows’ reproductive performance, growth, myogenesis, lipogenesis and intestine development of the progeny. ANIMAL PRODUCTION SCIENCE 2021. [DOI: 10.1071/an20498] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Context
It is hypothesised that protein supplementation in pregnant Nellore cows during the dry season would improve reproductive performance in the next breeding season, as well as growth, myogenesis and intramuscular lipogenesis of the progeny until weaning.
Aims
To evaluate the effect of maternal nutrition on cow reproductive performance, as well as on the growth, myogenesis and lipogenesis of the progeny until weaning.
Methods
A total of 92 pregnant cows were fed on pasture, and half of the cows were also fed a mineral–protein supplement (36% crude protein) from 124 ± 21 days of pregnancy to calving. Therefore, two treatments were tested: non-supplemented or supplemented cows. Progeny were weighed after birth, 130 days after birth and at weaning. Six newborn calves from each treatment were slaughtered to collect muscle and jejunum samples to analyse histology and gene expression. In addition, Longissimus thoracis muscle biopsies were collected at 11 days after birth and weaning for gene expression analyses.
Key results
Supplemented cows had greater bodyweight (P = 0.03) and body condition score (P = 0.05) during gestation, and the pregnancy rate in the subsequent breeding season had a tendency (P = 0.10) to be greater. The progeny from supplemented cows had greater bodyweight at birth (P = 0.05). However, no differences (P > 0.05) were found in bodyweight at weaning or in the average daily gain during this period. Non-supplemented calves had greater SLC27A4 (P = 0.04) expression and a tendency for greater expression of SLC5A1 (P = 0.08) in the jejunum. Muscle gene expression data showed that progeny from supplemented cows had greater expression of myogenic (WNT10B), adipogenic (PPARG, ZFP423, CD36) and fibrogenic (TGFβ1) markers at birth and at weaning (P ≤ 0.10).
Conclusions
In conclusion, protein supplementation of pregnant Nellore cows leads to positive effects for subsequent reproductive performance and for muscle development of their progeny. In addition, the progeny from feed-restricted cows increases prenatal intestinal development for better nutrients absorption under a potentially impaired environmental condition.
Implications
The use of protein supplementation in pregnant Nellore cows has a positive impact on the production system, increasing productivity in a cow/calf operation.
Collapse
|
47
|
Dankel SN, Grytten E, Bjune JI, Nielsen HJ, Dietrich A, Blüher M, Sagen JV, Mellgren G. COL6A3 expression in adipose tissue cells is associated with levels of the homeobox transcription factor PRRX1. Sci Rep 2020; 10:20164. [PMID: 33214660 PMCID: PMC7678848 DOI: 10.1038/s41598-020-77406-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 10/21/2020] [Indexed: 01/28/2023] Open
Abstract
Fibrillar collagen COL6α3 in adipose tissue has been associated with obesity, inflammation, insulin resistance and cancer. We here aimed to identify novel transcriptional regulators of COL6A3 expression. Based on a transcriptome dataset of adipose tissue, we identified strong correlations for 56 genes with COL6A3 mRNA, including targets of TGF-β/SMAD signaling. Among the identified candidates, the homeobox transcription factor PRRX1 showed a particularly striking co-expression with COL6A3, validated across several different cohorts, including patients with extreme obesity, insulin sensitive and resistant obesity (subcutaneous and omental), after profound fat loss (subcutaneous), and lean controls (subcutaneous). In human and mouse adipose cells, PRRX1 knockdown reduced COL6A3 mRNA and PRRX1 overexpression transactivated a reporter construct with the endogenous human COL6A3 promoter. Stable PRRX1 overexpression in 3T3-L1 cells induced Col6a3 mRNA threefold specifically after adipogenic induction, whereas TGF-β1 treatment upregulated Col6a3 mRNA also in the preadipocyte state. Interestingly, pro-inflammatory stimulus (i.e., TNF-α treatment) decreased PRRX1-mediated Col6a3 transactivation and mRNA expression, supporting a role for this mechanism in the regulation of adipose tissue inflammation. In conclusion, we identified the homeobox factor PRRX1 as a novel transcriptional regulator associated with COL6A3 expression, providing new insight into the regulatory mechanisms of altered adipose tissue function in obesity and insulin resistance.
Collapse
Affiliation(s)
- Simon N Dankel
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway. .,Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway.
| | - Elise Grytten
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway.,Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
| | - Jan-Inge Bjune
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway.,Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
| | | | - Arne Dietrich
- Department of Surgery, University of Leipzig, Leipzig, Germany
| | - Matthias Blüher
- Department of Medicine, University of Leipzig, Leipzig, Germany
| | - Jørn V Sagen
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway.,Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
| | - Gunnar Mellgren
- Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway. .,Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway.
| |
Collapse
|
48
|
Gharanei S, Shabir K, Brown JE, Weickert MO, Barber TM, Kyrou I, Randeva HS. Regulatory microRNAs in Brown, Brite and White Adipose Tissue. Cells 2020; 9:cells9112489. [PMID: 33207733 PMCID: PMC7696849 DOI: 10.3390/cells9112489] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/02/2020] [Accepted: 11/13/2020] [Indexed: 02/07/2023] Open
Abstract
MicroRNAs (miRNAs) constitute a class of short noncoding RNAs which regulate gene expression by targeting messenger RNA, inducing translational repression and messenger RNA degradation. This regulation of gene expression by miRNAs in adipose tissue (AT) can impact on the regulation of metabolism and energy homeostasis, particularly considering the different types of adipocytes which exist in mammals, i.e., white adipocytes (white AT; WAT), brown adipocytes (brown AT; BAT), and inducible brown adipocytes in WAT (beige or brite or brown-in-white adipocytes). Indeed, an increasing number of miRNAs has been identified to regulate key signaling pathways of adipogenesis in BAT, brite AT, and WAT by acting on transcription factors that promote or inhibit adipocyte differentiation. For example, MiR-328, MiR-378, MiR-30b/c, MiR-455, MiR-32, and MiR-193b-365 activate brown adipogenesis, whereas MiR-34a, MiR-133, MiR-155, and MiR-27b are brown adipogenesis inhibitors. Given that WAT mainly stores energy as lipids, whilst BAT mainly dissipates energy as heat, clarifying the effects of miRNAs in different types of AT has recently attracted significant research interest, aiming to also develop novel miRNA-based therapies against obesity, diabetes, and other obesity-related diseases. Therefore, this review presents an up-to-date comprehensive overview of the role of key regulatory miRNAs in BAT, brite AT, and WAT.
Collapse
Affiliation(s)
- Seley Gharanei
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK; (S.G.); (M.O.W.); (T.M.B.); (I.K.)
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
| | - Kiran Shabir
- Aston Medical Research Institute, Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham B4 7ET, UK; (K.S.); (J.E.B.)
| | - James E. Brown
- Aston Medical Research Institute, Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham B4 7ET, UK; (K.S.); (J.E.B.)
- School of Biosciences, College of Health and Life Sciences, Aston University, Birmingham B4 7ET, UK
| | - Martin O. Weickert
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK; (S.G.); (M.O.W.); (T.M.B.); (I.K.)
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
- Centre of Applied Biological & Exercise Sciences, Faculty of Health & Life Sciences, Coventry University, Coventry CV1 5FB, UK
| | - Thomas M. Barber
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK; (S.G.); (M.O.W.); (T.M.B.); (I.K.)
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
| | - Ioannis Kyrou
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK; (S.G.); (M.O.W.); (T.M.B.); (I.K.)
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
- Aston Medical Research Institute, Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham B4 7ET, UK; (K.S.); (J.E.B.)
| | - Harpal S. Randeva
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK; (S.G.); (M.O.W.); (T.M.B.); (I.K.)
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
- Aston Medical Research Institute, Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham B4 7ET, UK; (K.S.); (J.E.B.)
- Correspondence:
| |
Collapse
|
49
|
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: 21] [Impact Index Per Article: 4.2] [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
|
50
|
Zhang Z, Zhang Z, Oyelami FO, Sun H, Xu Z, Ma P, Wang Q, Pan Y. Identification of genes related to intramuscular fat independent of backfat thickness in Duroc pigs using single-step genome-wide association. Anim Genet 2020; 52:108-113. [PMID: 33073401 DOI: 10.1111/age.13012] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/29/2020] [Accepted: 09/19/2020] [Indexed: 12/11/2022]
Abstract
Intramuscular fat (IMF) is an important meat-quality trait of pigs, which influences pork's shearing force, hydraulics, tenderness and juicy flavor. However, to achieve a higher percentage of lean meat, pigs with lower backfat thickness (BF) are intensively selected for, which may lead to a reduction in pork quality. Therefore, the objective of this study was to locate loci that affect IMF without changing BF. A single-step GWAS was performed on 950 Duroc pigs genotyped by a 50K SNP chip in order to detect genomic variants relevant to IMF and BF. The significant SNPs detected were afterwards divided into a BF subset (seven SNPs), an IMF subset (11 SNPs) and a subset of both traits (12 SNPs), according to their P-value and LD. After SNP and QTL annotation, our results indicated that SSC1: 167938652, 166363826, 164829874 and 167171587 might be associated with IMF without changing BF. In the subset of both traits, we found that the combined effect of ALGA0006602 (SSC1: 159538854) and 12784636 (SSC1: 160773437) might improve the IMF without changing BF. Our gene annotation result showed that TLE3, ITGA11, SMAD6, PAQR5 and [RNF152A/G × MC4RA/A ] genes might affect IMF independently of BF. We believe that the SNPs and genes identified in this study will be valuable for the future molecular breeding of IMF in Duroc pigs.
Collapse
Affiliation(s)
- Z Zhang
- Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, 800# Dongchuan Road, Shanghai, East, 200240, China
| | - Z Zhang
- Department of Animal Science, College of Animal Science, Zhejiang University, 866# Yuhangtang Road, Hangzhou, East, 310058, China
| | - F O Oyelami
- Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, 800# Dongchuan Road, Shanghai, East, 200240, China
| | - H Sun
- Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, 800# Dongchuan Road, Shanghai, East, 200240, China
| | - Z Xu
- Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, 800# Dongchuan Road, Shanghai, East, 200240, China
| | - P Ma
- Department of Animal Science, School of Agriculture and Biology, Shanghai Jiao Tong University, 800# Dongchuan Road, Shanghai, East, 200240, China
| | - Q Wang
- Department of Animal Science, College of Animal Science, Zhejiang University, 866# Yuhangtang Road, Hangzhou, East, 310058, China
| | - Y Pan
- Department of Animal Science, College of Animal Science, Zhejiang University, 866# Yuhangtang Road, Hangzhou, East, 310058, China
| |
Collapse
|