1
|
De Francesco F, Sbarbati A, Sierra LAQ, Zingaretti N, Sarmadian Z, Parodi PC, Ricci G, Riccio M, Mobasheri A. Anatomy, Histology, and Embryonic Origin of Adipose Tissue: Insights to Understand Adipose Tissue Homofunctionality in Regeneration and Therapies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024. [PMID: 39107527 DOI: 10.1007/5584_2024_801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
Preadipocytes are formed during the 14th and 16th weeks of gestation. White adipose tissue, in particular, is generated in specific areas and thereby assembles after birth, rapidly increasing following the propagation of adipoblasts, which are considered the preadipocyte cell precursors. The second trimester of gestation is a fundamental phase of adipogenesis, and in the third trimester, adipocytes, albeit small may be present within the main deposition areas. In the course of late gestation, adipose tissue develops in the foetus and promotes the synthesis of large amounts of uncoupling protein 1, in similar quantities relative to differentiated brown adipose tissue. In mammals, differentiation occurs in two functionally different types of adipose cells: white adipose cells resulting from lipid storage and brown adipose cells from increased metabolic energy consumption. During skeletogenesis, synovial joints develop through the condensation of mesenchymal cells, which forms an insertional layer of flattened cells that umlaut skeletal elements, by sharing the same origin in the development of synovium. Peri-articular fat pads possess structural similarity with body subcutaneous white adipose tissue; however, they exhibit a distinct metabolic function due to the micro-environmental cues in which they are embedded. Fat pads are an important component of the synovial joint and play a key role in the maintenance of joint homeostasis. They are also implicated in pathological states such as osteoarthritis.In this paper we explore the therapeutic potential of adipocyte tissue mesenchymal precursor-based stem cell therapy linking it back to the anatomic origin of adipose tissue.
Collapse
Affiliation(s)
- Francesco De Francesco
- Department of Reconstructive Surgery and Hand Surgery, AOU Ospedali Riuniti delle Marche, Ancona, Italy
| | - Andrea Sbarbati
- Department of Neuroscience, Biomedicine and Movement, Human Anatomy and Histology Section, University of Verona, Verona, Italy
| | | | - Nicola Zingaretti
- Department of Medical Area (DAME), Clinic of Plastic and Reconstructive Surgery, Academic Hospital of Udine, University of Udine, Udine, Italy
| | - Zahra Sarmadian
- Research Unit of Health Sciences and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Pier Camillo Parodi
- Department of Medical Area (DAME), Clinic of Plastic and Reconstructive Surgery, Academic Hospital of Udine, University of Udine, Udine, Italy
| | - Giulia Ricci
- Department of Experimental Medicine, Università Degli Studi Della Campania "Luigi Vanvitelli", Naples, Italy
| | - Michele Riccio
- Department of Reconstructive Surgery and Hand Surgery, AOU Ospedali Riuniti delle Marche, Ancona, Italy
| | - Ali Mobasheri
- Research Unit of Health Sciences and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland.
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania.
- Department of Joint Surgery, Sun Yat-sen University, Guangzhou, People's Republic of China.
- World Health Organization Collaborating Center for Public Health Aspects of Musculoskeletal Health and Aging, Université de Liège, Liège, Belgium.
| |
Collapse
|
2
|
Kaimala S, Lootah SS, Mehra N, Kumar CA, Marzooqi SA, Sampath P, Ansari SA, Emerald BS. The Long Non-Coding RNA Obesity-Related (Obr) Contributes To Lipid Metabolism Through Epigenetic Regulation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401939. [PMID: 38704700 PMCID: PMC11234455 DOI: 10.1002/advs.202401939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Indexed: 05/07/2024]
Abstract
Obesity is a multifactorial disease that is part of today's epidemic and also increases the risk of other metabolic diseases. Long noncoding RNAs (lncRNAs) provide one tier of regulatory mechanisms to maintain metabolic homeostasis. Although lncRNAs are a significant constituent of the mammalian genome, studies aimed at their metabolic significance, including obesity, are only beginning to be addressed. Here, a developmentally regulated lncRNA, termed as obesity related (Obr), whose expression in metabolically relevant tissues such as skeletal muscle, liver, and pancreas is altered in diet-induced obesity, is identified. The Clone 9 cell line and high-fat diet-induced obese Wistar rats are used as a model system to verify the function of Obr. By using stable expression and antisense oligonucleotide-mediated downregulation of the expression of Obr followed by different molecular biology experiments, its role in lipid metabolism is verified. It is shown that Obr associates with the cAMP response element-binding protein (Creb) and activates different transcription factors involved in lipid metabolism. Its association with the Creb histone acetyltransferase complex, which includes the cAMP response element-binding protein (CBP) and p300, positively regulates the transcription of genes involved in lipid metabolism. In addition, Obr is regulated by Pparγ in response to lipid accumulation.
Collapse
Affiliation(s)
- Suneesh Kaimala
- Department of Anatomy, College of Medicine and Health Sciences, UAE University, Al Ain, P.O. Box 15551, UAE
| | - Shareena Saeed Lootah
- Department of Anatomy, College of Medicine and Health Sciences, UAE University, Al Ain, P.O. Box 15551, UAE
| | - Neha Mehra
- Department of Anatomy, College of Medicine and Health Sciences, UAE University, Al Ain, P.O. Box 15551, UAE
| | - Challagandla Anil Kumar
- Department of Anatomy, College of Medicine and Health Sciences, UAE University, Al Ain, P.O. Box 15551, UAE
| | - Saeeda Al Marzooqi
- Department of Pathology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, P.O. Box 15551, UAE
| | - Prabha Sampath
- A*STAR Skin Research Laboratory, Agency for Science Technology & Research (A*STAR), Singapore, 138648, Singapore
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore
- Genome Institute of Singapore, Agency for Science Technology & Research (A*STAR), Singapore, 138672, Singapore
| | - Suraiya Anjum Ansari
- Department of Biochemistry and Molecular Biology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, P.O. Box 15551, UAE
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, P.O. Box 15551, UAE
- ASPIRE Precision Medicine, Research Institute Abu Dhabi, Al Ain, Abu Dhabi, P.O. Box 15551, UAE
| | - Bright Starling Emerald
- Department of Anatomy, College of Medicine and Health Sciences, UAE University, Al Ain, P.O. Box 15551, UAE
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, P.O. Box 15551, UAE
- ASPIRE Precision Medicine, Research Institute Abu Dhabi, Al Ain, Abu Dhabi, P.O. Box 15551, UAE
| |
Collapse
|
3
|
Sekine Y, Kikkawa K, Honda S, Sasaki Y, Kawahara S, Mizushima A, Togi S, Fujimuro M, Oritani K, Matsuda T. STAP-2 facilitates insulin signaling through binding to CAP/c-Cbl and regulates adipocyte differentiation. Sci Rep 2024; 14:5799. [PMID: 38461189 PMCID: PMC10925025 DOI: 10.1038/s41598-024-56533-0] [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: 10/20/2023] [Accepted: 03/07/2024] [Indexed: 03/11/2024] Open
Abstract
Signal-transducing adaptor protein-2 (STAP-2) is an adaptor molecule involved in several cellular signaling cascades. Here, we attempted to identify novel STAP-2 interacting molecules, and identified c-Cbl associated protein (CAP) as a binding protein through the C-terminal proline-rich region of STAP-2. Expression of STAP-2 increased the interaction between CAP and c-Cbl, suggesting that STAP-2 bridges these proteins and enhances complex formation. CAP/c-Cbl complex is known to regulate GLUT4 translocation in insulin signaling. STAP-2 overexpressed human hepatocyte Hep3B cells showed enhanced GLUT4 translocation after insulin treatment. Elevated levels of Stap2 mRNA have been observed in 3T3-L1 cells and mouse embryonic fibroblasts (MEFs) during adipocyte differentiation. The differentiation of 3T3-L1 cells into adipocytes was highly promoted by retroviral overexpression of STAP-2. In contrast, STAP-2 knockout (KO) MEFs exhibited suppressed adipogenesis. The increase in body weight with high-fat diet feeding was significantly decreased in STAP-2 KO mice compared to WT animals. These data suggest that the expression of STAP-2 correlates with adipogenesis. Thus, STAP-2 is a novel regulatory molecule that controls insulin signal transduction by forming a c-Cbl/STAP-2/CAP ternary complex.
Collapse
Affiliation(s)
- Yuichi Sekine
- Department of Cell Biology, Kyoto Pharmaceutical University, Kyoto, 607-8412, Japan.
| | - Kazuna Kikkawa
- Department of Cell Biology, Kyoto Pharmaceutical University, Kyoto, 607-8412, Japan
| | - Sachie Honda
- Department of Cell Biology, Kyoto Pharmaceutical University, Kyoto, 607-8412, Japan
| | - Yuto Sasaki
- Department of Immunology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan
| | - Shoya Kawahara
- Department of Immunology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan
| | - Akihiro Mizushima
- Department of Immunology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan
| | - Sumihito Togi
- Division of Genomic Medicine, Department of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, Kahoku, Ishikawa, 920-0293, Japan
| | - Masahiro Fujimuro
- Department of Cell Biology, Kyoto Pharmaceutical University, Kyoto, 607-8412, Japan
| | - Kenji Oritani
- Department of Hematology, International University of Health and Welfare, Narita, Chiba, 286-8686, Japan
| | - Tadashi Matsuda
- Department of Immunology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan.
| |
Collapse
|
4
|
Carobbio S, Pellegrinelli V, Vidal-Puig A. Adipose Tissue Dysfunction Determines Lipotoxicity and Triggers the Metabolic Syndrome: Current Challenges and Clinical Perspectives. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:231-272. [PMID: 39287854 DOI: 10.1007/978-3-031-63657-8_8] [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
The adipose tissue organ is organised as distinct anatomical depots located all along the body axis, and it is constituted of three different types of adipocytes: white, beige and brown, which are integrated with vascular, immune, neural, and extracellular stroma cells. These distinct adipocytes serve different specialised functions. The main function of white adipocytes is to ensure healthy storage of excess nutrients/energy and its rapid mobilisation to supply the demand of energy imposed by physiological cues in other organs, whereas brown and beige adipocytes are designed for heat production through uncoupling lipid oxidation from energy production. The concerted action of the three types of adipocytes/tissues ensures an optimal metabolic status. However, when one or several of these adipose depots become dysfunctional because of sustained lipid/nutrient overload, then insulin resistance and associated metabolic complications ensue. These metabolic alterations close a vicious cycle that negatively affects the adipose tissue functionality and compromises global metabolic homeostasis. Optimising white adipose tissue expandability and ensuring its functional metabolic flexibility and/or promoting brown/beige mediated thermogenic activity are complementary strategies that counteract obesity and its associated lipotoxic metabolic effects. However, the development of these therapeutic approaches requires a deep understanding of adipose tissue in all broad aspects. In this chapter, we will discuss the characteristics of the different adipose tissue depots with respect to origins and precursors recruitment, plasticity, cellular composition, and expandability capacity potential as well as molecular and metabolic characteristic signatures in both physiological and pathophysiological conditions. Current antilipotoxic strategies for future clinical application are also discussed in this chapter.
Collapse
Affiliation(s)
- Stefania Carobbio
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
- Centro de Investigación Principe Felipe, Valencia, Spain.
| | - Vanessa Pellegrinelli
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Antonio Vidal-Puig
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
- Centro de Investigación Principe Felipe, Valencia, Spain.
| |
Collapse
|
5
|
Ryu SW, Moon JC, Oh BS, Yu SY, Bak JE, Heo ES, Jeong JH, Lee JH. Anti-obesity activity of human gut microbiota Bacteroides stercoris KGMB02265. Arch Microbiol 2023; 206:19. [PMID: 38086977 DOI: 10.1007/s00203-023-03750-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 11/16/2023] [Accepted: 11/16/2023] [Indexed: 12/18/2023]
Abstract
Obesity is a global health threat that causes various complications such as type 2 diabetes and nonalcoholic fatty liver disease. Gut microbiota is closely related to obesity. In particular, a higher Firmicutes to Bacteroidetes ratio has been reported as a biomarker of obesity, suggesting that the phylum Bacteroidetes may play a role in inhibiting obesity. Indeed, the genus Bacteroides was enriched in the healthy subjects based on metagenome analysis. In this study, we determined the effects of Bacteroides stercoris KGMB02265, a species belonging to the phylum Bacteroidetes, on obesity both in vitro and in vivo. The cell-free supernatant of B. stercoris KGMB02265 inhibited lipid accumulation in 3T3-L1 preadipocytes, in which the expression of adipogenic marker genes was repressed. In vivo study showed that the oral administration of B. stercoris KGMB02265 substantially reduced body weight and fat weight in high-fat diet induced obesity in mice. Furthermore, obese mice orally administered with B. stercoris KGMB02265 restored glucose sensitivity and reduced leptin and triglyceride levels. Taken together, our study reveals that B. stercoris KGMB02265 has anti-obesity activity and suggests that it may be a promising candidate for treating obesity.
Collapse
Affiliation(s)
- Seoung Woo Ryu
- Korean Collection for Type Cultures, Biological Resource Center, Institute of Bioscience and Biotechnology, Jeongeup, 56212, Republic of Korea
- BioMedical Sciences Graduate Program, Chonnam National University, Hwasun, 58128, Republic of Korea
| | - Jeong Chan Moon
- National Institute of Ecology, Yeongyang, 36531, Republic of Korea
| | - Byeong Seob Oh
- Korean Collection for Type Cultures, Biological Resource Center, Institute of Bioscience and Biotechnology, Jeongeup, 56212, Republic of Korea
| | - Seung Yeob Yu
- Korean Collection for Type Cultures, Biological Resource Center, Institute of Bioscience and Biotechnology, Jeongeup, 56212, Republic of Korea
- BioMedical Sciences Graduate Program, Chonnam National University, Hwasun, 58128, Republic of Korea
| | - Jeong Eun Bak
- Korean Collection for Type Cultures, Biological Resource Center, Institute of Bioscience and Biotechnology, Jeongeup, 56212, Republic of Korea
- BioMedical Sciences Graduate Program, Chonnam National University, Hwasun, 58128, Republic of Korea
| | - Eun Seo Heo
- Korean Collection for Type Cultures, Biological Resource Center, Institute of Bioscience and Biotechnology, Jeongeup, 56212, Republic of Korea
- University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Jae-Ho Jeong
- BioMedical Sciences Graduate Program, Chonnam National University, Hwasun, 58128, Republic of Korea.
| | - Ju Huck Lee
- Korean Collection for Type Cultures, Biological Resource Center, Institute of Bioscience and Biotechnology, Jeongeup, 56212, Republic of Korea.
- University of Science and Technology (UST), Daejeon, 34113, Republic of Korea.
| |
Collapse
|
6
|
Yue J, Sun C, Tang J, Zhang Q, Lou M, Sun H, Zhang L. Downregulation of miRNA-155-5p contributes to the adipogenic activity of 2-ethylhexyl diphenyl phosphate in 3T3-L1 preadipocytes. Toxicology 2023; 487:153452. [PMID: 36764644 DOI: 10.1016/j.tox.2023.153452] [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: 12/24/2022] [Revised: 02/03/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023]
Abstract
2-Ethylhexyl diphenyl phosphate (EHDPP) is a commonly used organophosphorus flame retardant and food packaging material. Because of its high lipophilic and bioaccumulative properties, adipocytes are the primary target of EHDPP. However, the toxicity of EHDPP on preadipocytes and the potential mechanism have not been fully elucidated. MicroRNAs (miRNAs) are thought to be an important mediator that contribute to the toxicity of environmental contaminants. To identify the miRNAs specifically responsible for EHDPP exposure and their role in EGDPP's toxicity in preadipocytes, the adipogenic effects and miRNA expression profiling were performed on 3T3-L1 preadipocytes exposed to EHDPP. EHDPP at concentrations of 1-10 μM promoted adipocyte differentiation, as evidenced by lipid staining, triglyceride content, and expression of adipogenesis markers. MiRNA-seq analysis revealed that 7 differentially expressed miRNAs were recognized under EHDPP exposure, with miR-155-5p being the top down-regulated miRNA. Quantitative reverse transcription PCR (RT-qPCR) analysis showed that miR-155-5p level fell sharply during the first 2 days and continued to fall dose-dependently throughout the EHDPP exposure period. MiR-155-5p inhibition promotes adipocyte differentiation, whereas its overexpression counteracted EHDPP-induced adipogenesis. Luciferase reporter assay identified CCAAT/enhancer-binding protein beta (C/EBPβ) as a target of miR-155-5p in 3T3-L1 preadipocytes in response to EHDPP. Taken together, EHDPP exposure down-regulated miR-155-5p, which then increased C/EBPβ and peroxisome proliferator-activated receptor γ (PPARγ) expression and promoted adipogenesis in preadipocytes.
Collapse
Affiliation(s)
- Junjie Yue
- Tianjin Key Laboratory of Hazardous Waste Safety Disposal and Recycling Technology, School of Environmental Science and Safety Engineering, Tianjin University of Technology, Tianjin 300384, China
| | - Caiting Sun
- Tianjin Key Laboratory of Hazardous Waste Safety Disposal and Recycling Technology, School of Environmental Science and Safety Engineering, Tianjin University of Technology, Tianjin 300384, China
| | - Jinyuan Tang
- Tianjin Key Laboratory of Hazardous Waste Safety Disposal and Recycling Technology, School of Environmental Science and Safety Engineering, Tianjin University of Technology, Tianjin 300384, China
| | - Qiyuan Zhang
- Tianjin Key Laboratory of Hazardous Waste Safety Disposal and Recycling Technology, School of Environmental Science and Safety Engineering, Tianjin University of Technology, Tianjin 300384, China
| | - Mengjie Lou
- Tianjin Key Laboratory of Hazardous Waste Safety Disposal and Recycling Technology, School of Environmental Science and Safety Engineering, Tianjin University of Technology, Tianjin 300384, China
| | - Hongwen Sun
- Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin 300071, China
| | - Lianying Zhang
- Tianjin Key Laboratory of Hazardous Waste Safety Disposal and Recycling Technology, School of Environmental Science and Safety Engineering, Tianjin University of Technology, Tianjin 300384, China; Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin 300071, China.
| |
Collapse
|
7
|
Nartey MNN, Jisaka M, Syeda PK, Nishimura K, Shimizu H, Yokota K. Prostaglandin D 2 Added during the Differentiation of 3T3-L1 Cells Suppresses Adipogenesis via Dysfunction of D-Prostanoid Receptor P1 and P2. Life (Basel) 2023; 13:life13020370. [PMID: 36836727 PMCID: PMC9963520 DOI: 10.3390/life13020370] [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: 12/11/2022] [Revised: 01/20/2023] [Accepted: 01/27/2023] [Indexed: 01/31/2023] Open
Abstract
We previously reported that the addition of prostaglandin, (PG)D2, and its chemically stable analog, 11-deoxy-11-methylene-PGD2 (11d-11m-PGD2), during the maturation phase of 3T3-L1 cells promotes adipogenesis. In the present study, we aimed to elucidate the effects of the addition of PGD2 or 11d-11m-PGD2 to 3T3-L1 cells during the differentiation phase on adipogenesis. We found that both PGD2 and 11d-11m-PGD2 suppressed adipogenesis through the downregulation of peroxisome proliferator-activated receptor gamma (PPARγ) expression. However, the latter suppressed adipogenesis more potently than PGD2, most likely because of its higher resistance to spontaneous transformation into PGJ2 derivatives. In addition, this anti-adipogenic effect was attenuated by the coexistence of an IP receptor agonist, suggesting that the effect depends on the intensity of the signaling from the IP receptor. The D-prostanoid receptors 1 (DP1) and 2 (DP2, also known as a chemoattractant receptor-homologous molecule expressed on Th2 cells) are receptors for PGD2. The inhibitory effects of PGD2 and 11d-11m-PGD2 on adipogenesis were slightly attenuated by a DP2 agonist. Furthermore, the addition of PGD2 and 11d-11m-PGD2 during the differentiation phase reduced the DP1 and DP2 expression during the maturation phase. Overall, these results indicated that the addition of PGD2 or 11d-11m-PGD2 during the differentiation phase suppresses adipogenesis via the dysfunction of DP1 and DP2. Therefore, unidentified receptor(s) for both molecules may be involved in the suppression of adipogenesis.
Collapse
Affiliation(s)
- Michael N. N. Nartey
- The United Graduate School of Agricultural Sciences, Tottori University, 4-101 Koyama-Minami, Tottori 680-8553, Japan
- Council for Scientific and Industrial Research-Animal Research Institute, Achimota, Accra P.O. Box AH20, Ghana
| | - Mitsuo Jisaka
- The United Graduate School of Agricultural Sciences, Tottori University, 4-101 Koyama-Minami, Tottori 680-8553, Japan
- Department of Life Science and Biotechnology, Shimane University, 1060 Nishikawatsu-Cho, Matsue 690-8504, Japan
- Institute of Agricultural and Life Sciences, Academic Assembly, Shimane University, 1060 Nishikawatsu-Cho, Matsue 690-8504, Japan
- Correspondence:
| | - Pinky Karim Syeda
- Department of Life Science and Biotechnology, Shimane University, 1060 Nishikawatsu-Cho, Matsue 690-8504, Japan
| | - Kohji Nishimura
- The United Graduate School of Agricultural Sciences, Tottori University, 4-101 Koyama-Minami, Tottori 680-8553, Japan
- Department of Life Science and Biotechnology, Shimane University, 1060 Nishikawatsu-Cho, Matsue 690-8504, Japan
- Institute of Agricultural and Life Sciences, Academic Assembly, Shimane University, 1060 Nishikawatsu-Cho, Matsue 690-8504, Japan
- Interdisciplinary Center for Science Research, Shimane University, 1060 Nishikawatsu-Cho, Matsue 690-8504, Japan
| | - Hidehisa Shimizu
- The United Graduate School of Agricultural Sciences, Tottori University, 4-101 Koyama-Minami, Tottori 680-8553, Japan
- Department of Life Science and Biotechnology, Shimane University, 1060 Nishikawatsu-Cho, Matsue 690-8504, Japan
- Institute of Agricultural and Life Sciences, Academic Assembly, Shimane University, 1060 Nishikawatsu-Cho, Matsue 690-8504, Japan
- Interdisciplinary Center for Science Research, Shimane University, 1060 Nishikawatsu-Cho, Matsue 690-8504, Japan
| | - Kazushige Yokota
- The United Graduate School of Agricultural Sciences, Tottori University, 4-101 Koyama-Minami, Tottori 680-8553, Japan
- Department of Life Science and Biotechnology, Shimane University, 1060 Nishikawatsu-Cho, Matsue 690-8504, Japan
- Institute of Agricultural and Life Sciences, Academic Assembly, Shimane University, 1060 Nishikawatsu-Cho, Matsue 690-8504, Japan
| |
Collapse
|
8
|
Verma R, Fu M, Yang G, Wu L, Wang R. Hydrogen Sulfide Promotes Adipocyte Differentiation, Hyperplasia, and Hypertrophy. ENGINEERING 2023; 20:36-48. [DOI: 10.1016/j.eng.2022.09.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
9
|
Modaresi SMS, Wei W, Emily M, DaSilva NA, Slitt AL. Per- and polyfluoroalkyl substances (PFAS) augment adipogenesis and shift the proteome in murine 3T3-L1 adipocytes. Toxicology 2022; 465:153044. [PMID: 34800597 PMCID: PMC8756374 DOI: 10.1016/j.tox.2021.153044] [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: 07/27/2021] [Revised: 10/27/2021] [Accepted: 11/16/2021] [Indexed: 01/17/2023]
Abstract
The Per- and polyfluoroalkyl substances (PFAS) are a wide group of fluorinated compounds, which the health effects of many of them have not been investigated. Perfluorinated sulfonates, such as perfluorooctane sulfonate (PFOS) and perfluorinated carboxylates, such as perfluorooctanoic acid (PFOA) are members of this broad group of PFAS, and previous studies have shown a correlation between the body accumulation of PFOS and PFOA and increased adipogenesis. PFOA and PFOS have been withdrawn from the market and use is limited because of their persistence, toxicity, and bioaccumulative properties. Instead, short chain PFAS have been created to replace PFOA and PFOS, but the health effects of other short chain PFAS are largely unknown. Therefore, herein we aimed to comprehensively determined the potential adipogenesis of ten different PFAS (PFBS, PFHxS, PFOS, PFBA, PFHxA, PFHA, PFOA, PFNA, PFDA, and HFPO-DA) and investigated the differences in protein expression of 3T3-L1 cells upon exposure to each PFAS. 3T3-L1 cells were differentiated with or without each PFAS for 4-days, and cellular lipid was quantified using Nile Red staining. Analysis of the adipocyte proteome was performed to identify the pathways related to adipogenesis and quantify proteins significantly affected by each PFAS. The results showed that in general, every PFAS investigated in our study has the potential to induce the 3T3-L1 differentiation to adipocytes in the presence of rosiglitazone, with the concentrations that range between 0.25 and 25 μM. Proteomics analysis revealed specific markers regarding to adipogenesis upregulated upon exposure to each of the ten PFAS.
Collapse
Affiliation(s)
| | - Wei Wei
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, 02881, USA
| | - Marques Emily
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, 02881, USA
| | - Nicholas A DaSilva
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, 02881, USA
| | - Angela L Slitt
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, 02881, USA.
| |
Collapse
|
10
|
Bruder J, Fromme T. Global Adipose Tissue Remodeling During the First Month of Postnatal Life in Mice. Front Endocrinol (Lausanne) 2022; 13:849877. [PMID: 35250892 PMCID: PMC8892685 DOI: 10.3389/fendo.2022.849877] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 01/21/2022] [Indexed: 12/13/2022] Open
Abstract
During the first month of postnatal life, adipose tissue depots of mice go through a drastic, but transient, remodeling process. Between postnatal days 10 and 20, several white fat depots display a strong and sudden surge in beige adipocyte emergence that reverts until day 30. At the same time, brown fat depots appear to undergo an opposite phenomenon. We comprehensively describe these events, their depot specificity and known environmental and genetic interactions, such as maternal diet, housing temperature and mouse strain. We further discuss potential mechanisms and plausible purposes, including the tempting hypothesis that postnatal transient remodeling creates a lasting adaptive capacity still detectable in adult animals. Finally, we propose postnatal adipose tissue remodeling as a model process to investigate mechanisms of beige adipocyte recruitment advantageous to cold exposure or adrenergic stimulation in its entirely endogenous sequence of events without external manipulation.
Collapse
Affiliation(s)
- Johanna Bruder
- Else Kröner-Fresenius Center for Nutritional Medicine (EKFZ), Technical University of Munich, Freising, Germany
- Chair of Molecular Nutritional Medicine, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Tobias Fromme
- Chair of Molecular Nutritional Medicine, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
- *Correspondence: Tobias Fromme,
| |
Collapse
|
11
|
Kulus M, Sibiak R, Stefańska K, Zdun M, Wieczorkiewicz M, Piotrowska-Kempisty H, Jaśkowski JM, Bukowska D, Ratajczak K, Zabel M, Mozdziak P, Kempisty B. Mesenchymal Stem/Stromal Cells Derived from Human and Animal Perinatal Tissues-Origins, Characteristics, Signaling Pathways, and Clinical Trials. Cells 2021; 10:cells10123278. [PMID: 34943786 PMCID: PMC8699543 DOI: 10.3390/cells10123278] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/13/2021] [Accepted: 11/19/2021] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are currently one of the most extensively researched fields due to their promising opportunity for use in regenerative medicine. There are many sources of MSCs, of which cells of perinatal origin appear to be an invaluable pool. Compared to embryonic stem cells, they are devoid of ethical conflicts because they are derived from tissues surrounding the fetus and can be safely recovered from medical waste after delivery. Additionally, perinatal MSCs exhibit better self-renewal and differentiation properties than those derived from adult tissues. It is important to consider the anatomy of perinatal tissues and the general description of MSCs, including their isolation, differentiation, and characterization of different types of perinatal MSCs from both animals and humans (placenta, umbilical cord, amniotic fluid). Ultimately, signaling pathways are essential to consider regarding the clinical applications of MSCs. It is important to consider the origin of these cells, referring to the anatomical structure of the organs of origin, when describing the general and specific characteristics of the different types of MSCs as well as the pathways involved in differentiation.
Collapse
Affiliation(s)
- Magdalena Kulus
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland; (M.K.); (K.R.)
| | - Rafał Sibiak
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland; (R.S.); (K.S.)
- Division of Reproduction, Department of Obstetrics, Gynecology, and Gynecologic Oncology, Poznan University of Medical Sciences, 60-535 Poznan, Poland
| | - Katarzyna Stefańska
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland; (R.S.); (K.S.)
| | - Maciej Zdun
- Department of Basic and Preclinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland; (M.Z.); (M.W.); (H.P.-K.)
| | - Maria Wieczorkiewicz
- Department of Basic and Preclinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland; (M.Z.); (M.W.); (H.P.-K.)
| | - Hanna Piotrowska-Kempisty
- Department of Basic and Preclinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland; (M.Z.); (M.W.); (H.P.-K.)
- Department of Toxicology, Poznan University of Medical Sciences, 60-631 Poznan, Poland
| | - Jędrzej M. Jaśkowski
- Department of Diagnostics and Clinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland; (J.M.J.); (D.B.)
| | - Dorota Bukowska
- Department of Diagnostics and Clinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland; (J.M.J.); (D.B.)
| | - Kornel Ratajczak
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland; (M.K.); (K.R.)
| | - Maciej Zabel
- Division of Anatomy and Histology, University of Zielona Gora, 65-046 Zielona Gora, Poland;
| | - Paul Mozdziak
- Prestage Department of Poultry Science, North Carolina State University, Raleigh, NC 27695, USA;
| | - Bartosz Kempisty
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland; (M.K.); (K.R.)
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland; (R.S.); (K.S.)
- Prestage Department of Poultry Science, North Carolina State University, Raleigh, NC 27695, USA;
- Department of Anatomy, Poznan University of Medical Sciences, 60-781 Poznan, Poland
- Correspondence:
| |
Collapse
|
12
|
Huang K, Wang Y, Zhu J, Xiong Y, Lin Y. Regulation of fibroblast growth factor 9 on the differentiation of goat intramuscular adipocytes. Anim Sci J 2021; 92:e13627. [PMID: 34477270 DOI: 10.1111/asj.13627] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 05/05/2021] [Accepted: 08/04/2021] [Indexed: 12/26/2022]
Abstract
It has been found that fibroblast growth factor receptor (FGF-FGFR) signaling can regulate the expression of adipocyte differentiation genes. FGF9 is one of the members of FGFs that mainly binds receptors FGFR2 and FGFR3. FGF9 is highly expressed in the adipose tissue of humans and mice, but there are few reports on the role of FGF9 in goat intramuscular adipocyte differentiation. Therefore, this study explored the effect of FGF9 on adipocyte differentiation through cell culture, interference, and overexpression. The expression of receptors FGFR1-FGFR4 in adipocyte differentiation and their effects on differentiation were detected to screen receptor gene of FGF9. Finally, the interaction between FGF9 and the receptor was tested by cotransfection. Our results showed that FGF9 interacts with FGFR2 to inhibit goat intramuscular adipocyte differentiation by regulating peroxisome proliferator-activated receptor gamma (PPARγ) and preadipocyte factor 1 (Pref1), which is a data support for subsequent pathway research.
Collapse
Affiliation(s)
- Kai Huang
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Education Ministry, Southwest Minzu University, Chengdu, China.,Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Sichuan Province, Southwest Minzu University, Chengdu, China
| | - Yong Wang
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Education Ministry, Southwest Minzu University, Chengdu, China.,Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Sichuan Province, Southwest Minzu University, Chengdu, China
| | - Jiangjiang Zhu
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Education Ministry, Southwest Minzu University, Chengdu, China.,Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Sichuan Province, Southwest Minzu University, Chengdu, China
| | - Yan Xiong
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Education Ministry, Southwest Minzu University, Chengdu, China.,Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Sichuan Province, Southwest Minzu University, Chengdu, China
| | - Yaqiu Lin
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Education Ministry, Southwest Minzu University, Chengdu, China.,Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization of Sichuan Province, Southwest Minzu University, Chengdu, China
| |
Collapse
|
13
|
Fiorani M, De Matteis R, Canonico B, Blandino G, Mazzoli A, Montanari M, Guidarelli A, Cantoni O. Temporal correlation of morphological and biochemical changes with the recruitment of different mechanisms of reactive oxygen species formation during human SW872 cell adipogenic differentiation. Biofactors 2021; 47:837-851. [PMID: 34260117 PMCID: PMC8597007 DOI: 10.1002/biof.1769] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 06/29/2021] [Indexed: 12/22/2022]
Abstract
Human SW872 preadipocyte conversion to mature adipocytes is associated with time-dependent changes in differentiation markers' expression and with morphological changes accompanied by the accumulation of lipid droplets (LDs) as well as by increased mitochondriogenesis and mitochondrial membrane potential. Under identical conditions, the formation of reactive oxygen species (ROS) revealed with a general probe was significant at days 3 and 10 of differentiation and bearly detectable at day 6. NADPH oxidase (NOX)-2 activity determined with an immunocytochemical approach followed a very similar pattern. There was no evidence of mitochondrial ROS (mROS), as detected with a selective fluorescence probe, at days 3 and 6, possibly due to the triggering of the Nrf-2 antioxidant response. mROS were instead clearly detected at day 10, concomitantly with the accumulation of very large LDs, oxidation of both cardiolipin and thioredoxin 2, and decreased mitochondrial glutathione. In conclusion, the morphological and biochemical changes of differentiating SW872 cells are accompanied by the discontinuous formation of ROS derived from NOX-2, increasingly implicated in adipogenesis and adipose tissue dysfunction. In addition, mROS formation was significant only in the late phase of differentiation and was associated with mitochondrial dysfunction.
Collapse
Affiliation(s)
- Mara Fiorani
- Department of Biomolecular SciencesUniversity of Urbino Carlo BoUrbinoItaly
| | - Rita De Matteis
- Department of Biomolecular SciencesUniversity of Urbino Carlo BoUrbinoItaly
| | - Barbara Canonico
- Department of Biomolecular SciencesUniversity of Urbino Carlo BoUrbinoItaly
| | - Giulia Blandino
- Department of Biomolecular SciencesUniversity of Urbino Carlo BoUrbinoItaly
| | - Alessandro Mazzoli
- Department of Biomolecular SciencesUniversity of Urbino Carlo BoUrbinoItaly
| | - Mariele Montanari
- Department of Biomolecular SciencesUniversity of Urbino Carlo BoUrbinoItaly
| | - Andrea Guidarelli
- Department of Biomolecular SciencesUniversity of Urbino Carlo BoUrbinoItaly
| | - Orazio Cantoni
- Department of Biomolecular SciencesUniversity of Urbino Carlo BoUrbinoItaly
| |
Collapse
|
14
|
Murugan DD, Balan D, Wong PF. Adipogenesis and therapeutic potentials of antiobesogenic phytochemicals: Insights from preclinical studies. Phytother Res 2021; 35:5936-5960. [PMID: 34219306 DOI: 10.1002/ptr.7205] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 05/21/2021] [Accepted: 06/17/2021] [Indexed: 12/11/2022]
Abstract
Obesity is one of the most serious public health problems in both developed and developing countries in recent years. While lifestyle and diet modifications are the most important management strategies of obesity, these may be insufficient to ensure long-term weight reduction in certain individuals and alternative strategies including pharmacotherapy need to be considered. However, drugs option remains limited due to low efficacy and adverse effects associated with their use. Hence, identification of safe and effective alternative therapeutic agents remains warranted to combat obesity. In recent years, bioactive phytochemicals are considered as valuable sources for the discovery of new pharmacological agents for the treatment of obesity. Adipocyte hypertrophy and hyperplasia increases with obesity and undergo molecular and cellular alterations that can affect systemic metabolism giving rise to metabolic syndrome and comorbidities such as type 2 diabetes and cardiovascular diseases. Many phytochemicals have been reported to target adipocytes by inhibiting adipogenesis, inducing lipolysis, suppressing the differentiation of preadipocytes to mature adipocytes, reducing energy intake, and boosting energy expenditure mainly in vitro and in animal studies. Nevertheless, further high-quality studies are needed to firmly establish the clinical efficacy of these phytochemicals. This review outlines common pathways involved in adipogenesis and phytochemicals targeting effector molecules of these pathways, the challenges faced and the way forward for the development of phytochemicals as antiobesity agents.
Collapse
Affiliation(s)
- Dharmani Devi Murugan
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Dharvind Balan
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Pooi-Fong Wong
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
15
|
Lv B, An T, Wang T, Bao X, Lian J, Wu Y, Hu Y, Zhu J, Zheng C, Hu X, Gao S, Jiang G. Effects of salvianolic acid B on glycometabolism and lipid metabolism in rodents: Meta-analysis. TRADITIONAL MEDICINE AND MODERN MEDICINE 2021. [DOI: 10.1142/s2575900020300040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022] Open
Abstract
Danshen (Salvia miltiorrhiza) is a herb which has been widely used in China. Salvianolic acid B (SalB) is an aqueous bioactive component derived from Danshen. Here, we aimed to estimate the effect of SalB on glycometabolism and lipid metabolism in rats and mice. We searched four databases until November 2020. The outcome measures were fasting blood glucose (FBG), total cholesterol (TC), triglycerides (TG), high-density lipoprotein cholesterol (HDLc), and low-density lipoprotein cholesterol (LDLc). Twenty-four studies involving 547 animals were included. The meta-analysis showed effects of SalB on decreasing the level of FBG, TC, TG, LDLc, and increasing the level of HDLc compared with the control group. In conclusion, the result showed that SalB may regulate the glycometabolism and lipid metabolism in rats or mice, and may be a potential agent for treating metabolic diseases such as diabetes and hyperlipidemia.
Collapse
Affiliation(s)
- Bohan Lv
- Traditional Chinese Medicine School, Beijing University of Chinese Medicine, Beijing, P. R. China
| | - Tian An
- Traditional Chinese Medicine School, Beijing University of Chinese Medicine, Beijing, P. R. China
| | - Tingye Wang
- Traditional Chinese Medicine School, Beijing University of Chinese Medicine, Beijing, P. R. China
| | - Xueli Bao
- The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, P. R. China
| | - Juan Lian
- Traditional Chinese Medicine School, Beijing University of Chinese Medicine, Beijing, P. R. China
| | - Yanxiang Wu
- Traditional Chinese Medicine School, Beijing University of Chinese Medicine, Beijing, P. R. China
| | - Yuanyuan Hu
- Traditional Chinese Medicine School, Beijing University of Chinese Medicine, Beijing, P. R. China
| | - Jiajian Zhu
- Traditional Chinese Medicine School, Beijing University of Chinese Medicine, Beijing, P. R. China
| | - Chunyan Zheng
- Traditional Chinese Medicine School, Beijing University of Chinese Medicine, Beijing, P. R. China
| | - Xuehong Hu
- Traditional Chinese Medicine School, Beijing University of Chinese Medicine, Beijing, P. R. China
| | - Sihua Gao
- Traditional Chinese Medicine School, Beijing University of Chinese Medicine, Beijing, P. R. China
| | - Guangjian Jiang
- Traditional Chinese Medicine School, Beijing University of Chinese Medicine, Beijing, P. R. China
| |
Collapse
|
16
|
Han JH, Jang KW, Myung CS. Garcinia cambogia attenuates adipogenesis by affecting CEBPB and SQSTM1/p62-mediated selective autophagic degradation of KLF3 through RPS6KA1 and STAT3 suppression. Autophagy 2021; 18:518-539. [PMID: 34101546 DOI: 10.1080/15548627.2021.1936356] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The overexpansion of adipose tissues leads to obesity and eventually results in metabolic disorders. Garcinia cambogia (G. cambogia) has been used as an antiobesity supplement. However, the molecular mechanisms underlying the effects of G. cambogia on cellular processes have yet to be fully understood. Here, we discovered that G. cambogia attenuated the expression of CEBPB (CCAAT/enhancer binding protein (C/EBP), beta), an important adipogenic factor, suppressing its transcription in differentiated cells. In addition, G. cambogia inhibited macroautophagic/autophagic flux by decreasing autophagy-related gene expression and autophagosome formation. Notably, G. cambogia markedly elevated the expression of KLF3 (Kruppel-like factor 3 (basic)), a negative regulator of adipogenesis, by reducing SQSTM1/p62-mediated selective autophagic degradation. Furthermore, increased KLF3 induced by G. cambogia interacted with CTBP2 (C-terminal binding protein 2) to form a transcriptional repressor complex and inhibited Cebpa and Pparg transcription. Importantly, we found that RPS6KA1 and STAT3 were involved in the G. cambogia-mediated regulation of CEBPB and autophagic flux. In an obese animal model, G. cambogia reduced high-fat diet (HFD)-induced obesity by suppressing epididymal and inguinal subcutaneous white adipose tissue mass and adipocyte size, which were attributed to the regulation of targets that had been consistently identified in vitro. These findings provide new insight into the mechanism of G. cambogia-mediated regulation of adipogenesis and suggest molecular links to therapeutic targets for the treatment of obesity.
Collapse
Affiliation(s)
- Joo-Hui Han
- Department of Pharmacology, College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
| | - Keun-Woo Jang
- Department of Pharmacology, College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
| | - Chang-Seon Myung
- Department of Pharmacology, College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
17
|
Brandão BB, Poojari A, Rabiee A. Thermogenic Fat: Development, Physiological Function, and Therapeutic Potential. Int J Mol Sci 2021; 22:5906. [PMID: 34072788 PMCID: PMC8198523 DOI: 10.3390/ijms22115906] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 04/30/2021] [Accepted: 05/27/2021] [Indexed: 12/11/2022] Open
Abstract
The concerning worldwide increase of obesity and chronic metabolic diseases, such as T2D, dyslipidemia, and cardiovascular disease, motivates further investigations into preventive and alternative therapeutic approaches. Over the past decade, there has been growing evidence that the formation and activation of thermogenic adipocytes (brown and beige) may serve as therapy to treat obesity and its associated diseases owing to its capacity to increase energy expenditure and to modulate circulating lipids and glucose levels. Thus, understanding the molecular mechanism of brown and beige adipocytes formation and activation will facilitate the development of strategies to combat metabolic disorders. Here, we provide a comprehensive overview of pathways and players involved in the development of brown and beige fat, as well as the role of thermogenic adipocytes in energy homeostasis and metabolism. Furthermore, we discuss the alterations in brown and beige adipose tissue function during obesity and explore the therapeutic potential of thermogenic activation to treat metabolic syndrome.
Collapse
Affiliation(s)
- Bruna B. Brandão
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA;
| | - Ankita Poojari
- Department of Physiology & Pharmacology, Thomas J. Long School of Pharmacy & Health Sciences, University of the Pacific, Stockton, CA 95211, USA;
| | - Atefeh Rabiee
- Department of Physiology & Pharmacology, Thomas J. Long School of Pharmacy & Health Sciences, University of the Pacific, Stockton, CA 95211, USA;
| |
Collapse
|
18
|
Partial Deficiency of Zfp217 Resists High-Fat Diet-Induced Obesity by Increasing Energy Metabolism in Mice. Int J Mol Sci 2021; 22:ijms22105390. [PMID: 34065474 PMCID: PMC8161011 DOI: 10.3390/ijms22105390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/09/2021] [Accepted: 05/11/2021] [Indexed: 11/17/2022] Open
Abstract
Obesity-induced adipose tissue dysfunction and disorders of glycolipid metabolism have become a worldwide research priority. Zfp217 plays a crucial role in adipogenesis of 3T3-L1 preadipocytes, but about its functions in animal models are not yet clear. To explore the role of Zfp217 in high-fat diet (HFD)-induced obese mice, global Zfp217 heterozygous knockout (Zfp217+/−) mice were constructed. Zfp217+/− mice and Zfp217+/+ mice fed a normal chow diet (NC) did not differ significantly in weight gain, percent body fat mass, glucose tolerance, or insulin sensitivity. When challenged with HFD, Zfp217+/− mice had less weight gain than Zfp217+/+ mice. Histological observations revealed that Zfp217+/− mice fed a high-fat diet had much smaller white adipocytes in inguinal white adipose tissue (iWAT). Zfp217+/− mice had improved metabolic profiles, including improved glucose tolerance, enhanced insulin sensitivity, and increased energy expenditure compared to the Zfp217+/+ mice under HFD. We found that adipogenesis-related genes were increased and metabolic thermogenesis-related genes were decreased in the iWAT of HFD-fed Zfp217+/+ mice compared to Zfp217+/− mice. In addition, adipogenesis was markedly reduced in mouse embryonic fibroblasts (MEFs) from Zfp217-deleted mice. Together, these data indicate that Zfp217 is a regulator of energy metabolism and it is likely to provide novel insight into treatment for obesity.
Collapse
|
19
|
Choi EY, Park CY, Ho SH, Park SJ, Kim D, Han B, Kim SH. Anti-Obesity Effects of Artemisia annua Extract in Zucker Fatty Rats and High-Fat Diet Sprague Dawley Rats through Upregulation of Uncoupling Protein 1. J Obes Metab Syndr 2021; 30:32-43. [PMID: 33479187 PMCID: PMC8017327 DOI: 10.7570/jomes20097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/30/2020] [Accepted: 12/02/2020] [Indexed: 11/12/2022] Open
Abstract
Background Obesity is a widespread disease and is caused mainly by excessive adipocyte differentiation and fat accumulation. Peroxisome proliferation-activated receptor γ (PPARγ) and CCAAT/enhancer-binding proteins (C/EBP) are major components for regulating adipocyte differentiation. Uncoupling protein 1 (UCP1) is a transmembrane protein that can convert white fat to brown adipose tissue. Artemisia annua L. has long been used in East Asia as an herbal drug for anti-oxidant, anti-bacterial, and anti-obesity purposes. Methods We investigated the effects of water extracts of A. annua (WEAA) in C3H10T1/2, a mesenchymal stem cell line, by measuring the level of intracellular fat accumulation and the expression of genes associated with adipocyte differentiation. We also evaluated anti-obesity effects of WEAA in Zucker rats, a genetic model for the study of obesity, and in Sprague Dawley rats with high-fat diet (HFD)-induced obesity. Results In this study, WEAA reduced the expression levels of PPARγ and C/EBPα in C3H10T1/2 cells, as well as the expression of enzymes that regulate fatty acid metabolism. In the Zucker fatty rat model and the HFD-induced obesity rat model, WEAA significantly decreased adipogenic differentiation and white fat accumulation between the scapulae, in contrast to the brown fat that remained unchanged between the groups. A. annua suppressed the expression of the adipocyte differentiation-promoting genes, while increasing the expression of UCP1. Conclusion These results indicated that WEAA could reduce adipocyte differentiation and fat accumulation in in vitro and in vivo model systems, resulting in suppression of obesity and the occurrence of fatty liver due to a HFD.
Collapse
Affiliation(s)
| | | | | | | | - Donghyun Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea.,Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Korea
| | - Byoungduck Han
- Department of Family Medicine, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Korea
| | | |
Collapse
|
20
|
Bifidobacterium adolescentis Isolated from Different Hosts Modifies the Intestinal Microbiota and Displays Differential Metabolic and Immunomodulatory Properties in Mice Fed a High-Fat Diet. Nutrients 2021; 13:nu13031017. [PMID: 33801119 PMCID: PMC8004121 DOI: 10.3390/nu13031017] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/15/2021] [Accepted: 03/18/2021] [Indexed: 01/07/2023] Open
Abstract
The incidence of obesity, which is closely associated with the gut microbiota and chronic inflammation, has rapidly increased in the past 40 years. Therefore, the probiotic-based modification of the intestinal microbiota composition has been developed as a strategy for the treatment of obesity. In this study, we selected four Bifidobacterium adolescentis strains isolated from the feces of newborn and elderly humans to investigate whether supplementation with B. adolescentis of various origins could alleviate obesity in mice. Male C57BL/6J mice fed a high-fat diet (HFD, 60% energy as fat) received one of the following 14-week interventions: (i) B. adolescentis N4_N3, (ii) B. adolescentis Z25, (iii) B. adolescentis 17_3, (iv) B. adolescentis 2016_7_2, and (v) phosphate-buffered saline. The metabolic parameters, thermogenesis, and immunity of all treated mice were measured. Cecal and colonic microbial profiles were determined by 16S rRNA gene sequencing. Intestinal concentrations of short-chain fatty acids (SCFAs) were measured by gas chromatography-mass spectrometry (GC-MS). The B. adolescentis strains isolated from the feces of elderly humans (B. adolescentis Z25, 17_3, and 2016_7_2) decreased the body weight or weight gain of mice, whilst the strain isolated from the newborn (B. adolescentis N4_N3) increased the body weight of mice. The B. adolescentis strains isolated from the elderly also increased serum leptin concentrations and induced the expression of thermogenesis- and lipid metabolism-related genes in brown adipose tissue. All the B. adolescentis strains alleviated inflammations in the spleen and brain and modified the cecal and colonic microbiota. Particularly, all strains reversed the HFD-induced depletion of Bifidobacterium and reduced the development of beta-lactam resistance. In addition, the B. adolescentis strains isolated from the elderly increased the relative abundances of potentially beneficial genera, such as Bacteroides, Parabacteroides, and Faecalibaculum. We speculate that such increased abundance of commensal bacteria may have mediated the alleviation of obesity, as B. adolescentis supplementation decreased the intestinal production of SCFAs, thereby reducing energy delivery to the host mice. Our results revealed that certain strains of B. adolescentis can alleviate obesity and modify the gut microbiota of mice. The tested strains of B. adolescentis showed different effects on lipid metabolism and immunity regulation, with these effects related to whether they had been isolated from the feces of newborn or elderly humans. This indicates that B. adolescentis from different sources may have disparate effects on host health possibly due to the transmission of origin-specific functions to the host.
Collapse
|
21
|
Janson B, Prasomthong J, Malakul W, Boonsong T, Tunsophon S. Hibiscus sabdariffa L. calyx extract prevents the adipogenesis of 3T3-L1 adipocytes, and obesity-related insulin resistance in high-fat diet-induced obese rats. Biomed Pharmacother 2021; 138:111438. [PMID: 33721756 DOI: 10.1016/j.biopha.2021.111438] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 02/11/2021] [Accepted: 02/23/2021] [Indexed: 10/21/2022] Open
Abstract
Roselle (Hibiscus sabdariffa) is reported to be beneficial in treating obesity which can develop into a range of metabolic disorders. The molecular mechanisms by which roselle extract works to prevent obesity-related insulin resistance remains poorly understood. We hypothesized that the roselle extract can decrease lipid accumulation and improve insulin resistance by downregulating adipogenesis. The aim of this study is to investigate the protective effect of roselle extract on the mechanism of adipogenesis and prevent complications of the obesity-related insulin resistance in high-fat diet-induced obese rats for 8 weeks. Male Sprague Dawley rats were divided into 4 groups: control (C), high-fat diet (HFD), high-fat diet supplemented with 250 mg/kg BW of roselle (R250), and high-fat diet supplemented with 500 mg/kg BW of roselle (R500). The results demonstrated that roselle had the potential to reduce body weight, food intake, lipid profiles, inflammatory cytokines, lipid peroxidation, serum leptin, insulin and duodenal glucose absorption, while significantly increased glucose uptake of adipose tissue and muscle when compared to the HFD group. Roselle can prevent lipid accumulation by suppressing differentiation of 3T3-L1 adipocyte by downregulating the adipogenic gene expression. The results of this study demonstrated that the molecular mechanism underlying the protective effect of roselle, could be an alternative approach for obesity-related insulin resistance prevention.
Collapse
Affiliation(s)
- Benjarat Janson
- Department of Physiology, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand
| | - Janjira Prasomthong
- Department of Physiology, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand
| | - Wachirawadee Malakul
- Department of Physiology, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand
| | - Tantip Boonsong
- Department of Biochemistry, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand
| | - Sakara Tunsophon
- Department of Physiology, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand; Centre of Excellence for Innovation in Chemistry, Naresuan University, Phitsanulok 65000 Thailand.
| |
Collapse
|
22
|
Evseeva MN, Dyikanov DT, Karagyaur MN, Prikazchikova TA, Sheptulina AF, Balashova MS, Zatsepin TS, Rubtsov YP, Kulebyakin KY. Hematopoietically-expressed homeobox protein HHEX regulates adipogenesis in preadipocytes. Biochimie 2021; 185:68-77. [PMID: 33677034 DOI: 10.1016/j.biochi.2021.02.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 02/15/2021] [Accepted: 02/17/2021] [Indexed: 11/18/2022]
Abstract
Obesity is a key health problem and is associated with a high risk of type 2 diabetes and other metabolic diseases. Increased weight as well as dysregulation of adipocyte homeostasis are the main drivers of obesity. Pathological adipogenesis plays a central role in obesity-related complications such as type 2 diabetes, hypertension and others. Thus, an understanding of the molecular mechanisms involved in physiological and pathogenic adipogenesis can help to develop new strategies to prevent or cure obesity and related diseases. Previously, genetic polymorphisms in the HHEX gene that encodes the homeobox transcription factor HEX (PRH) were found to be associated with type 2 diabetes and high body mass index at birth by GWAS in distinct human populations. To understand whether HHEX has a regulatory function in adipogenesis, we performed RNAi-mediated knockdown of Hhex in preadipocyte cell line 3T3-L1 in vitro, and studied changes in the efficacy of adipogenesis. We found that Hhex knockdown blocks adipogenesis in preadipocytes in a dose-dependent manner and leads to a significant decrease of PPAR-gamma protein - the main regulator of adipogenesis. We also propose that Hhex can play an important role in adipocyte differentiation by affecting the level of the PPAR-gamma protein. Our study supports the claim that Hhex plays an important role in adipocyte differentiation program and can contribute to fat tissue homeostasis.
Collapse
Affiliation(s)
- Maria N Evseeva
- Faculty of Medicine, Lomonosov Moscow State University, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Russia.
| | | | | | - Tatyana A Prikazchikova
- Skolkovo Institute of Science and Technology, Skolkovo, Moscow, Russia; Department of Chemistry, Lomonosov Moscow State University, Moscow, Russia
| | - Anna F Sheptulina
- Department of Gastroenterology and Hepatology, Sechenov 1st State Medical University, Moscow, Russia
| | - Maria S Balashova
- Department of Medical Genetics, Sechenov 1st State Medical University, Moscow, Russia
| | - Timofei S Zatsepin
- Skolkovo Institute of Science and Technology, Skolkovo, Moscow, Russia; Department of Chemistry, Lomonosov Moscow State University, Moscow, Russia
| | - Yury P Rubtsov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Russia
| | | |
Collapse
|
23
|
B G M, Manjappara UV. Obestatin and Rosiglitazone Differentially Modulate Lipid Metabolism Through Peroxisome Proliferator-activated Receptor-γ (PPARγ) in Pre-adipose and Mature 3T3-L1 Cells. Cell Biochem Biophys 2021; 79:73-85. [PMID: 33432549 DOI: 10.1007/s12013-020-00958-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/26/2020] [Indexed: 10/22/2022]
Abstract
Obestatin is a 23-residue peptide, obtained after posttranslational modification of preproghrelin. It has been shown, in Swiss albino mice, to upregulate glycerolipid metabolism and PPARγ signaling. It was opined that the by-products of increased glycerolipid metabolism triggered PPARγ signaling. It was hypothesized that obestatin upon co-administration with a full agonist of PPARγ should reveal the comparative significance or possible synergy in PPARγ signaling. We postulated they would act synergistically by obestatin increasing PPARγ expression and rosiglitazone enhancing PPARγ activity. We evaluated the combination in DIO-C57BL/6 mice and observed that obestatin completely reversed the increase in subcutaneous fat brought about by rosiglitazone. To understand their role at the adipocyte level, 3T3-L1 cells were treated with a combination of obestatin and rosiglitazone during (1) initiation of differentiation and (2) after 14 days from initiation of differentiation when the adipocytes were mature. Interestingly, their influence was mainly adipogenic and showed double lipid accumulation when estimated 14 days after initiation of differentiation. There was an upregulation of Pparγ by fourfold, Hsl by eightfold, Glut4 by fourfold, Leptin by 2.7-fold, Atgl by sixfold, Fasn by sixfold, and Fabp4 by sevenfold at the mRNA level, whereas in mature adipocytes there was a significant decrease in fat accumulation by 20%. There was downregulation of Pparγ, Hsl, Lpl, and Fasn by 0.5-fold at the mRNA level. These results show that the combined influence of obestatin and rosiglitazone is significant and the outcome is dependent on the metabolic stage of the adipocyte.
Collapse
Affiliation(s)
- Mallikarjuna B G
- Department of Lipid Science, CSIR-Central Food Technological Research Institute, Mysore, 570020, India
| | - Uma V Manjappara
- Department of Lipid Science, CSIR-Central Food Technological Research Institute, Mysore, 570020, India.
| |
Collapse
|
24
|
Puckett D, Alquraishi M, Alani DS, Chahed S, Frankel VD, Donohoe D, Voy B, Whelan J, Bettaieb A. Zyflamend, a unique herbal blend, induces cell death and inhibits adipogenesis through the coordinated regulation of PKA and JNK. Adipocyte 2020; 9:454-471. [PMID: 32779962 PMCID: PMC7469463 DOI: 10.1080/21623945.2020.1803642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
The prevalence of obesity and its comorbidities has sparked a worldwide concern to address rates of adipose tissue accrual. Recent studies have demonstrated a novel role of Zyflamend, a blend of natural herbal extracts, in regulating lipid metabolism in several cancer cell lines through the activation of the AMPK signalling pathway. Yet, the role of Zyflamend in adipogenic differentiation and lipid metabolism remains largely unexplored. The objective of this study is to investigate the effects of Zyflamend on white 3T3-MBX pre-adipocyte differentiation and elucidate the molecular mechanisms. We demonstrate that Zyflamend treatment altered cell cycle progression, attenuated proliferation, and increased cell death of 3T3-MBX pre-adipocytes. In addition, treatment with Zyflamend inhibited lipid accumulation during the differentiation of 3T3-MBX cells, consistent with decreased expression of lipogenic genes and increased lipolysis. Mechanistically, Zyflamend-induced alterations in adipogenesis were mediated, at least in part, through the activation of AMPK, PKA, and JNK. Inhibition of AMPK partially reversed Zyflamend-induced inhibition of differentiation, whereas the inhibition of either JNK or PKA fully restored adipocyte differentiation and decreased lipolysis. Taken together, the present study demonstrates that Zyflamend, as a novel anti-adipogenic bioactive mix, inhibits adipocyte differentiation through the activation of the PKA and JNK pathways. Abbreviation: 7-AAD: 7-amino-actinomycin D; ACC: acetyl-CoA carboxylase; AKT: protein kinase B; AMPK: AMP-activated protein kinase; ATGL: adipose triglyceride lipase; C/EBPα: CCAAT-enhancer binding protein alpha; DMEM: Dulbecco’s Modified Eagle Medium; DMSO: dimethyl sulphoxide; DTT: dithiothreitol; EGTA: ethylene glycol-bis-(2-aminoethyl)-N,N,N’,N’-tetraacetic acid; ERK: extracellular signal–regulated kinases; FASN: fatty acid synthase; FBS: foetal bovine serum; GLUT: glucose transporter; HSL: hormone-sensitive lipase; IR: insulin receptor; IRS: insulin receptor substrate; JNK: c-JUN N-terminal kinase; MGL: monoacylglycerol lipase; NaF: sodium fluoride; NF-κB: nuclear factor kappa-light-chain-enhancer of activated B cells; PBS: phosphate buffered- saline; PCB: pyruvate carboxylase; PDE: phosphodiesterase; PKA: protein kinase cAMP-dependent; PMSF: phenylmethylsulfonyl fluoride; PPARγ: perilipin peroxisome proliferator-activated receptor gamma; PREF-1: pre-adipocyte factor 1; PVDF: polyvinylidene fluoride; RIPA: radio-immunoprecipitation assay; SDS-PAGE: sodium dodecyl sulphate polyacrylamide gel electrophoresis; SEM: standard error of the mean; SOX9: suppressor of cytokine signalling 9; TGs: triacylglycerols.
Collapse
Affiliation(s)
- Dexter Puckett
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN, USA
| | - Mohammed Alquraishi
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN, USA
| | - Dina S. Alani
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN, USA
| | - Samah Chahed
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN, USA
| | - Victoria D. Frankel
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN, USA
| | - Dallas Donohoe
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN, USA
| | - Brynn Voy
- Tennessee Agricultural Experiment Station, University of Tennessee Institute of Agriculture, Knoxville, TN, USA
- Graduate School of Genome Science and Technology, University of Tennessee, Knoxville, TN, USA
| | - Jay Whelan
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN, USA
- Tennessee Agricultural Experiment Station, University of Tennessee Institute of Agriculture, Knoxville, TN, USA
| | - Ahmed Bettaieb
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN, USA
- Graduate School of Genome Science and Technology, University of Tennessee, Knoxville, TN, USA
- Department of Biochemistry, Cellular and Molecular Biology, University of Tennessee, Knoxville, TN, USA
| |
Collapse
|
25
|
Comparison of Orbital Preadipocyte Adipogenesis Between Pediatric and Adult Graves Ophthalmopathy. J Craniofac Surg 2020; 31:727-730. [DOI: 10.1097/scs.0000000000005907] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
26
|
Moon S, Kim YH, Choi K. Inhibition of 3T3-L1 Adipocyte Differentiation by D-allulose. BIOTECHNOL BIOPROC E 2020. [DOI: 10.1007/s12257-019-0352-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
27
|
Mallikarjuna BG, Manjappara UV. Co-administration with Obestatin Reduces Accumulation of Subcutaneous Fat Due to Rosiglitazone Administration in DIO-C57BL/6 Mice. Int J Pept Res Ther 2020. [DOI: 10.1007/s10989-020-10028-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
28
|
Abstract
Understanding the mammalian energy balance can pave the way for future therapeutics that enhance energy expenditure as an anti-obesity and anti-diabetic strategy. Several studies showed that brown adipose tissue activity increases daily energy expenditure. However, the size and activity of brown adipose tissue is reduced in individuals with obesity and type two diabetes. Humans have an abundance of functionally similar beige adipocytes that have the potential to contribute to increased energy expenditure. This makes beige adipocytes a promising target for metabolic disease therapies. While brown adipocytes tend to be stable, beige adipocytes have a high level of plasticity that allows for the rapid and dynamic induction of thermogenesis by external stimuli such as low environmental temperatures. This means that after browning stimuli have been withdrawn beige adipocytes quickly transition back to their white adipose state. The detailed molecular mechanisms regulating beige adipocytes development, function, and reversibility are not fully understood. The goal of this review is to give a comprehensive overview of beige fat development and origins, along with the transcriptional and epigenetic programs that lead to beige fat formation, and subsequent thermogenesis in humans. An improved understanding of the molecular pathways of beige adipocyte plasticity will enable us to selectively manipulate beige cells to induce and maintain their thermogenic output thus improving the whole-body energy homeostasis.
Collapse
|
29
|
Chen Y, Zhao Y, Jin W, Li Y, Zhang Y, Ma X, Sun G, Han R, Tian Y, Li H, Kang X, Li G. MicroRNAs and their regulatory networks in Chinese Gushi chicken abdominal adipose tissue during postnatal late development. BMC Genomics 2019; 20:778. [PMID: 31653195 PMCID: PMC6815035 DOI: 10.1186/s12864-019-6094-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Accepted: 09/10/2019] [Indexed: 12/13/2022] Open
Abstract
Background Abdominal fat is the major adipose tissue in chickens. The growth status of abdominal fat during postnatal late development ultimately affects meat yield and quality in chickens. MicroRNAs (miRNAs) are endogenous small noncoding RNAs that regulate gene expression at the post-transcriptional level. Studies have shown that miRNAs play an important role in the biological processes involved in adipose tissue development. However, few studies have investigated miRNA expression profiles and their interaction networks associated with the postnatal late development of abdominal adipose tissue in chickens. Results We constructed four small RNA libraries from abdominal adipose tissue obtained from Chinese domestic Gushi chickens at 6, 14, 22, and 30 weeks. A total of 507 known miRNAs and 53 novel miRNAs were identified based on the four small RNA libraries. Fifty-one significant differentially expressed (SDE) miRNAs were identified from six combinations by comparative analysis, and the expression patterns of these SDE miRNAs were divided into six subclusters by cluster analysis. Gene ontology enrichment analysis showed that the SDE miRNAs were primarily involved in the regulation of fat cell differentiation, regulation of lipid metabolism, regulation of fatty acid metabolism, and unsaturated fatty acid metabolism in the lipid metabolism- or deposition-related biological process categories. In addition, we constructed differentially expressed miRNA–mRNA interaction networks related to abdominal adipose development. The results showed that miRNA families, such as mir-30, mir-34, mir-199, mir-8, and mir-146, may have key roles in lipid metabolism, adipocyte proliferation and differentiation, and cell junctions during abdominal adipose tissue development in chickens. Conclusions This study determined the dynamic miRNA transcriptome and characterized the miRNA–mRNA interaction networks in Gushi chicken abdominal adipose tissue for the first time. The results expanded the number of known miRNAs in abdominal adipose tissue and provide novel insights and a valuable resource to elucidate post-transcriptional regulation mechanisms during postnatal late development of abdominal adipose tissue in chicken.
Collapse
Affiliation(s)
- Yi Chen
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zheng zhou, Henan Province, 450002, People's Republic of China
| | - Yinli Zhao
- College of Biological Engineering, Henan University of Technology, Zheng zhou, Henan Province, 450001, People's Republic of China
| | - Wenjiao Jin
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zheng zhou, Henan Province, 450002, People's Republic of China
| | - Yuanfang Li
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zheng zhou, Henan Province, 450002, People's Republic of China
| | - Yanhua Zhang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zheng zhou, Henan Province, 450002, People's Republic of China
| | - Xuejie Ma
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zheng zhou, Henan Province, 450002, People's Republic of China
| | - Guirong Sun
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zheng zhou, Henan Province, 450002, People's Republic of China
| | - Ruili Han
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zheng zhou, Henan Province, 450002, People's Republic of China
| | - Yadong Tian
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zheng zhou, Henan Province, 450002, People's Republic of China
| | - Hong Li
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zheng zhou, Henan Province, 450002, People's Republic of China
| | - Xiangtao Kang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zheng zhou, Henan Province, 450002, People's Republic of China
| | - Guoxi Li
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zheng zhou, Henan Province, 450002, People's Republic of China.
| |
Collapse
|
30
|
Han XQ, Xu SQ, Lin JG. Curcumin Recovers Intracellular Lipid Droplet Formation Through Increasing Perilipin 5 Gene Expression in Activated Hepatic Stellate Cells In Vitro. Curr Med Sci 2019; 39:766-777. [DOI: 10.1007/s11596-019-2104-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 09/02/2019] [Indexed: 02/06/2023]
|
31
|
Wang Z, Dai Z, Luo Z, Zuo C. Identification of Pyrvinium, an Anthelmintic Drug, as a Novel Anti-Adipogenic Compound Based on the Gene Expression Microarray and Connectivity Map. Molecules 2019; 24:molecules24132391. [PMID: 31261664 PMCID: PMC6650900 DOI: 10.3390/molecules24132391] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 06/23/2019] [Accepted: 06/27/2019] [Indexed: 02/07/2023] Open
Abstract
Obesity is a serious health problem, while the current anti-obesity drugs are not very effective. The Connectivity Map (C-Map), an in-silico drug screening approach based on gene expression profiles, has recently been indicated as a promising strategy for drug repositioning. In this study, we performed mRNA expression profile analysis using microarray technology and identified 435 differentially expressed genes (DEG) during adipogenesis in both C3H10T1/2 and 3T3-L1 cells. Then, DEG signature was uploaded into C-Map, and using pattern-matching methods we discovered that pyrvinium, a classical anthelminthic, is a novel anti-adipogenic differentiation agent. Pyrvinium suppressed adipogenic differentiation in a dose-dependent manner, as evidenced by Oil Red O staining and the mRNA levels of adipogenic markers. Furthermore, we identified that the inhibitory effect of pyrvinium was resulted primarily from the early stage of adipogenesis. Molecular studies showed that pyrvinium downregulated the expression of key transcription factors C/EBPa and PPARγ. The mRNA levels of notch target genes Hes1 and Hey1 were obviously reduced after pyrvinium treatment. Taken together, this study identified many differentially expressed genes involved in adipogenesis and demonstrated for the first time that pyrvinium is a novel anti-adipogenic compound for obesity therapy. Meanwhile, we provided a new strategy to explore potential anti-obesity drugs.
Collapse
Affiliation(s)
- Zonggui Wang
- Department of Biochemistry and Molecular Biology, Guangdong Medical University, Dongguan 523808, Guangdong, China
| | - Zhong Dai
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Zhanjiang 524023, Guangdong, China
- School of Pharmacy, Guangdong Medical University, Dongguan 523808, Guangdong, China
| | - Zhicong Luo
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Zhanjiang 524023, Guangdong, China
- School of Pharmacy, Guangdong Medical University, Dongguan 523808, Guangdong, China
| | - Changqing Zuo
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Zhanjiang 524023, Guangdong, China.
- School of Pharmacy, Guangdong Medical University, Dongguan 523808, Guangdong, China.
| |
Collapse
|
32
|
Jang M, Choi HY, Kim GH. Phenolic components rich ethyl acetate fraction of Orostachys japonicus inhibits lipid accumulation by regulating reactive oxygen species generation in adipogenesis. J Food Biochem 2019; 43:e12939. [PMID: 31368568 DOI: 10.1111/jfbc.12939] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 03/13/2019] [Accepted: 04/23/2019] [Indexed: 12/11/2022]
Abstract
In this study, Orostachys japonicus was extracted with ethyl alcohol and fractionated by a serial of organic solvents. The ethyl acetate fraction was found to be the most effective among the tested five fractions. High-performance liquid chromatography and mass spectrometry analysis of the ethyl acetate fraction presented epicatechin gallate, quercetin-3-O-glucoside, and kaempferol-3-O-rutinoside. Treatment with O. japonicus inhibited reactive oxygen species (ROS) generation and lipid accumulation during adipogenesis. The gene expression of enzymes involved in the antioxidant system increased in O. japonicus-treated cells. messeanger RNA (mRNA) and protein expression of the pro-oxidant enzymes such as nicotinamide adenine dinucleotide phosphate hydrogen oxidase4 and glucose-6-phosphate dehydrogenase suppressed in O. japonicus-treated cells. O. japonicus also inhibited the mRNA and protein levels of adipogenic transcription factors (including proliferator activated receptor-γ and CCAAT/enhancer-binding protein-α) and their target gene (adipocyte protein 2). These results suggest that O. japonicus inhibits adipogenesis by controlling pro-/anti-oxidant enzyme responses and adipogenic transcription factors. PRACTICAL APPLICATIONS: ROS generation is markedly related to the pathogenesis and development of metabolic disorders. Treatment with O. japonicus inhibited ROS generation and lipid accumulation during adipogenesis. This result indicates that O. japonicus inhibit adipogenesis by controlling pro-/anti-oxidant enzyme responses and adipogenic mediators.
Collapse
Affiliation(s)
- Miran Jang
- Plant Resources Research Institute, Duksung Women's University, Seoul, Republic of Korea
| | - Hye-Young Choi
- Plant Resources Research Institute, Duksung Women's University, Seoul, Republic of Korea
| | - Gun-Hee Kim
- Plant Resources Research Institute, Duksung Women's University, Seoul, Republic of Korea
| |
Collapse
|
33
|
Tian JJ, Lei CX, Ji H, Zhou JS, Yu HB, Li Y, Yu EM, Xie J. Dietary arachidonic acid decreases the expression of transcripts related to adipocyte development and chronic inflammation in the adipose tissue of juvenile grass carp, Ctenopharyngodon idella. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY D-GENOMICS & PROTEOMICS 2019; 30:122-132. [DOI: 10.1016/j.cbd.2019.02.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 02/18/2019] [Accepted: 02/20/2019] [Indexed: 12/30/2022]
|
34
|
Koo SY, Hwang JH, Yang SH, Um JI, Hong KW, Kang K, Pan CH, Hwang KT, Kim SM. Anti-Obesity Effect of Standardized Extract of Microalga Phaeodactylum tricornutum Containing Fucoxanthin. Mar Drugs 2019; 17:md17050311. [PMID: 31137922 PMCID: PMC6562887 DOI: 10.3390/md17050311] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 05/23/2019] [Accepted: 05/24/2019] [Indexed: 12/22/2022] Open
Abstract
Fucoxanthin (FX), a marine carotenoid found in macroalgae and microalgae, exhibits several beneficial effects to health. The anti-obesity activity of FX is well documented, but FX has not been mass-produced or applied extensively or commercially because of limited availability of raw materials and complex extraction techniques. In this study, we investigated the anti-obesity effect of standardized FX powder (Phaeodactylum extract (PE)) developed from microalga Phaeodactylum tricornutum as a commercial functional food. The effects of PE on adipogenesis inhibition in 3T3-L1 adipocytes and anti-obesity in high-fat diet (HFD)-fed C57BL/6J mice were evaluated. PE and FX dose-dependently decreased intracellular lipid contents in adipocytes without cytotoxicity. In HFD-fed obese mice, PE supplementation for six weeks decreased body weight, organ weight, and adipocyte size. In the serum parameter analysis, the PE-treated groups showed attenuation of lipid metabolism dysfunction and liver damage induced by HFD. In the liver, uncoupling protein-1 (UCP1) upregulation and peroxisome proliferator activated receptor γ (PPARγ) downregulation were detected in the PE-treated groups. Additionally, micro computed tomography revealed lower fat accumulation in PE-treated groups compared to that in the HFD group. These results indicate that PE exerts anti-obesity effects by inhibiting adipocytic lipogenesis, inducing fat mass reduction and decreasing intracellular lipid content, adipocyte size, and adipose weight.
Collapse
Affiliation(s)
- Song Yi Koo
- Natural Product Informatics Center, KIST Gangneung Institute of Natural Products, Gangneung 25451, Korea.
- Department of Food and Nutrition, and Research Institute of Human Ecology, Seoul National University, Seoul 08826, Korea.
| | - Ji-Hyun Hwang
- Natural Product Informatics Center, KIST Gangneung Institute of Natural Products, Gangneung 25451, Korea.
| | - Seung-Hoon Yang
- Department of Medical Biotechnology, College of Life Science and Biotechnology, Dongguk University, Seoul 04620, Korea.
| | - Jae-In Um
- R&D Department, AlgaeTech Co. Ltd., Gangneung 25457, Korea.
| | - Kwang Won Hong
- R&D Department, AlgaeTech Co. Ltd., Gangneung 25457, Korea.
| | - Kyungsu Kang
- Natural Product Informatics Center, KIST Gangneung Institute of Natural Products, Gangneung 25451, Korea.
- Division of Bio-Medical Science & Technology, University of Science & Technology, Daejeon 34113, Korea.
| | - Cheol-Ho Pan
- Natural Product Informatics Center, KIST Gangneung Institute of Natural Products, Gangneung 25451, Korea.
- Division of Bio-Medical Science & Technology, University of Science & Technology, Daejeon 34113, Korea.
| | - Keum Taek Hwang
- Department of Food and Nutrition, and Research Institute of Human Ecology, Seoul National University, Seoul 08826, Korea.
| | - Sang Min Kim
- Smart Farm Research Center, KIST Gangneung Institute of Natural Products, Gangneung 25451, Korea.
- Division of Bio-Medical Science & Technology, University of Science & Technology, Daejeon 34113, Korea.
| |
Collapse
|
35
|
Park M, Sharma A, Lee HJ. Anti-Adipogenic Effects of Delphinidin-3- O- β-Glucoside in 3T3-L1 Preadipocytes and Primary White Adipocytes. Molecules 2019; 24:E1848. [PMID: 31091729 PMCID: PMC6571603 DOI: 10.3390/molecules24101848] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 05/04/2019] [Accepted: 05/08/2019] [Indexed: 12/17/2022] Open
Abstract
Delphinidin-3-O-β-glucoside (D3G) is a health-promoting anthocyanin whose anti-obesity activity has not yet been thoroughly investigated. We examined the effects of D3G on adipogenesis and lipogenesis in 3T3-L1 adipocytes and primary white adipocytes using real-time RT-PCR and immunoblot analysis. D3G significantly inhibited the accumulation of lipids in a dose-dependent manner without displaying cytotoxicity. In the 3T3-L1 adipocytes, D3G downregulated the expression of key adipogenic and lipogenic markers, which are known as peroxisome proliferator-activated receptor gamma (PPARγ), sterol regulatory element-binding transcription factor 1 (SREBP1), CCAAT/enhancer-binding protein alpha (C/EBPα), and fatty acid synthase (FAS). Moreover, the relative protein expression of silent mating type information regulation 2 homolog 1 (SIRT1) and carnitine palmitoyltransferase-1 (CPT-1) were increased, alongside reduced lipid levels and the presence of several small lipid droplets. Furthermore, D3G increased the phosphorylation of adenosine monophosphate-activated protein kinase (AMPK) and acetyl-CoA carboxylase (ACC), which suggests that D3G may play a role in AMPK and ACC activation in adipocytes. Our data indicate that D3G attenuates adipogenesis and promotes lipid metabolism by activating AMPK-mediated signaling, and, hence, could have a therapeutic role in the management and treatment of obesity.
Collapse
Affiliation(s)
- Miey Park
- Department of Food and Nutrition, Gachon University, Gyeonggi-do 13120, Korea.
| | - Anshul Sharma
- Department of Food and Nutrition, Gachon University, Gyeonggi-do 13120, Korea.
| | - Hae-Jeung Lee
- Department of Food and Nutrition, Gachon University, Gyeonggi-do 13120, Korea.
| |
Collapse
|
36
|
Li W, Zhang Y, Chen C, Tian W, Wang H. Increased Angiogenic and Adipogenic Differentiation Potentials in Adipose-Derived Stromal Cells from Thigh Subcutaneous Adipose Depots Compared with Cells from the Abdomen. Aesthet Surg J 2019; 39:NP140-NP149. [PMID: 30358810 DOI: 10.1093/asj/sjy252] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Adipose-derived stromal cells (ADSCs) may play a pivotal role by differentiating into multilineage cells or by secreting growth factors or cytokines in cell-assisted lipotransfer, which participates in adipose tissue regeneration. The angiogenic potential of various ADSCs from different anatomical regions remains uncertain. OBJECTIVES The authors sought to offer appropriate choices of sources of adipose-derived stromal cells for cell-assisted lipotransfer and tissue engineering. METHODS ADSCs were harvested from subcutaneous adipose depots in the abdomen and thighs. The expression of adipocyte-specific markers was evaluated, and Oil Red O staining was performed to assess the capacity for adipogenic differentiation. Angiogenic differentiation potential was evaluated by detecting the expression of vascular endothelial growth factor, vascular endothelial growth factor 2, and CD31. A tube formation assay was also performed to analyze the angiogenic differentiation capacity. RESULTS ADSCs from the thigh showed more significant angiogenic and adipogenic potential. More lipogenesis was identified in ADSCs from the thigh, and this was accompanied by the enhancement of adipocyte markers. Angiogenesis was more vigorous in the thigh-derived stromal cells, and ADSCs from the thigh depot showed more junctions and longer tubule formation on Matrigel in vitro. CONCLUSIONS Thigh-derived ADSCs exhibited greater capacity for adipogenic and angiogenic differentiation and would be a better option for cell-assisted lipotransfer and tissue engineering.
Collapse
Affiliation(s)
- Wei Li
- Department of Oral and Maxillofacial Surgery, Changzhou Stomatologic Hospital, Changzhou, China
| | - Yan Zhang
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chang Chen
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Weidong Tian
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology
| | - Hang Wang
- Department of Oral and Maxillofacial Surgery, Sichuan University, Chengdu, China
- Department of Cosmetic Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
37
|
Chen J, Lu Y, Tian M, Huang Q. Molecular mechanisms of FOXO1 in adipocyte differentiation. J Mol Endocrinol 2019; 62:R239-R253. [PMID: 30780132 DOI: 10.1530/jme-18-0178] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 02/05/2019] [Indexed: 12/14/2022]
Abstract
Forkhead box-O1 (FOXO1) is a downstream target of AKT and plays crucial roles in cell cycle control, apoptosis, metabolism and adipocyte differentiation. It is thought that FOXO1 affects adipocyte differentiation by regulating lipogenesis and cell cycle. With the deepening in the understanding of this field, it is currently believed that FOXO1 translocation between nuclei and cytoplasm is involved in the regulation of FOXO1 activity, thus affecting adipocyte differentiation. Translocation of FOXO1 depends on its post-translational modifications and interactions with 14-3-3. Based on these modifications and interactions, FOXO1 could regulate lipogenesis through PPARγ and the adipocyte cell cycle through p21 and p27. In this review, we aim to provide a comprehensive FOXO1 regulation network in adipocyte differentiation by linking together distinct functions mentioned above to explain their effects on adipocyte differentiation and to emphasize the regulatory role of FOXO1. In addition, we also focus on the novel findings such as the use of miRNAs in FOXO1 regulation and highlight the improvable issues, such as RNA modifications, for future research in the field.
Collapse
Affiliation(s)
- Junye Chen
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, People's Republic of China
- Nanchang Joint Programme, Queen Mary, University of London, London, UK
| | - Yi Lu
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi Province, People's Republic of China
- Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, Jiangxi Province, People's Republic of China
| | - Mengyuan Tian
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, People's Republic of China
- Nanchang Joint Programme, Queen Mary, University of London, London, UK
| | - Qiren Huang
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi Province, People's Republic of China
- Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, Jiangxi Province, People's Republic of China
| |
Collapse
|
38
|
Berkoz M. EFFECT OF HYPEROSIDE ON THE INHIBITION OF ADIPOGENESIS IN 3T3-L1 ADIPOCYTES. ACTA ENDOCRINOLOGICA-BUCHAREST 2019; 15:165-172. [PMID: 31508172 DOI: 10.4183/aeb.2019.165] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Context The inhibition of adipocyte differentiation has a significant role on the prevention of obesity and obesity-associated complications. Objective In this study, we aimed to detect whether hyperoside is able to inhibit the conversion of pre-adiposits into mature adiposits. Design and Methods 3T3-L1 pre-adipocytes were stimulated so as to differentiate into mature adipocytes. Hyperoside in non-cytotoxic concentrations (1, 2, 5, and 10 µM) were separately applied to differentiated 3T3-L1 cells. Oil red O staining was performed and triacylglycerol contents were measured. Furthermore, gene and protein expressions of transcription factors, adipogenic genes and adipokines were examined in order to investigate the effect of hyperoside on adipocyte differentiation. Results Hyperoside in high concentrations significantly suppressed the adipogenic process by inhibiting the expression of transcription factors and adipogenic genes and reducing lipid accumulation in adipocytes (p<0.05). Low doses of hyperoside are able to inhibit adipogenesis, but higher doses are needed to reduce fat accumulation in mature adipocytes. In the case of maturing preadipocytes, 5 μM of hyperoside exerts its antiadipogenic effect at the early stages of adipogenesis, whereas 10 μM of hyperoside acts at the later stages (p<0.05). Conclusion These results suggest that hyperoside has a beneficial effect on the prevention and treatment of obesity.
Collapse
Affiliation(s)
- M Berkoz
- Yuzuncu Yil University, Faculty of Pharmacy - Department of Biochemistry, Van, Turkey
| |
Collapse
|
39
|
Xie YY, Mo CL, Cai YH, Wang WJ, Hong XX, Zhang KK, Liu QF, Liu YJ, Hong JJ, He T, Zheng ZZ, Mo W, Li BA. Pygo2 Regulates Adiposity and Glucose Homeostasis via β-Catenin-Axin2-GSK3β Signaling Pathway. Diabetes 2018; 67:2569-2584. [PMID: 30279163 DOI: 10.2337/db18-0311] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 09/13/2018] [Indexed: 11/13/2022]
Abstract
Wnt/β-catenin signaling plays a key role in regulating adipogenesis through indirectly inhibiting the expression of C/EBPα and peroxisome proliferator-activated receptor γ (PPARγ); however, the detailed molecular mechanism remains poorly understood. Moreover, the factor(s) that determines the Wnt/β-catenin output level during adipogenesis is also not completely defined. In this study, we showed that Pygo2 exhibited a declined expression pattern during adipocyte differentiation, resulting in an attenuated Wnt/β-catenin output level. The mechanism study indicated that Pygo2 inhibition led to the downregulation of Axin2, a constitutive Wnt target, in the cytoplasm. Consequently, Axin2-bound GSK3β was released and translocated into the nucleus to phosphorylate C/EBPβ and Snail, resulting in an increase in the DNA binding activity of C/EBPβ and decreased protein stability of Snail, which subsequently activated the expression of C/EBPα and PPARγ. Consistent with this, embryonic fibroblasts from Pygo2-/- mice exhibited spontaneous adipocyte differentiation, and adipocyte precursor-specific Pygo2-deficient mice exhibited increased adiposity with decreased energy expenditure. We further showed impaired glucose tolerance and decreased systemic insulin sensitivity in Pygo2-deficient mice. Our study revealed an association between Pygo2 function and obesity or diabetes.
Collapse
Affiliation(s)
- Yuan-Yuan Xie
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
- Engineering Research Center of Molecular Diagnostics, Ministry of Education, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Chun-Li Mo
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
- Engineering Research Center of Molecular Diagnostics, Ministry of Education, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Yi-Huang Cai
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
- Engineering Research Center of Molecular Diagnostics, Ministry of Education, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Wen-Jie Wang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
- Engineering Research Center of Molecular Diagnostics, Ministry of Education, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Xin-Xin Hong
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
- Engineering Research Center of Molecular Diagnostics, Ministry of Education, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Kun-Kun Zhang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
- Engineering Research Center of Molecular Diagnostics, Ministry of Education, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Qing-Feng Liu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
- Engineering Research Center of Molecular Diagnostics, Ministry of Education, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Yun-Jia Liu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
- Engineering Research Center of Molecular Diagnostics, Ministry of Education, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Jing-Jing Hong
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
- Engineering Research Center of Molecular Diagnostics, Ministry of Education, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Ting He
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
- Engineering Research Center of Molecular Diagnostics, Ministry of Education, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Zhong-Zheng Zheng
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
- Engineering Research Center of Molecular Diagnostics, Ministry of Education, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Wei Mo
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
- Engineering Research Center of Molecular Diagnostics, Ministry of Education, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Bo-An Li
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
- Engineering Research Center of Molecular Diagnostics, Ministry of Education, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
40
|
Lee HW, Pyo S. Acrylamide induces adipocyte differentiation and obesity in mice. Chem Biol Interact 2018; 298:24-34. [PMID: 30409764 DOI: 10.1016/j.cbi.2018.10.021] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 10/19/2018] [Accepted: 10/24/2018] [Indexed: 10/28/2022]
Abstract
Obesity is a critical risk factor for various diseases including type II diabetes, cerebral infarction, cardiovascular diseases, and various cancers. Acrylamide (ACR) is present in wide range of foods, including fried potato products, root vegetables, bakery products, chips, cakes, cereals, and coffee. In this study, ACR treatment dramatically increased the accumulation of lipid droplets. We also examined expression levels of peroxisome proliferator-activated receptors γ (PPARγ), CCAAT enhancer binding protein α (c/EBPα), and CCAAT enhancer binding protein β (c/EBPβ) as adipogenic transcription factors for adipocyte differentiation. They were dose-dependently increased by treatment of ACR. Moreover, effects of ACR on mitogen-activated protein kinases (MAPKs) and 5' AMP-activated protein kinase (AMPK)-Acetyl-CoA carboxylase (ACC) activation were investigated. Results also showed that ACR induced phosphorylation of MAPKs and AMPK-ACC. ACR also induced expression of adipocyte fatty acid-binding protein (aP2), lipoprotein lipase (LPL), sterol regulatory element-binding protein (SREBP)-1c, and fatty acid synthase (FAS). Exposure of ACR to high fat diet (HFD)-fed mice significantly increased body weight, organ weight, and fat mass of mice. Collectively, these result showed that ACR can act as an enhancer of adipocyte. Therefore, we suggest that up-regulation of the adipogenesis by ACR may be related to the regulation of the MAPKs and AMPK-ACC pathway.
Collapse
Affiliation(s)
- Hee-Weon Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Suhkneung Pyo
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| |
Collapse
|
41
|
Lu T, Pei W, Wang K, Zhang S, Chen F, Wu Y, Guan W. In vitro culture and biological properties of broiler adipose-derived stem cells. Exp Ther Med 2018; 16:2399-2407. [PMID: 30210592 PMCID: PMC6122567 DOI: 10.3892/etm.2018.6445] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Accepted: 05/04/2018] [Indexed: 02/07/2023] Open
Abstract
In the past 10 years, adipose-derived stem cells (ADSCs) have been applied due to their pluripotency. Experimental tissues have been frequently obtained from mammals, including rabbits, mice and humans, but rarely from broilers, Gallus gallus domesticus. In the present study, ADSCs were obtained from 20-day-old broiler embryos. Primary ADSCs were sub-cultured to passage 37 in vitro. The surface markers of ADSCs, namely CD29, CD31, CD44, CD71 and CD73, were detected by reverse transcription polymerase chain reaction and immunofluorescence assays. The result indicated that CD29, CD44, CD71 and CD73 were expressed on the surface of cells at various passages, but not CD31. The growth curve of cells at the different passages had a typical sigmoidal shape. Furthermore, ADSCs were successfully induced to differentiate into osteoblasts, adipocytes and hepatocyte-like cells. The results denote that the ADSCs isolated from broilers have similar biological properties to those of ADSCs obtained from other animals. The present study provided a theoretical and experimental foundation for the use of poultry as a source of stem cells, and laid a foundation for adipose tissue engineering and strategies in regenerative medicine.
Collapse
Affiliation(s)
- Tengfei Lu
- Department of Animal Genetic Resources, Institute of Animal Science, Chinese Academy of Agricultural Science, Beijing 100193, P.R. China
| | - Wenhua Pei
- Department of Animal Genetic Resources, Institute of Animal Science, Chinese Academy of Agricultural Science, Beijing 100193, P.R. China
| | - Kunfu Wang
- College of Wildlife Resources, Northeast Forestry University, Harbin, Heilongjiang 150040, P.R. China
| | - Shuang Zhang
- Scientific Experiment Research Center, Harbin Sport University, Harbin, Heilongjiang 150008, P.R. China
| | - Fenghao Chen
- College of Human Movement Science, Harbin Sport University, Harbin, Heilongjiang 150008, P.R. China
| | - Yangnan Wu
- Department of Animal Genetic Resources, Institute of Animal Science, Chinese Academy of Agricultural Science, Beijing 100193, P.R. China
| | - Weijun Guan
- Department of Animal Genetic Resources, Institute of Animal Science, Chinese Academy of Agricultural Science, Beijing 100193, P.R. China
| |
Collapse
|
42
|
Liu P, Hsieh P, Lin H, Liu T, Wu H, Chen C, Chen Y. Grail is involved in adipocyte differentiation and diet-induced obesity. Cell Death Dis 2018; 9:525. [PMID: 29743578 PMCID: PMC5943410 DOI: 10.1038/s41419-018-0596-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 04/04/2018] [Accepted: 04/16/2018] [Indexed: 01/24/2023]
Abstract
Grail is a crucial regulator of various biological processes, including the development of T-cell anergy, antiviral innate immune response, and cancer. However, the role of Grail in adipogenesis and obesity remains unclear. Here, we demonstrated that Grail knockdown in vitro leads to a decrease in PPARγ expression, resulting in adipogenesis inhibition. However, Grail overexpression induced the same effects. Grail was shown to interact with PPARγ, targeting it for degradation and modulating its adipogenic activity. PPARγ expression was shown to be considerably reduced in Grail knockout (KO) mice fed normal diet or high-fat diet (HFD). The administration of both normal diet or HFD to Grail KO mice led to lower adipose mass and body weight than those in the wild-type mice. HFD-fed Grail KO mice had improved glucose and insulin tolerance. Taken together, our results indicate that Grail plays a pivotal role in adipogenesis and diet-induced obesity by regulating PPARγ activity.
Collapse
Affiliation(s)
- Peiyao Liu
- Department of Physiology & Biophysics, National Defense Medical Center, Taipei, Taiwan, 114, Republic of China
| | - Poshiuan Hsieh
- Department of Physiology & Biophysics, National Defense Medical Center, Taipei, Taiwan, 114, Republic of China.,Institute of Preventive Medicine, National Defense Medical Center, New Taipei City, Taiwan, 114, Republic of China
| | - Huitsu Lin
- Institute of Preventive Medicine, National Defense Medical Center, New Taipei City, Taiwan, 114, Republic of China
| | - Tejung Liu
- Department of Physical Medicine and Rehabilitation, Tri-Service General Hospital, Taipei, Taiwan, 114, Republic of China.,Department of Physical Medicine and Rehabilitation, School of Medicine, National Defense Medical Center, Taipei, Taiwan, 114, Republic of China.,Department of Physical Medicine and Rehabilitation, Taoyuan Armed Force General Hospital, Taoyuan, Taiwan, 114, Republic of China
| | - Hsuehling Wu
- Institute of Preventive Medicine, National Defense Medical Center, New Taipei City, Taiwan, 114, Republic of China
| | - Chengcheung Chen
- Institute of Preventive Medicine, National Defense Medical Center, New Taipei City, Taiwan, 114, Republic of China
| | - Yingchuan Chen
- Institute of Preventive Medicine, National Defense Medical Center, New Taipei City, Taiwan, 114, Republic of China.
| |
Collapse
|
43
|
Liotti A, Cabaro S, Cimmino I, Ricci S, Procaccini C, Paciello O, Raciti GA, Spinelli R, Iossa S, Matarese G, Miele C, Formisano P, Beguinot F, Oriente F. Prep1 deficiency improves metabolic response in white adipose tissue. Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1863:515-525. [PMID: 29474930 DOI: 10.1016/j.bbalip.2018.02.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 02/09/2018] [Accepted: 02/17/2018] [Indexed: 12/13/2022]
Abstract
Prep1 is a gene encoding for a homeodomain transcription factor which induces hepatic and muscular insulin resistance. In this study, we show that Prep1 hypomorphic heterozygous (Prep1i/+) mice, expressing low levels of protein, featured a 23% and a 25% reduction of total body lipid content and epididymal fat, respectively. The percentage of the small adipocytes (25-75 μm) was 30% higher in Prep1i/+ animals than in the WT, with a reciprocal difference in the large adipose cells (100-150 and >150 μm). Insulin-stimulated insulin receptor tyrosine and Akt serine phosphorylation markedly increased in Prep1i/+ mice, paralleled by 3-fold higher glucose uptake and a significant increase of proadipogenic genes such as C/EBPα, GLUT4, and FABP4. Moreover, T cells infiltration and TNF-α, IFNγ and leptin expression were reduced in adipose tissue from Prep1i/+ mice, while adiponectin levels were 2-fold higher. Furthermore, Prep1i/+ mature adipocytes released lower amounts of pro-inflammatory cytokines and higher amount of adiponectin compared to WT cells. Incubation of murine liver cell line (NMuLi) with conditioned media (CM) from mature adipocytes of Prep1i/+ mice improved glucose metabolism, while those from WT mice had no effect. Consistent with these data, Prep1 overexpression in 3T3-L1 adipocytes impaired adipogenesis and insulin signaling, and increased proinflammatory cytokine secretion. All these findings suggest that Prep1 silencing reduces inflammatory response and increases insulin sensitivity in adipose tissue. In addition, CM from mature adipocytes of Prep1i/+ mice improve metabolism in hepatic cells.
Collapse
Affiliation(s)
- Antonietta Liotti
- Department of Translational Medicine, Federico II University of Naples and URT "Genomic of Diabetes" of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Naples, Italy
| | - Serena Cabaro
- Department of Translational Medicine, Federico II University of Naples and URT "Genomic of Diabetes" of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Naples, Italy
| | - Ilaria Cimmino
- Department of Translational Medicine, Federico II University of Naples and URT "Genomic of Diabetes" of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Naples, Italy
| | - Serena Ricci
- Department of Translational Medicine, Federico II University of Naples and URT "Genomic of Diabetes" of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Naples, Italy
| | - Claudio Procaccini
- Laboratory of Immunology, National Council of Research (CNR), Naples, Italy
| | - Orlando Paciello
- Department of Veterinary Medicine and Animal Production, Federico II University of Naples, Naples, Italy
| | - Gregory A Raciti
- Department of Translational Medicine, Federico II University of Naples and URT "Genomic of Diabetes" of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Naples, Italy
| | - Rosa Spinelli
- Department of Translational Medicine, Federico II University of Naples and URT "Genomic of Diabetes" of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Naples, Italy
| | - Susanna Iossa
- Department of Biology, Federico II University of Naples, Naples, Italy
| | - Giuseppe Matarese
- Laboratory of Immunology, National Council of Research (CNR), Naples, Italy
| | - Claudia Miele
- Department of Translational Medicine, Federico II University of Naples and URT "Genomic of Diabetes" of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Naples, Italy
| | - Pietro Formisano
- Department of Translational Medicine, Federico II University of Naples and URT "Genomic of Diabetes" of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Naples, Italy
| | - Francesco Beguinot
- Department of Translational Medicine, Federico II University of Naples and URT "Genomic of Diabetes" of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Naples, Italy
| | - Francesco Oriente
- Department of Translational Medicine, Federico II University of Naples and URT "Genomic of Diabetes" of Institute of Experimental Endocrinology and Oncology, National Council of Research (CNR), Naples, Italy.
| |
Collapse
|
44
|
Jin A, Lei CX, Tian JJ, Sun J, Ji H. Dietary docosahexaenoic acid decreased lipid accumulation via inducing adipocytes apoptosis of grass carp, Ctenopharygodon idella. FISH PHYSIOLOGY AND BIOCHEMISTRY 2018; 44:197-207. [PMID: 28918543 DOI: 10.1007/s10695-017-0424-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 09/04/2017] [Indexed: 06/07/2023]
Abstract
The purpose of this study was to explore the mechanism of by which docosahexaenoic acid (DHA) inhibit the accumulation of adipose tissue lipid in grass carp (Ctenopharyngodon idella). We therefore designed two semi-purified diets, namely DHA-free (control) and DHA-supplemented, and fed them to grass carp (22.19 ± 1.76 g) for 3 and 6 weeks. DHA supplementation led to a significantly lower intraperitoneal fat index (IPFI) than that in the control group by reducing the number of adipocytes but significantly higher adipocyte size (P < 0.05). In the intraperitoneal adipose tissue, the DHA-fed group showed significantly higher peroxisome proliferator-activated receptor (PPAR)γ, CCAAT enhancer-binding protein (C/EBP)α, and sterol regulatory element-binding protein (SREBP)1c mRNA expression levels at both 3 and 6 weeks (P < 0.05). However, the ratio of the expression levels of B cell leukemia 2 (Bcl-2) and Bcl-2-associated X protein (Bax) was significantly lower in the DHA-fed group than in the control group (P < 0.05), and the protein expression levels of the apoptosis-related proteins caspase 3, caspase 8, and caspase 9 were also significantly higher (P < 0.05). Overall, although DHA promotes lipid synthesis, it is more likely that DHA could suppress the lipid accumulation in adipocytes of grass carp by inducing adipocyte apoptosis.
Collapse
Affiliation(s)
- Ai Jin
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, People's Republic of China
| | - Cai-Xia Lei
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, People's Republic of China
| | - Jing-Jing Tian
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, People's Republic of China
| | - Jian Sun
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, People's Republic of China
| | - Hong Ji
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, People's Republic of China.
| |
Collapse
|
45
|
Liu P, Tian JJ, Ji H, Sun J, Li C, Huang JQ, Li Y, Yu HB, Yu EM, Xie J. The Wnt/β-catenin pathway contributes to the regulation of adipocyte development induced by docosahexaenoic acid in grass carp, Ctenopharyngodon idellus. Comp Biochem Physiol B Biochem Mol Biol 2018; 216:18-24. [DOI: 10.1016/j.cbpb.2017.11.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 08/20/2017] [Accepted: 11/14/2017] [Indexed: 12/16/2022]
|
46
|
Li J, Curley JL, Floyd ZE, Wu X, Halvorsen YDC, Gimble JM. Isolation of Human Adipose-Derived Stem Cells from Lipoaspirates. Methods Mol Biol 2018; 1773:155-165. [PMID: 29687388 DOI: 10.1007/978-1-4939-7799-4_13] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Adipose tissue is as an abundant and accessible source of stem cells with multipotent properties suitable for tissue engineering and regenerative medical applications. Here, we describe methods from our own laboratory and the literature for the isolation and expansion of adipose-derived stem cells (ASCs). We present a large scale procedure suitable for processing >100 mL volumes of lipoaspirate tissue specimens by collagenase digestion and a related procedure suitable for processing adipose tissue aspirates without digestion.
Collapse
Affiliation(s)
- Jie Li
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA, USA
- LaCell LLC, New Orleans, LA, USA
| | - J Lowry Curley
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA, USA
- LaCell LLC, New Orleans, LA, USA
| | - Z Elizabeth Floyd
- Ubiquitin Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA
| | - Xiying Wu
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA, USA
- LaCell LLC, New Orleans, LA, USA
| | - Yuan Di C Halvorsen
- Center for Computational and Integrative Biology, Harvard Medical School, Boston, MA, USA
| | - Jeffrey M Gimble
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA, USA.
- LaCell LLC, New Orleans, LA, USA.
| |
Collapse
|
47
|
Zhai X, Cheng F, Ji L, Zhu X, Cao Q, Zhang Y, Jia X, Zhou Q, Guan W, Zhou Y. Leptin reduces microRNA-122 level in hepatic stellate cells in vitro and in vivo. Mol Immunol 2017; 92:68-75. [PMID: 29054053 DOI: 10.1016/j.molimm.2017.10.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 10/09/2017] [Accepted: 10/09/2017] [Indexed: 12/13/2022]
Abstract
Obese patients, often accompanied by hyperleptinemia, are more likely to develop liver fibrosis. Leptin, an adipocyte-derived hormone, augments inflammatory in liver and promotes hepatic stellate cell (HSC) activation (a key step for liver fibrogenesis) and liver fibrosis. microRNA-122 (miR-122) is the most abundant liver-specific miRNA and can attenuate liver fibrosis. This study examined the effect of leptin on miR-122 level in HSCs in vivo and in vitro. Results demonstrated that leptin reduced the levels of both miR-122 (mature miR-122) and primary miR-122 (pri-miR-122). The effects of leptin on the levels of miR-122 and pri-miR-122 were through at least hedgehog pathway. Leptin-induced decrease in sterol regulatory element-binding protein-1c (SREBP-1c) has been shown to contribute to leptin-induced HSC activation. We revealed a mutual promotional effect between SREBP-1c and miR-122. Further experiments indicated that miR-122 inhibited leptin-induced liver fibrosis in leptin-deficient mouse model. These data have potential implications for clarifying the mechanisms of hepatic fibrogenesis associated with elevated leptin level in human such as obese patients.
Collapse
Affiliation(s)
- Xuguang Zhai
- Department of Biochemistry & Molecular Biology, Medical College, Nantong University, Qi xiou road 19, Nantong 226001, Jiangsu, China
| | - Fangyun Cheng
- Department of Biochemistry & Molecular Biology, Medical College, Nantong University, Qi xiou road 19, Nantong 226001, Jiangsu, China
| | - Li Ji
- Department of Pharmacology, School of Pharmacy, Nantong University, Qi xiou road 19, Nantong 226001, Jiangsu, China
| | - Xiaofei Zhu
- Department of Pharmacology, School of Pharmacy, Nantong University, Qi xiou road 19, Nantong 226001, Jiangsu, China
| | - Qing Cao
- Department of Pharmacology, School of Pharmacy, Nantong University, Qi xiou road 19, Nantong 226001, Jiangsu, China
| | - Yali Zhang
- Department of Biochemistry & Molecular Biology, Medical College, Nantong University, Qi xiou road 19, Nantong 226001, Jiangsu, China
| | - Xin Jia
- Department of Biochemistry & Molecular Biology, Medical College, Nantong University, Qi xiou road 19, Nantong 226001, Jiangsu, China
| | - Qian Zhou
- Department of Pharmacology, School of Pharmacy, Nantong University, Qi xiou road 19, Nantong 226001, Jiangsu, China
| | - Wei Guan
- Department of Biochemistry & Molecular Biology, Medical College, Nantong University, Qi xiou road 19, Nantong 226001, Jiangsu, China
| | - Yajun Zhou
- Department of Biochemistry & Molecular Biology, Medical College, Nantong University, Qi xiou road 19, Nantong 226001, Jiangsu, China.
| |
Collapse
|
48
|
Adipose Tissue Function and Expandability as Determinants of Lipotoxicity and the Metabolic Syndrome. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 960:161-196. [PMID: 28585199 DOI: 10.1007/978-3-319-48382-5_7] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The adipose tissue organ is organised as distinct anatomical depots located all along the body axis and it is constituted of three different types of adipocytes : white, beige and brown which are integrated with vascular, immune, neural and extracellular stroma cells. These distinct adipocytes serve different specialised functions. The main function of white adipocytes is to ensure healthy storage of excess nutrients/energy and its rapid mobilisation to supply the demand of energy imposed by physiological cues in other organs, whereas brown and beige adipocytes are designed for heat production through uncoupling lipid oxidation from energy production. The concert action of the three type of adipocytes/tissues has been reported to ensure an optimal metabolic status in rodents. However, when one or multiple of these adipose depots become dysfunctional as a consequence of sustained lipid/nutrient overload, then insulin resistance and associated metabolic complications ensue. These metabolic alterations negatively affects the adipose tissue functionality and compromises global metabolic homeostasis. Optimising white adipose tissue expandability and its functional metabolic flexibility and/or promoting brown/beige mediated thermogenic activity counteracts obesity and its associated lipotoxic metabolic effects. The development of these therapeutic approaches requires a deep understanding of adipose tissue in all broad aspects. In this chapter we will discuss the characteristics of the different adipose tissue depots with respect to origins and precursors recruitment, plasticity, cellular composition and expandability capacity as well as molecular and metabolic signatures in both physiological and pathophysiological conditions.
Collapse
|
49
|
Xu Y, Hadjiargyrou M, Rafailovich M, Mironava T. Cell-based cytotoxicity assays for engineered nanomaterials safety screening: exposure of adipose derived stromal cells to titanium dioxide nanoparticles. J Nanobiotechnology 2017; 15:50. [PMID: 28693576 PMCID: PMC5504822 DOI: 10.1186/s12951-017-0285-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 07/03/2017] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Increasing production of nanomaterials requires fast and proper assessment of its potential toxicity. Therefore, there is a need to develop new assays that can be performed in vitro, be cost effective, and allow faster screening of engineered nanomaterials (ENMs). RESULTS Herein, we report that titanium dioxide (TiO2) nanoparticles (NPs) can induce damage to adipose derived stromal cells (ADSCs) at concentrations which are rated as safe by standard assays such as measuring proliferation, reactive oxygen species (ROS), and lactate dehydrogenase (LDH) levels. Specifically, we demonstrated that low concentrations of TiO2 NPs, at which cellular LDH, ROS, or proliferation profiles were not affected, induced changes in the ADSCs secretory function and differentiation capability. These two functions are essential for ADSCs in wound healing, energy expenditure, and metabolism with serious health implications in vivo. CONCLUSIONS We demonstrated that cytotoxicity assays based on specialized cell functions exhibit greater sensitivity and reveal damage induced by ENMs that was not otherwise detected by traditional ROS, LDH, and proliferation assays. For proper toxicological assessment of ENMs standard ROS, LDH, and proliferation assays should be combined with assays that investigate cellular functions relevant to the specific cell type.
Collapse
Affiliation(s)
- Yan Xu
- Department of Materials Science and Engineering, Stony Brook University, Stony Brook, NY USA
| | - M. Hadjiargyrou
- Department of Life Sciences, New York Institute of Technology, Old Westbury, NY USA
| | - Miriam Rafailovich
- Department of Materials Science and Engineering, Stony Brook University, Stony Brook, NY USA
| | - Tatsiana Mironava
- Department of Materials Science and Engineering, Stony Brook University, Stony Brook, NY USA
| |
Collapse
|
50
|
Cho BY, Park MR, Lee JH, Ra MJ, Han KC, Kang IJ, Lee OH. Standardized Cirsium setidens Nakai Ethanolic Extract Suppresses Adipogenesis and Regulates Lipid Metabolisms in 3T3-L1 Adipocytes and C57BL/6J Mice Fed High-Fat Diets. J Med Food 2017; 20:763-776. [PMID: 28686516 DOI: 10.1089/jmf.2017.3965] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Cirsium setidens Nakai, a wild perennial herb, grows mainly in Gangwon province, Korea, and has been reported to contain bioactive ingredients with various medicinal activities, including the treatment of edema, bleeding, and hemoptysis. However, the potential antiobesity effects of C. setidens Nakai have not been fully investigated. This study evaluated the antiobesity effect of standardized C. setidens Nakai ethanolic extract (CNE) in 3T3-L1 adipocytes and in obese C57BL/6J mice fed a high-fat diet. CNE suppressed the expression of lipogenic genes and increased the expression of lipolytic genes. The antiadipogenic and antilipogenic effects of CNE appear to be mediated by the inhibition of peroxisome proliferator-activated receptor γ (PPARγ) and CCAAT/enhancer-binding protein (C/EBP) expressions. Moreover, CNE stimulated fatty acid oxidation in an AMPK-dependent manner. CNE-treated groups of C57BL/6J mice showed reduced body weights and adipose tissue weight and improved serum lipid profiles through the downregulation of PPARγ, C/EBPα, fatty acid binding protein 4 (FABP4), sterol regulatory element binding protein-1c (SREBP-1c), and fatty acid synthase (FAS) and the upregulation of adiponectin and carnitine palmitoyltransferase-1 (CPT-1) in obese C57BL/6J mice fed a high-fat diet. These results suggest that CNE may have an antiobesity effect on adipogenesis and lipid metabolism in vitro and in vivo and present the possibility of developing a treatment for obesity with nontoxic natural resources.
Collapse
Affiliation(s)
- Bong-Yeon Cho
- 1 Department of Food Science and Biotechnology, Kangwon National University , Chuncheon, Republic of Korea
| | - Mi-Ryeong Park
- 2 Department of Food Science and Nutrition, Hallym University , Chuncheon, Republic of Korea
| | - Jin-Ha Lee
- 1 Department of Food Science and Biotechnology, Kangwon National University , Chuncheon, Republic of Korea
| | - Moon-Jin Ra
- 3 Hongcheon Institute of Medicinal Herb , Hongcheon, Republic of Korea
| | | | - Il-Jun Kang
- 2 Department of Food Science and Nutrition, Hallym University , Chuncheon, Republic of Korea
| | - Ok-Hwan Lee
- 1 Department of Food Science and Biotechnology, Kangwon National University , Chuncheon, Republic of Korea
| |
Collapse
|