251
|
Ahn YA, Baek H, Choi M, Park J, Son SJ, Seo HJ, Jung J, Seong JK, Lee J, Kim S. Adipogenic effects of prenatal exposure to bisphenol S (BPS) in adult F1 male mice. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 728:138759. [PMID: 32403013 DOI: 10.1016/j.scitotenv.2020.138759] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 04/13/2020] [Accepted: 04/15/2020] [Indexed: 05/20/2023]
Abstract
Bisphenol S (BPS) has been increasingly used as a substitute for bisphenol A (BPA), a known endocrine disruptor. Early-life exposure to BPA affects fetal development and the risk of obesity in adolescence and adulthood. However, the effects of fetal exposure BPS in later life are unknown. This study aimed to investigate the effects of prenatal BPS exposure on adiposity in adult F1 mice. Pregnant C57BL/6 N mice were exposed to BPS (0, 0.05, 0.5, 5, and 50 mg/kg/d) via drinking water from gestation day 9 until delivery. Thereafter, two groups of offspring (6 weeks old) were either administered a standard diet (STD) or a high-fat diet (HFD) for 4 weeks until euthanasia. The body weight and gonadal white adipose tissue (gWAT) mass were determined, and the energy expenditure for the adiposity phenotype was computed especially for male mice, followed by histological analysis of the gWAT. Thereafter, the expression levels of adipogenic marker genes (Pparg, Cebpa, Fabp4, Lpl, and Adipoq) were analyzed in the gWAT via reverse-transcription PCR analysis. BPS-exposed male mice displayed apparent gWAT hypertrophy, consistent with the significant increase in adipocyte size in the gWAT and upregulation of Pparg and its direct target genes among HFD mice in comparison with the control mice. These results suggest that prenatal BPS exposure potentially increases the susceptibility to HFD-induced adipogenesis in male adult mice.
Collapse
Affiliation(s)
- Young-Ah Ahn
- Department of Environmental Health Sciences, Graduate School of Public Health, Seoul National University, Seoul 08826, Republic of Korea.
| | - Hwayoung Baek
- Department of Environmental Health Sciences, Graduate School of Public Health, Seoul National University, Seoul 08826, Republic of Korea.
| | - Miso Choi
- Department of Environmental Health Sciences, Graduate School of Public Health, Seoul National University, Seoul 08826, Republic of Korea.
| | - Junbo Park
- Department of Environmental Health Sciences, Graduate School of Public Health, Seoul National University, Seoul 08826, Republic of Korea.
| | - Soo Jin Son
- Laboratory of Developmental Biology and Genomics, Research Institute for Veterinary Science, and BK21 Program for Advanced Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea; Korea Mouse Phenotyping Center, Seoul National University, Seoul 08826, Republic of Korea.
| | - Hyun Ju Seo
- Laboratory of Developmental Biology and Genomics, Research Institute for Veterinary Science, and BK21 Program for Advanced Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea; Korea Mouse Phenotyping Center, Seoul National University, Seoul 08826, Republic of Korea.
| | - Jaeyun Jung
- Laboratory of Developmental Biology and Genomics, Research Institute for Veterinary Science, and BK21 Program for Advanced Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea; Korea Mouse Phenotyping Center, Seoul National University, Seoul 08826, Republic of Korea.
| | - Je Kyung Seong
- Laboratory of Developmental Biology and Genomics, Research Institute for Veterinary Science, and BK21 Program for Advanced Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea; Korea Mouse Phenotyping Center, Seoul National University, Seoul 08826, Republic of Korea.
| | - Jaehyouk Lee
- Department of Environmental Health Sciences, Graduate School of Public Health, Seoul National University, Seoul 08826, Republic of Korea.
| | - Sungkyoon Kim
- Department of Environmental Health Sciences, Graduate School of Public Health, Seoul National University, Seoul 08826, Republic of Korea; Institute of Health and Environment, Graduate School of Public Health, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea.
| |
Collapse
|
252
|
Od-Ek P, Deenin W, Malakul W, Phoungpetchara I, Tunsophon S. Anti-obesity effect of Carica papaya in high-fat diet fed rats. Biomed Rep 2020; 13:30. [PMID: 32802327 PMCID: PMC7412661 DOI: 10.3892/br.2020.1337] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 07/01/2020] [Indexed: 12/14/2022] Open
Abstract
The present study evaluated the anti-obesity properties of papaya in high-fat (HF) diet fed rats. In the in vitro portion of the present study, the effects of papaya juice on pancreatic lipase enzyme activity was assessed, and it was shown that papaya exhibited an inhibitory effect on these enzymes. In the in vivo portion of the study, papaya was found to reduce the expression levels of markers of obesity, inflammation and oxidative stress in rats. Obesity was induced in 28 male Sprague Dawley rats by feeding them a HF diet for 12 weeks. The anti-obesity effects of papaya was evaluated by feeding papaya juice orally in with two experimental doses: 0.5 ml (HFL) and 1.0 ml (HFH) per 100 g of body weight. The HF diet resulted in significant increases in the body weight, serum triglyceride, serum total cholesterol and serum low-density lipoprotein cholesterol levels, as well as a decrease in serum high-density lipoprotein cholesterol levels. The HF diet also induced adipocyte hypertrophy, lipid accumulation and increased malondialdehyde levels. Papaya reversed all of these changes and significantly increased serum superoxide dismutase and decreased serum cytokine (interleukin-6) levels. The protein expression of levels PPARγ in the HF group was significantly increased compared with the other groups, but was decreased significantly in the HFH group. Histological observations of epididymal adipose tissue provided evidence for the lipid-lowering effects of papaya. The results of the present study demonstrate that papaya has the potential to reduce the risk of obesity associated with adiposity, anti-inflammation and anti-oxidation.
Collapse
Affiliation(s)
- Phichittra Od-Ek
- Department of Physiology, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand
| | - Wanwisa Deenin
- 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
| | - Ittipon Phoungpetchara
- Department of Anatomy, 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
|
253
|
Soundharrajan I, Kuppusamy P, Srisesharam S, Lee JC, Sivanesan R, Kim D, Choi KC. Positive metabolic effects of selected probiotic bacteria on diet‐induced obesity in mice are associated with improvement of dysbiotic gut microbiota. FASEB J 2020; 34:12289-12307. [DOI: 10.1096/fj.202000971r] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/26/2020] [Accepted: 06/30/2020] [Indexed: 12/21/2022]
Affiliation(s)
- Ilavenil Soundharrajan
- Grassland and Forage Division, National Institute of Animal Science Rural Development Administration Cheonan Republic of Korea
| | - Palaniselvam Kuppusamy
- Grassland and Forage Division, National Institute of Animal Science Rural Development Administration Cheonan Republic of Korea
| | - Srigopalram Srisesharam
- Grassland and Forage Division, National Institute of Animal Science Rural Development Administration Cheonan Republic of Korea
| | - Jeong Chae Lee
- Research Center of Bioactive Materials, Institute of Molecular Biology and Genetics Chonbuk National University Jeonju Republic of Korea
| | - Ravikumar Sivanesan
- Department of Zoology Rajah Serfoji Government Arts College Thanjavur Tamilnadu India
| | - Dahye Kim
- Faculty of Biotechnology College of Applied Life Science Jeju National University Jeju Republic of Korea
| | - Ki Choon Choi
- Grassland and Forage Division, National Institute of Animal Science Rural Development Administration Cheonan Republic of Korea
| |
Collapse
|
254
|
Ezzati-Mobaser S, Malekpour-Dehkordi Z, Nourbakhsh M, Tavakoli-Yaraki M, Ahmadpour F, Golpour P, Nourbakhsh M. The up-regulation of markers of adipose tissue fibrosis by visfatin in pre-adipocytes as well as obese children and adolescents. Cytokine 2020; 134:155193. [PMID: 32707422 DOI: 10.1016/j.cyto.2020.155193] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 06/25/2020] [Accepted: 07/04/2020] [Indexed: 01/01/2023]
Abstract
Adipocytes are surrounded by a three-dimensional network of extracellular matrix (ECM) proteins. Aberrant ECM accumulation and remodeling leads to adipose tissue fibrosis. Visfatin is one of the adipocytokines that is increased in obesity and is implicated in insulin resistance. The objective of this study was to investigate the effect of visfatin on major components of ECM remodeling. In this study, plasma levels of both endotrophin and visfatin in obese children and adolescents were significantly higher than those in control subjects and they showed a positive correlation with each other. Treatment of 3T3-L1 pre-adipocytes with visfatin caused significant up-regulation of Osteopontin (Opn), Collagen type VI (Col6), matrix metalloproteinases MMP-2 and MMP-9. By using inhibitors of major signaling pathways it was shown that visfatin exerted its effect on Col6a3 gene expression through PI3K, JNK, and NF-кB pathways, while induced Opn gene expression via PI3K, JNK, MAPK/ERK, and NOTCH1. Our conclusion is that, the relationship between visfatin, endotrophin and insulin resistance parameters in obesity as well as increased expression of ECM proteins by visfatin suggests adipose tissue fibrosis as a mechanism for devastating effects of visfatin in obesity.
Collapse
Affiliation(s)
- Samira Ezzati-Mobaser
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Malekpour-Dehkordi
- Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular -Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mona Nourbakhsh
- Hazrat Aliasghar Children's Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Masoumeh Tavakoli-Yaraki
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Ahmadpour
- Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Pegah Golpour
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mitra Nourbakhsh
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran; Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
255
|
Lynch GM, Murphy CH, Castro EDM, Roche HM. Inflammation and metabolism: the role of adiposity in sarcopenic obesity. Proc Nutr Soc 2020; 79:1-13. [PMID: 32669148 DOI: 10.1017/s0029665120007119] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Sarcopenic obesity is characterised by the double burden of diminished skeletal muscle mass and the presence of excess adiposity. From a mechanistic perspective, both obesity and sarcopenia are associated with sub-acute, chronic pro-inflammatory states that impede metabolic processes, disrupting adipose and skeletal functionality, which may potentiate disease. Recent evidence suggests that there is an important cross-talk between metabolism and inflammation, which has shifted focus upon metabolic-inflammation as a key emerging biological interaction. Dietary intake, physical activity and nutritional status are important environmental factors that may modulate metabolic-inflammation. This paradigm will be discussed within the context of sarcopenic obesity risk. There is a paucity of data in relation to the nature and the extent to which nutritional status affects metabolic-inflammation in sarcopenic obesity. Research suggests that there may be scope for the modulation of sarcopenic obesity with alterations in diet. The potential impact of increasing protein consumption and reconfiguration of dietary fat composition in human dietary interventions are evaluated. This review will explore emerging data with respect to if and how different dietary components may modulate metabolic-inflammation, particularly with respect to adiposity, within the context of sarcopenic obesity.
Collapse
Affiliation(s)
- G M Lynch
- Nutrigenomics Research Group, School of Public Health, Physiotherapy and Sports Science, UCD Institute of Food and Health, Diabetes Complications Research Centre, University College Dublin, Dublin, Ireland
| | - C H Murphy
- Nutrigenomics Research Group, School of Public Health, Physiotherapy and Sports Science, UCD Institute of Food and Health, Diabetes Complications Research Centre, University College Dublin, Dublin, Ireland
| | - E de Marco Castro
- Nutrigenomics Research Group, School of Public Health, Physiotherapy and Sports Science, UCD Institute of Food and Health, Diabetes Complications Research Centre, University College Dublin, Dublin, Ireland
| | - H M Roche
- Nutrigenomics Research Group, School of Public Health, Physiotherapy and Sports Science, UCD Institute of Food and Health, Diabetes Complications Research Centre, University College Dublin, Dublin, Ireland
- Institute for Global Food Security, Queen's University Belfast, Belfast, UK
| |
Collapse
|
256
|
Tian N, Liu Q, Li Y, Tong L, Lu Y, Zhu Y, Zhang P, Chen H, Hu L, Meng J, Feng M, Li M, Zheng L, Li B, Xu T, Wu L, Tong X. Transketolase Deficiency in Adipose Tissues Protects Mice From Diet-Induced Obesity by Promoting Lipolysis. Diabetes 2020; 69:1355-1367. [PMID: 32295803 DOI: 10.2337/db19-1087] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 04/09/2020] [Indexed: 01/06/2023]
Abstract
Obesity has recently become a prevalent health threat worldwide. Although emerging evidence has suggested a strong link between the pentose phosphate pathway (PPP) and obesity, the role of transketolase (TKT), an enzyme in the nonoxidative branch of the PPP that connects PPP and glycolysis, remains obscure in adipose tissues. In this study, we specifically deleted TKT in mouse adipocytes and found no obvious phenotype upon normal diet feeding. However, adipocyte TKT abrogation attenuated high-fat diet-induced obesity, reduced hepatic steatosis, improved glucose tolerance, alleviated insulin resistance, and increased energy expenditure. Mechanistically, TKT deficiency accumulated nonoxidative PPP metabolites and decreased glycolysis and pyruvate input into the mitochondria, leading to increased lipolytic enzyme gene expression and enhanced lipolysis, fatty acid oxidation, and mitochondrial respiration. Therefore, our data not only identify a novel role of TKT in regulating lipolysis and obesity but also suggest that limiting glucose-derived carbon into the mitochondria induces lipid catabolism and energy expenditure.
Collapse
Affiliation(s)
- Na Tian
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qi Liu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yakui Li
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lingfeng Tong
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Lu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yemin Zhu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ping Zhang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hanbei Chen
- Department of Endocrinology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Hu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian Meng
- School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Ming Feng
- School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Minle Li
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Liang Zheng
- Pediatric Translational Medicine Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bin Li
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tianle Xu
- Collaborative Innovation Center for Brain Science, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lifang Wu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xuemei Tong
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
257
|
A Bibliometric Review of Artificial Extracellular Matrices Based on Tissue Engineering Technology Literature: 1990 through 2019. MATERIALS 2020; 13:ma13132891. [PMID: 32605069 PMCID: PMC7372414 DOI: 10.3390/ma13132891] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/23/2020] [Accepted: 06/24/2020] [Indexed: 01/06/2023]
Abstract
Artificial extracellular matrices (aECMs) are an extension of biomaterials that were developed as in-vitro model environments for tissue cells that mimic the native in vivo target tissues’ structure. This bibliometric analysis evaluated the research productivity regarding aECM based on tissue engineering technology. The Web of Science citation index was examined for articles published from 1990 through 2019 using three distinct aECM-related topic sets. Data were also visualized using network analyses (VOSviewer). Terms related to in-vitro, scaffolds, collagen, hydrogels, and differentiation were reoccurring in the aECM-related literature over time. Publications with terms related to a clinical direction (wound healing, stem cells, artificial skin, in-vivo, and bone regeneration) have steadily increased, as have the number of countries and institutions involved in the artificial extracellular matrix. As progress with 3D scaffolds continues to advance, it will become the most promising technology to provide a therapeutic option to repair or replace damaged tissue.
Collapse
|
258
|
Park M, Han J, Lee HJ. Anti-Adipogenic Effect of Neferine in 3T3-L1 Cells and Primary White Adipocytes. Nutrients 2020; 12:nu12061858. [PMID: 32580414 PMCID: PMC7353287 DOI: 10.3390/nu12061858] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 06/11/2020] [Accepted: 06/17/2020] [Indexed: 02/06/2023] Open
Abstract
Neferine, an alkaloid component extracted from lotus seed embryos, is known for its anti-inflammatory, anticancer, and antioxidant properties. However, the anti-adipogenic activity of neferine has not been thoroughly investigated. In this study, neferine was found to inhibit lipid accumulation in a dose-dependent manner during the differentiation of 3T3-L1 cells without inducing cytotoxicity. Real-time polymerase chain reaction and immunoblot analysis revealed the downregulation in the expression of peroxisome proliferator activated receptor gamma (PPARγ), CCAAT/enhancer-binding protein alpha (C/EBPα), sterol regulatory element-binding protein-1c (SREBP-1c), and fatty acid synthase (FAS) and the upregulation in carnitine palmitoyltransferase-1 (CPT-1) and sirtuin 1 (SIRT1) levels following neferine treatment. Furthermore, neferine increased the phosphorylation of adenosine monophosphate-activated protein kinase (AMPK) and acetyl-CoA carboxylase (ACC), which is an important regulator of fatty acid oxidation. Our result indicates that neferine attenuates adipogenesis and promotes lipid metabolism by activating AMPK-mediated signaling. Therefore, neferine may serve as a therapeutic candidate for obesity treatment.
Collapse
Affiliation(s)
- Miey Park
- Department of Food and Nutrition, College of BioNano Technology, Gachon University, Gyeonggi-do 13120, Korea; (M.P.); (J.H.)
- Institute for Aging and Clinical Nutrition Research, Gachon University, Gyeonggi-do 13120, Korea
| | - Jinyoung Han
- Department of Food and Nutrition, College of BioNano Technology, Gachon University, Gyeonggi-do 13120, Korea; (M.P.); (J.H.)
| | - Hae-Jeung Lee
- Department of Food and Nutrition, College of BioNano Technology, Gachon University, Gyeonggi-do 13120, Korea; (M.P.); (J.H.)
- Institute for Aging and Clinical Nutrition Research, Gachon University, Gyeonggi-do 13120, Korea
- Correspondence: ; Tel.: +82-31-750-5968; Fax: +82-31-724-4411
| |
Collapse
|
259
|
Al Haj G, Rey F, Giallongo T, Colli M, Marzani B, Giuliani G, Gorio A, Zuccotti GV, Di Giulio AM, Carelli S. A New Selective PPARγ Modulator Inhibits Triglycerides Accumulation during Murine Adipocytes' and Human Adipose-Derived Mesenchymal Stem Cells Differentiation. Int J Mol Sci 2020; 21:ijms21124415. [PMID: 32575918 PMCID: PMC7352648 DOI: 10.3390/ijms21124415] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/08/2020] [Accepted: 06/18/2020] [Indexed: 12/19/2022] Open
Abstract
Understanding the molecular basis of adipogenesis is vital to identify new therapeutic targets to improve anti-obesity drugs. The adipogenic process could be a new target in the management of this disease. Our aim was to evaluate the effect of GMG-43AC, a selective peroxisome proliferator-activated receptor γ (PPARγ) modulator, during adipose differentiation of murine pre-adipocytes and human Adipose Derived Stem Cells (hADSCs). We differentiated 3T3-L1 cells and primary hADSCs in the presence of various doses of GMG-43AC and evaluated the differentiation efficiency measuring lipid accumulation, the expression of specific differentiation markers and the quantification of accumulated triglycerides. The treatment with GMG-43AC is not toxic as shown by cell viability assessments after the treatments. Our findings demonstrate the inhibition of lipid accumulation and the significant decrease in the expression of adipocyte-specific genes, such as PPARγ, FABP-4, and leptin. This effect was long lasting, as the removal of GMG-43AC from culture medium did not allow the restoration of adipogenic process. The above actions were confirmed in hADSCs exposed to adipogenic stimuli. Together, these results indicate that GMG-43AC efficiently inhibits adipocytes differentiation in murine and human cells, suggesting its possible function in the reversal of adipogenesis and modulation of lipolysis.
Collapse
Affiliation(s)
- Ghina Al Haj
- Department of Health Sciences, University of Milan, Via Antonio di Rudinì 8, 20142 Milan, Italy; (G.A.H.); (F.R.); (T.G.); (M.C.); (A.G.)
| | - Federica Rey
- Department of Health Sciences, University of Milan, Via Antonio di Rudinì 8, 20142 Milan, Italy; (G.A.H.); (F.R.); (T.G.); (M.C.); (A.G.)
| | - Toniella Giallongo
- Department of Health Sciences, University of Milan, Via Antonio di Rudinì 8, 20142 Milan, Italy; (G.A.H.); (F.R.); (T.G.); (M.C.); (A.G.)
| | - Mattia Colli
- Department of Health Sciences, University of Milan, Via Antonio di Rudinì 8, 20142 Milan, Italy; (G.A.H.); (F.R.); (T.G.); (M.C.); (A.G.)
| | - Barbara Marzani
- Research and Development, Giuliani SpA, Via Pelagio Palagi, 2, 20129 Milan, Italy; (B.M.); (G.G.)
| | - Giammaria Giuliani
- Research and Development, Giuliani SpA, Via Pelagio Palagi, 2, 20129 Milan, Italy; (B.M.); (G.G.)
| | - Alfredo Gorio
- Department of Health Sciences, University of Milan, Via Antonio di Rudinì 8, 20142 Milan, Italy; (G.A.H.); (F.R.); (T.G.); (M.C.); (A.G.)
| | - Gian Vicenzo Zuccotti
- Department of Biomedical and Clinical Sciences, University of Milan, Via G.B. Grassi 74, 20157 Milan, Italy;
- Pediatric Research Center “Romeo ed Enrica Invernizzi”, University of Milan, Via G.B. Grassi 74, 20157 Milan, Italy
| | - Anna Maria Di Giulio
- Pediatric Research Center “Romeo ed Enrica Invernizzi”, University of Milan, Via G.B. Grassi 74, 20157 Milan, Italy
- Correspondence: (A.M.D.G.); (S.C.)
| | - Stephana Carelli
- Department of Biomedical and Clinical Sciences, University of Milan, Via G.B. Grassi 74, 20157 Milan, Italy;
- Pediatric Research Center “Romeo ed Enrica Invernizzi”, University of Milan, Via G.B. Grassi 74, 20157 Milan, Italy
- Correspondence: (A.M.D.G.); (S.C.)
| |
Collapse
|
260
|
Prediction of the Secretome and the Surfaceome: A Strategy to Decipher the Crosstalk between Adipose Tissue and Muscle during Fetal Growth. Int J Mol Sci 2020; 21:ijms21124375. [PMID: 32575512 PMCID: PMC7353064 DOI: 10.3390/ijms21124375] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/08/2020] [Accepted: 06/16/2020] [Indexed: 12/16/2022] Open
Abstract
Crosstalk between adipose and muscular tissues is hypothesized to regulate the number of muscular and adipose cells during fetal growth, with post-natal consequences on lean and fat masses. Such crosstalk largely remains, however, to be described. We hypothesized that a characterization of the proteomes of adipose and muscular tissues from bovine fetuses may enhance the understanding of the crosstalk between these tissues through the prediction of their secretomes and surfaceomes. Proteomic experiments have identified 751 and 514 proteins in fetal adipose tissue and muscle. These are mainly involved in the regulation of cell proliferation or differentiation, but also in pathways such as apoptosis, Wnt signalling, or cytokine-mediated signalling. Of the identified proteins, 51 adipokines, 11 myokines, and 37 adipomyokines were predicted, together with 26 adipose and 13 muscular cell surface proteins. Analysis of protein–protein interactions suggested 13 links between secreted and cell surface proteins that may contribute to the adipose–muscular crosstalk. Of these, an interaction between the adipokine plasminogen and the muscular cell surface alpha-enolase may regulate the fetal myogenesis. The in silico secretome and surfaceome analyzed herein exemplify a powerful strategy to enhance the elucidation of the crosstalk between cell types or tissues.
Collapse
|
261
|
Xu HY, Shao J, Yin BZ, Zhang LM, Fang JC, Zhang JS, Xia GJ. Bovine bta-microRNA-1271 Promotes Preadipocyte Differentiation by Targeting Activation Transcription Factor 3. BIOCHEMISTRY (MOSCOW) 2020; 85:749-757. [DOI: 10.1134/s0006297920070032] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
262
|
Qiu Y, Yu H, Zeng R, Guo S, Daniyal M, Deng Z, Wang A, Wang W. Recent Development on Anti-Obesity Compounds and their Mechanisms of Action: A Review. Curr Med Chem 2020; 27:3577-3597. [DOI: 10.2174/0929867326666190215114359] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 12/13/2018] [Accepted: 12/14/2018] [Indexed: 12/18/2022]
Abstract
Obesity, associated with a series of complications such as diabetes, hypertension, and
heart disease, is a great threat to human health and leads to increased morbidity and mortality. Despite
the presence of anti-obesity agents on the market, the application of these drugs is limited because
of their typical side effects. More effective and safe weight-loss drugs are being pursued by
many researchers, correspondingly, growing small molecules and natural products with anti-obesity
effects have been identified and the molecular mechanisms underlying the action of the novel and
known compounds have at least partially been revealed. Therefore, the field does witness great progress
year by year. In this review, we intend to provide a comprehensive and updated view on the
known and novel compounds which possess anti-obesity effects and further classify them according
to the molecular mechanisms of their actions in regulating the major anti-obesity pathways.
Collapse
Affiliation(s)
- Yixing Qiu
- TCM and Ethnomedicine Innovation & Development International Laboratory, Innovative Drug Research Institute, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Huanghe Yu
- TCM and Ethnomedicine Innovation & Development International Laboratory, Innovative Drug Research Institute, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Rong Zeng
- TCM and Ethnomedicine Innovation & Development International Laboratory, Innovative Drug Research Institute, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Shiyin Guo
- College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, Hunan, China
| | - Muhammad Daniyal
- TCM and Ethnomedicine Innovation & Development International Laboratory, Innovative Drug Research Institute, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Zeyu Deng
- TCM and Ethnomedicine Innovation & Development International Laboratory, Innovative Drug Research Institute, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Aibing Wang
- The Key Laboratory of Animal Vaccine & Protein Engineering, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, Hunan, China
| | - Wei Wang
- TCM and Ethnomedicine Innovation & Development International Laboratory, Innovative Drug Research Institute, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| |
Collapse
|
263
|
The Role of Pref-1 during Adipogenic Differentiation: An Overview of Suggested Mechanisms. Int J Mol Sci 2020; 21:ijms21114104. [PMID: 32526833 PMCID: PMC7312882 DOI: 10.3390/ijms21114104] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/25/2020] [Accepted: 05/30/2020] [Indexed: 12/15/2022] Open
Abstract
Obesity contributes significantly to the global health burden. A better understanding of adipogenesis, the process of fat formation, may lead to the discovery of novel treatment strategies. However, it is of concern that the regulation of adipocyte differentiation has predominantly been studied using the murine 3T3-L1 preadipocyte cell line and murine experimental animal models. Translation of these findings to the human setting requires confirmation using experimental models of human origin. The ability of mesenchymal stromal/stem cells (MSCs) to differentiate into adipocytes is an attractive model to study adipogenesis in vitro. Differences in the ability of MSCs isolated from different sources to undergo adipogenic differentiation, may be useful in investigating elements responsible for regulating adipogenic differentiation potential. Genes involved may be divided into three broad categories: early, intermediate and late-stage regulators. Preadipocyte factor-1 (Pref-1) is an early negative regulator of adipogenic differentiation. In this review, we briefly discuss the adipogenic differentiation potential of MSCs derived from two different sources, namely adipose-derived stromal/stem cells (ASCs) and Wharton’s Jelly derived stromal/stem cells (WJSCs). We then discuss the function and suggested mechanisms of action of Pref-1 in regulating adipogenesis, as well as current findings regarding Pref-1’s role in human adipogenesis.
Collapse
|
264
|
Harms MJ, Li Q, Lee S, Zhang C, Kull B, Hallen S, Thorell A, Alexandersson I, Hagberg CE, Peng XR, Mardinoglu A, Spalding KL, Boucher J. Mature Human White Adipocytes Cultured under Membranes Maintain Identity, Function, and Can Transdifferentiate into Brown-like Adipocytes. Cell Rep 2020; 27:213-225.e5. [PMID: 30943403 DOI: 10.1016/j.celrep.2019.03.026] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 01/23/2019] [Accepted: 03/06/2019] [Indexed: 12/24/2022] Open
Abstract
White adipose tissue (WAT) is a central factor in the development of type 2 diabetes, but there is a paucity of translational models to study mature adipocytes. We describe a method for the culture of mature white adipocytes under a permeable membrane. Compared to existing culture methods, MAAC (membrane mature adipocyte aggregate cultures) better maintain adipogenic gene expression, do not dedifferentiate, display reduced hypoxia, and remain functional after long-term culture. Subcutaneous and visceral adipocytes cultured as MAAC retain depot-specific gene expression, and adipocytes from both lean and obese patients can be cultured. Importantly, we show that rosiglitazone treatment or PGC1α overexpression in mature white adipocytes induces a brown fat transcriptional program, providing direct evidence that human adipocytes can transdifferentiate into brown-like adipocytes. Together, these data show that MAAC are a versatile tool for studying phenotypic changes of mature adipocytes and provide an improved translational model for drug development.
Collapse
Affiliation(s)
- Matthew J Harms
- Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Qian Li
- Department of Cell and Molecular Biology (CMB), Karolinska Institutet, Stockholm 17177, Sweden
| | - Sunjae Lee
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm 17121, Sweden
| | - Cheng Zhang
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm 17121, Sweden
| | - Bengt Kull
- Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Stefan Hallen
- Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Anders Thorell
- Department of Clinical Sciences, Danderyds Hospital, Karolinska Institutet and Department of Surgery, Ersta Hospital, Stockholm 11691, Sweden
| | - Ida Alexandersson
- Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Carolina E Hagberg
- Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre (KI/AZ ICMC), Department of Medicine, Karolinska Institutet, Stockholm 17176, Sweden
| | - Xiao-Rong Peng
- Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Adil Mardinoglu
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm 17121, Sweden; Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London SE1 9RT, United Kingdom
| | - Kirsty L Spalding
- Department of Cell and Molecular Biology (CMB), Karolinska Institutet, Stockholm 17177, Sweden; Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre (KI/AZ ICMC), Department of Medicine, Karolinska Institutet, Stockholm 17176, Sweden
| | - Jeremie Boucher
- Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden; The Lundberg Laboratory for Diabetes Research, University of Gothenburg, Sweden; Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Sweden.
| |
Collapse
|
265
|
Nowakowski P, Naliwajko SK, Markiewicz‐Żukowska R, Borawska MH, Socha K. The two faces of Coprinus comatus-Functional properties and potential hazards. Phytother Res 2020; 34:2932-2944. [PMID: 32462723 PMCID: PMC7754439 DOI: 10.1002/ptr.6741] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 04/23/2020] [Accepted: 05/07/2020] [Indexed: 01/01/2023]
Abstract
Mushrooms have been used for centuries not only as food but also in traditional medicine as a source of components with pro‐health activity. One of them is Coprinus comatus (O.F.Müll.) Pers. also called shaggy mane, chicken drumstick mushroom, or lawyer's wig. In Asian countries, C. comatus (CC) is approved as edible mushroom and often cultivated for consumption, whereas in many other countries, although it is widespread, it is unrecognized and not used. In this review, for the first time, we discussed about the composition related to functional properties as well as the potential risks associated with consumption of CC by reviewing scientific literature. The information has been collected in order to get to know this species thoroughly. Various studies show many of the physiological activities, such as antioxidant, anticancer, antiandrogenic, hepatoprotective, acetylcholinesterase inhibitory, antiinflammatory, antidiabetic, antiobesity, antibacterial, antifungal, antinematode, and antiviral. Besides positive physiological properties, CC has also negative features, for example, skin reactions in patients with dermatitis and atopic predisposition, risk of confusion with poisonous mushrooms, quick autolysis after collection, and contamination of toxic elements.
Collapse
Affiliation(s)
- Patryk Nowakowski
- Department of Bromatology, Faculty of Pharmacy with the Division of Laboratory MedicineMedical University of BialystokBialystokPoland
| | - Sylwia K. Naliwajko
- Department of Bromatology, Faculty of Pharmacy with the Division of Laboratory MedicineMedical University of BialystokBialystokPoland
| | - Renata Markiewicz‐Żukowska
- Department of Bromatology, Faculty of Pharmacy with the Division of Laboratory MedicineMedical University of BialystokBialystokPoland
| | - Maria H. Borawska
- Department of Bromatology, Faculty of Pharmacy with the Division of Laboratory MedicineMedical University of BialystokBialystokPoland
| | - Katarzyna Socha
- Department of Bromatology, Faculty of Pharmacy with the Division of Laboratory MedicineMedical University of BialystokBialystokPoland
| |
Collapse
|
266
|
Ida Y, Hikage F, Itoh K, Ida H, Ohguro H. Prostaglandin F2α agonist-induced suppression of 3T3-L1 cell adipogenesis affects spatial formation of extra-cellular matrix. Sci Rep 2020; 10:7958. [PMID: 32409724 PMCID: PMC7224398 DOI: 10.1038/s41598-020-64674-1] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 04/20/2020] [Indexed: 12/11/2022] Open
Abstract
To establish a deepening of the upper eyelid sulcus (DUES) model that can be induced by prostaglandin (PG) analogues, a three-dimension (3D) tissue culture was employed. Upon adipogenesis of the 3T3-L1 organoid, the effects of either Bimatoprost acid (BIM-A), or PGF2α were examined. During the adipogenesis, organoid size, lipid staining by BODIPY and expression of the extracellular matrix (ECM) by immunocytochemistry and/or quantitative PCR were employed. The size of the organoid increased remarkably during the adipogenesis, while such increases were significantly inhibited by the presence of PGF2α or BIM-A. BODIPY positive lipid-laden cells significantly increased during the adipogenesis, while in contrast they were greatly suppressed by the presence of PGF2α. Characteristic and spatial changes in ECM expressions observed upon adipogenesis were greatly modified by the presence of PGs. Our present study using a 3D tissue culture may be a suitable strategy toward understanding disease etiology of DUES.
Collapse
Affiliation(s)
- Yosuke Ida
- Departments of Ophthalmology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Fumihito Hikage
- Departments of Ophthalmology, Sapporo Medical University School of Medicine, Sapporo, Japan.
| | - Kaku Itoh
- Departments of Ophthalmology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Haruka Ida
- Departments of Ophthalmology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hiroshi Ohguro
- Departments of Ophthalmology, Sapporo Medical University School of Medicine, Sapporo, Japan
| |
Collapse
|
267
|
Ramskov Tetzlaff CN, Svingen T, Vinggaard AM, Rosenmai AK, Taxvig C. Bisphenols B, E, F, and S and 4-cumylphenol induce lipid accumulation in mouse adipocytes similarly to bisphenol A. ENVIRONMENTAL TOXICOLOGY 2020; 35:543-552. [PMID: 31820560 DOI: 10.1002/tox.22889] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 11/27/2019] [Accepted: 12/01/2019] [Indexed: 05/27/2023]
Abstract
Bisphenol A (BPA) has been widely reported to exert endocrine disrupting effects, including the induction of adipogenesis in cultured preadipocytes and intact animals. Because of the potential harm to human health, BPA is being substituted by structurally related bisphenols. Whether or not such BPA analogues are safe substitutes, however, remains largely unknown. Here, we investigated the potential of bisphenol B (BPB), bisphenol E (BPE), bisphenol F (BPF), bisphenol S (BPS), and 4-cumylphenol (4-CP) to affect lipid and hormone levels in 3 T3-L1 cells. We found that BPB, BPE, BPF, BPS, and 4-CP all affected lipid accumulation and leptin levels to the same extent and potencies as BPA. Based on these and other results, we conclude that these BPA analogues and 4-CP most likely will elicit similar effects on adipocytes as BPA. Using them to substitute BPA in products should be done with caution.
Collapse
Affiliation(s)
- Cecilie N Ramskov Tetzlaff
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, Lyngby, Denmark
| | - Terje Svingen
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, Lyngby, Denmark
| | - Anne M Vinggaard
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, Lyngby, Denmark
| | - Anna K Rosenmai
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, Lyngby, Denmark
| | - Camilla Taxvig
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, Lyngby, Denmark
| |
Collapse
|
268
|
Urrutia O, Mendizabal JA, Alfonso L, Soret B, Insausti K, Arana A. Adipose Tissue Modification through Feeding Strategies and Their Implication on Adipogenesis and Adipose Tissue Metabolism in Ruminants. Int J Mol Sci 2020; 21:E3183. [PMID: 32365995 PMCID: PMC7246642 DOI: 10.3390/ijms21093183] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 04/22/2020] [Accepted: 04/28/2020] [Indexed: 11/25/2022] Open
Abstract
Dietary recommendations by health authorities have been advising of the importance of diminishing saturated fatty acids (SFA) consumption and replacing them by polyunsaturated fatty acids (PUFA), particularly omega-3. Therefore, there have been efforts to enhance food fatty acid profiles, helping them to meet human nutritional recommendations. Ruminant meat is the major dietary conjugated linoleic acid (CLA) source, but it also contains SFA at relatively high proportions, deriving from ruminal biohydrogenation of PUFA. Additionally, lipid metabolism in ruminants may differ from other species. Recent research has aimed to modify the fatty acid profile of meat, and other animal products. This review summarizes dietary strategies based on the n-3 PUFA supplementation of ruminant diets and their effects on meat fatty acid composition. Additionally, the role of n-3 PUFA in adipose tissue (AT) development and in the expression of key genes involved in adipogenesis and lipid metabolism is discussed. It has been demonstrated that linseed supplementation leads to an increase in α-linolenic acid (ALA) and eicosapentaenoic acid (EPA), but not in docosahexaenoic acid (DHA), whilst fish oil and algae increase DHA content. Dietary PUFA can alter AT adiposity and modulate lipid metabolism genes expression, although further research is required to clarify the underlying mechanism.
Collapse
Affiliation(s)
- Olaia Urrutia
- IS-FOOD Institute, Escuela Técnica Superior de Ingeniería Agronómica y Biociencias, Departamento de Agronomía, Biotecnología y Alimentación, Universidad Pública de Navarra, 31006 Pamplona, Spain; (J.A.M.); (L.A.); (B.S.); (K.I.); (A.A.)
| | | | | | | | | | | |
Collapse
|
269
|
Choi YR, Shim J, Kim MJ. Genistin: A Novel Potent Anti-Adipogenic and Anti-Lipogenic Agent. Molecules 2020; 25:E2042. [PMID: 32349444 PMCID: PMC7248826 DOI: 10.3390/molecules25092042] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 04/17/2020] [Accepted: 04/23/2020] [Indexed: 12/23/2022] Open
Abstract
Soy isoflavones are popular ingredients with anti-adipogenic and anti-lipogenic properties. The anti-adipogenic and anti-lipogenic properties of genistein are well-known, but those of genistin and glycitein remain unknown, and those of daidzein are characterized by contrasting data. Therefore, the purpose of our study was to investigate the effects of daidzein, glycitein, genistein, and genistin on adipogenesis and lipogenesis in 3T3-L1 cells. Proliferation of 3T3-L1 preadipocytes was unaffected by genistin and glycitein, but it was affected by 50 and 100 µM genistein and 100 µM daidzein for 48 h. Among the four isoflavones, only 50 and 100 µM genistin and genistein markedly suppressed lipid accumulation during adipogenesis in 3T3-L1 cells through a similar signaling pathway in a dose-dependent manner. Genistin and genistein suppress adipocyte-specific proteins and genes, such as peroxisome proliferator-activated receptor γ (PPARγ), CCAAT-enhancer-binding protein α (C/EBPα), and adipocyte binding protein 2 (aP2)/fatty acid-binding protein 4 (FABP4), and lipogenic enzymes such as ATP citrate lyase (ACL), acetyl-CoA carboxylase 1 (ACC1), and fatty acid synthase (FAS). Both isoflavones also activate AMP-activated protein kinase α (AMPKα), an essential factor in adipocyte differentiation, and inhibited sterol regulatory element-binding transcription factor 1c (SREBP-1c). These results indicate that genistin is a potent anti-adipogenic and anti-lipogenic agent.
Collapse
Affiliation(s)
- Yae Rim Choi
- Research Division of Food Functionality, Korea Food Research Institute, Wanju 55365, Korea (J.S.)
- Department of Food Science and Engineering, Ewha Womans University, Seoul 03760, Korea
| | - Jaewon Shim
- Research Division of Food Functionality, Korea Food Research Institute, Wanju 55365, Korea (J.S.)
| | - Min Jung Kim
- Research Division of Food Functionality, Korea Food Research Institute, Wanju 55365, Korea (J.S.)
| |
Collapse
|
270
|
Perera A, Ton SH, Moorthy M, Palanisamy UD. The insulin-sensitising properties of the ellagitannin geraniin and its metabolites from Nephelium lappaceum rind in 3T3-L1 cells. Int J Food Sci Nutr 2020; 71:940-953. [PMID: 32319838 DOI: 10.1080/09637486.2020.1754348] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
In this study, the insulin-like and insulin sensitising effects of the ellagitannins geraniin, corilagin, ellagic acid, gallic acid and Nephelium lappaceum rind extract in 3T3-L1 adipocytes was investigated. It was observed that non-toxic concentrations of geraniin and its metabolites (0.2-20 μM) and N. lappaceum extract (0.2-20 μg/mL) exhibited insulin-like properties in the absence of insulin and insulin-sensitising properties in the presence of insulin particularly with regards to glucose uptake in 3T3-L1 adipocytes. The compounds were further able to promote adipocyte differentiation and may be involved in the inhibition of lipolysis in 3T3-L1 adipocytes in the presence of insulin. However further study into the molecular mechanisms of action of these compounds need to be carried out to better understand the potential of these compounds/extracts to act as therapeutic agents for hyperglycaemia associated with diabetes mellitus and obesity.
Collapse
Affiliation(s)
- Asiri Perera
- Tropical Medicine and Biology Platform, School of Science, Monash University, Bandar Sunway, Selangor, Malaysia
| | - So Ha Ton
- Tropical Medicine and Biology Platform, School of Science, Monash University, Bandar Sunway, Selangor, Malaysia
| | - Mohanambal Moorthy
- Tropical Medicine and Biology Platform, School of Science, Monash University, Bandar Sunway, Selangor, Malaysia.,School of Medicine and Health Sciences, Monash University, Bandar Sunway, Selangor, Malaysia
| | - Uma Devi Palanisamy
- Tropical Medicine and Biology Platform, School of Science, Monash University, Bandar Sunway, Selangor, Malaysia.,School of Medicine and Health Sciences, Monash University, Bandar Sunway, Selangor, Malaysia
| |
Collapse
|
271
|
Wen Z, Tang Z, Li M, Zhang Y, Li J, Cao Y, Zhang D, Fu Y, Wang C. APPL1 knockdown blocks adipogenic differentiation and promotes adipocyte lipolysis. Mol Cell Endocrinol 2020; 506:110755. [PMID: 32045627 DOI: 10.1016/j.mce.2020.110755] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 01/19/2020] [Accepted: 02/06/2020] [Indexed: 01/12/2023]
Abstract
Adipocyte dysfunction is closely associated with the development of obesity, insulin resistance, and type 2 diabetes. In addition to having a positive effect on adiponectin pathway and insulin signaling through direct and/or indirect mechanisms, adapter protein APPL1 has also been reported to regulate body weight, brown fat tissues thermogenesis, and body fat distribution in diabetic individuals. However, there is dearth of data on the specific role of APPL1 on adipogenic differentiation and adipocyte lipolysis. In this study, APPL1's function in adipocyte differentiation and adipocyte lipolysis was evaluated, and the possible mechanisms were investigated. We found that APPL1 knockdown (KD) impeded differentiation of 3T3-L1 preadipocytes into mature 3T3-L1 adipocytes and enhanced basal and insulin-suppressed lipolysis in mature 3T3-L1 adipocytes. APPL1 KD cells presented a reduced autophagic activity in 3T3-L1 preadipocytes and mature 3T3-L1 adipocytes. In 3T3-L1 preadipocytes, APPL1 KD reduced PPARγ protein levels, which was prevented by administration with proteasome inhibitor MG132. Furthermore, APPL1 KD-reduced autophagic activity in mature 3T3-L1 adipocytes was markedly restored by inhibition of PKA, accompanied with prevention of APPL1-induced lipolysis. In addition, APPL1 KD caused insulin resistance in mature 3T3-L1 adipocytes. Unexpectedly, we found that APPL1 overexpression did not appear to play a role in adipogenic differentiation and adipocyte lipolysis. Our results confirmed that APPL1 KD inhibits adipogenic differentiation by suppressing autophagy and enhances adipocyte lipolysis through activating PKA respectively. These findings may deepen our understanding of APPL1 function, especially its regulation on adipocyte biology.
Collapse
Affiliation(s)
- Zhongyuan Wen
- Department of Endocrinology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zhao Tang
- Department of Pathology and Pathophysiology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China; Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Mingxin Li
- Department of Pathology and Pathophysiology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Yemin Zhang
- Department of Pathology and Pathophysiology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Junfeng Li
- Department of Endocrinology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yingkang Cao
- Department of Pathology and Pathophysiology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Deling Zhang
- Department of Pathology and Pathophysiology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Yalin Fu
- Department of Pathology and Pathophysiology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Changhua Wang
- Department of Pathology and Pathophysiology, Wuhan University School of Basic Medical Sciences, Wuhan, 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China.
| |
Collapse
|
272
|
Sharath SS, Ramu J, Nair SV, Iyer S, Mony U, Rangasamy J. Human Adipose Tissue Derivatives as a Potent Native Biomaterial for Tissue Regenerative Therapies. Tissue Eng Regen Med 2020; 17:123-140. [PMID: 31953618 PMCID: PMC7105544 DOI: 10.1007/s13770-019-00230-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/07/2019] [Accepted: 11/15/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Human adipose tissue is a great source of translatable biomaterials owing to its ease of availability and simple processing. Reusing discardable adipose tissue for tissue regeneration helps in mimicking the exact native microenvironment of tissue. Over the past 10 years, extraction, processing, tuning and fabrication of adipose tissue have grabbed the attention owing to their native therapeutic and regenerative potential. The present work gives the overview of next generation biomaterials derived from human adipose tissue and their development with clinical relevance. METHODS Around 300 articles have been reviewed to widen the knowledge on the isolation, characterization techniques and medical applications of human adipose tissue and its derivatives from bench to bedside. The prospective applications of adipose tissue derivatives like autologous fat graft, stromal vascular fraction, stem cells, preadipocyte, adipokines and extracellular matrix, their behavioural mechanism, rational property of providing native bioenvironment, circumventing their translational abilities, recent advances in featuring them clinically have been reviewed extensively to reveal the dormant side of human adipose tissue. RESULTS Basic understanding about the molecular and structural aspect of human adipose tissue is necessary to employ it constructively. This review has nailed the productive usage of human adipose tissue, in a stepwise manner from exploring the methods of extracting derivatives, concerns during processing and its formulations to turning them into functional biomaterials. Their performance as functional biomaterials for skin regeneration, wound healing, soft tissue defects, stem cell and other regenerative therapies under in vitro and in vivo conditions emphasizes the translational efficiency of adipose tissue derivatives. CONCLUSION In the recent years, research interest has inclination towards constructive tissue engineering and regenerative therapies. Unravelling the maximum utilization of human adipose tissue derivatives paves a way for improving existing tissue regeneration and cellular based therapies and other biomedical applications.
Collapse
Affiliation(s)
- Siva Sankari Sharath
- Centre for Nanosciences and Molecular Medicine, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi, 682041, India
| | - Janarthanan Ramu
- Department of Plastic and Reconstructive Surgery, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi, 682041, India
| | - Shantikumar Vasudevan Nair
- Centre for Nanosciences and Molecular Medicine, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi, 682041, India
| | - Subramaniya Iyer
- Department of Plastic and Reconstructive Surgery, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi, 682041, India
| | - Ullas Mony
- Centre for Nanosciences and Molecular Medicine, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi, 682041, India.
| | - Jayakumar Rangasamy
- Centre for Nanosciences and Molecular Medicine, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi, 682041, India.
| |
Collapse
|
273
|
Adipose Tissue and FoxO1: Bridging Physiology and Mechanisms. Cells 2020; 9:cells9040849. [PMID: 32244542 PMCID: PMC7226803 DOI: 10.3390/cells9040849] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 03/23/2020] [Accepted: 03/30/2020] [Indexed: 12/22/2022] Open
Abstract
Forkhead box O class proteins (FoxOs) are expressed nearly in all tissues and are involved in different functions such as energy metabolism, redox homeostasis, differentiation, and cell cycle arrest. The plasticity of FoxOs is demonstrated by post-translational modifications that determine diverse levels of transcriptional regulations also controlled by their subcellular localization. Among the different members of the FoxO family, we will focus on FoxO1 in adipose tissue, where it is abundantly expressed and is involved in differentiation and transdifferentiation processes. The capability of FoxO1 to respond differently in dependence of adipose tissue subtype underlines the specific involvement of the transcription factor in energy metabolism and the “browning” process of adipocytes. FoxO1 can localize to nuclear, cytoplasm, and mitochondrial compartments of adipocytes responding to different availability of nutrients and source of reactive oxygen species (ROS). Specifically, fasted state produced-ROS enhance the nuclear activity of FoxO1, triggering the transcription of lipid catabolism and antioxidant response genes. The enhancement of lipid catabolism, in combination with ROS buffering, allows systemic energetic homeostasis and metabolic adaptation of white/beige adipocytes. On the contrary, a fed state induces FoxO1 to accumulate in the cytoplasm, but also in the mitochondria where it affects mitochondrial DNA gene expression. The importance of ROS-mediated signaling in FoxO1 subcellular localization and retrograde communication will be discussed, highlighting key aspects of FoxO1 multifaceted regulation in adipocytes.
Collapse
|
274
|
Wang W, Li X, Ding N, Teng J, Zhang S, Zhang Q, Tang H. miR-34a regulates adipogenesis in porcine intramuscular adipocytes by targeting ACSL4. BMC Genet 2020; 21:33. [PMID: 32171241 PMCID: PMC7073017 DOI: 10.1186/s12863-020-0836-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 03/04/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Intramuscular fat (IMF) content is an important factor in porcine meat quality. Previously, we showed that miR-34a was less abundant in liver tissue from pigs with higher backfat thickness, compared to pigs with lower backfat thickness. The purpose of this present study was to explore the role of miR-34a in adipogenesis. RESULT Bioinformatics analysis identified Acyl-CoA synthetase long chain family member 4 (ACSL4) as a putative target of miR-34a. Using a luciferase reporter assay, we verified that miR-34a binds the ACSL4 mRNA at the 3'UTR. To examine the role of the miR-34a-ACSL4 interaction in IMF deposition in the pig, mRNA and protein expression of the ACSL4 gene was measured in primary intramuscular preadipocytes transfected with miR-34a mimic and inhibitor. Our results showed that ACSL4 is expressed throughout the entire differentiation process in pig preadipocytes, similar to the lipogenesis-associated genes PPARγ and aP2. Transfection with miR-34a mimic reduced lipid droplet formation during adipogenesis, while miR-34a inhibitor increased lipid droplet accumulation. Transfection with miR-34a mimic also reduced the mRNA and protein expression of ACSL4 and lipogenesis genes, including PPARγ, aP2, and SREBP-1C, but increased the expression of steatolysis genes such as ATGL and Sirt1. In contrast, the miR-34a inhibitor had the opposite effect on gene expression. Further, knockdown of ACSL4 decreased lipid droplet accumulation. CONCLUSIONS Our results support the hypothesis that miR-34a regulates intramuscular fat deposition in porcine adipocytes by targeting ACSL4.
Collapse
Affiliation(s)
- Wenwen Wang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, No. 61, Daizong Street, Tai’an City, 271018 Shandong Province China
| | - Xiuxiu Li
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, No. 61, Daizong Street, Tai’an City, 271018 Shandong Province China
| | - Ning Ding
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, No. 61, Daizong Street, Tai’an City, 271018 Shandong Province China
| | - Jun Teng
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, No. 61, Daizong Street, Tai’an City, 271018 Shandong Province China
| | - Shen Zhang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, No. 61, Daizong Street, Tai’an City, 271018 Shandong Province China
| | - Qin Zhang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, No. 61, Daizong Street, Tai’an City, 271018 Shandong Province China
| | - Hui Tang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, No. 61, Daizong Street, Tai’an City, 271018 Shandong Province China
| |
Collapse
|
275
|
Ma X, Sun J, Zhu S, Du Z, Li D, Li W, Li Z, Tian Y, Kang X, Sun G. MiRNAs and mRNAs Analysis during Abdominal Preadipocyte Differentiation in Chickens. Animals (Basel) 2020; 10:ani10030468. [PMID: 32168898 PMCID: PMC7143929 DOI: 10.3390/ani10030468] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 03/02/2020] [Accepted: 03/07/2020] [Indexed: 12/13/2022] Open
Abstract
Simple Summary We sequenced the miRNAs and mRNAs of preabdominal fat cells and differentiated adipocytes, and target genes of miRNA combined with mRNA transcriptome data jointly. We found that the MAPK signal pathway, insulin signal pathway, fatty acid metabolism, ECM( extracellular matrix)–receptor interaction, and other signal pathways were involved in the differentiation of preabdominal fat cells. In addition, we found that some miRNAs–mRNAs combinations were strongly related to the differentiation of fat cells (miR-214−ACSBG2, NFKB2, CAMK2A, ACLY, CCND3, PLK3, ITGB2; miR-148a-5p−ROCK2; miR-10a-5p−ELOVL5; miR-146b-5p−LAMA4; miR-6615-5p−FLNB; miR-1774−COL6A1). Our findings provide important resources for the study of adipocyte differentiation. Abstract The excessive deposition of abdominal fat has become an important factor in restricting the production efficiency of chickens, so reducing abdominal fat deposition is important for improving growth rate. It has been proven that miRNAs play an important role in regulating many physiological processes of organisms. In this study, we constructed a model of adipogenesis by isolating preadipocytes (Ab-Pre) derived from abdominal adipose tissue and differentiated adipocytes (Ab-Ad) in vitro. Deep sequencing of miRNAs and mRNAs expressed in Ab-Pre and Ab-Ad groups was conducted to explore the effect of miRNAs and mRNAs on fat deposition. We identified 80 differentially expressed miRNAs (DEMs) candidates, 58 of which were up-regulated and 22 down-regulated. Furthermore, six miRNAs and six mRNAs were verified by qRT-PCR, and the results showed that the expression of the DEMs and differentially expressed genes (DEGs) in the two groups was consistent with our sequencing results. When target genes of miRNA were combined with mRNA transcriptome data, a total of 891 intersection genes were obtained, we predicted the signal pathways of cross genes enrichment to the MAPK signal pathway, insulin signal pathway, fatty acid metabolism, and ECM–receptor interaction. Meanwhile, we constructed miRNA and negatively correlated mRNA target networks, including 12 miRNA–mRNAs pairs, which showed a strong association with the abdominal adipocyte differentiation (miR-214−ACSBG2, NFKB2, CAMK2A, ACLY, CCND3, PLK3, ITGB2; miR-148a-5p−ROCK2; miR-10a-5p−ELOVL5; miR-146b-5p−LAMA4; miR-6615-5p−FLNB; miR-1774−COL6A1). Overall, these findings provide a background for further research on lipid metabolism. Thus, we can better understand the molecular genetic mechanism of chicken abdominal fat deposition.
Collapse
|
276
|
Choi Y, Choi H, Yoon BK, Lee H, Seok JW, Kim HJ, Kim JW. Serpina3c Regulates Adipogenesis by Modulating Insulin Growth Factor 1 and Integrin Signaling. iScience 2020; 23:100961. [PMID: 32193145 PMCID: PMC7076559 DOI: 10.1016/j.isci.2020.100961] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 11/19/2019] [Accepted: 02/28/2020] [Indexed: 12/15/2022] Open
Abstract
Preadipocyte differentiation can be induced upon a hormonal treatment, and various factors secreted by the cells may contribute to adipogenesis. In this study, RNA-seq revealed Serpina3c as a critical factor regulating the signaling network during adipogenesis. Serpina3c is a secretory protein and is highly expressed in fat tissues. Knockdown of Serpina3c decreased adipogenesis by attenuating the mitotic clonal expansion of 3T3-L1 cells. These cells exhibited decreases in integrin α5, which abolished the phosphorylation of integrin β3. We found that Serpina3c inhibits a serine protease that regulates integrin α5 degradation. Knockdown of Serpina3c disrupted integrin-mediated insulin growth factor 1 (IGF-1) signaling and ERK activation. Serpina3c-mediated regulation of integrin-IGF-1 signaling is also associated with AKT activation, which affects the nuclear translocation of GSK3β. Altogether, our results indicate that Serpina3c secreted from differentiating adipocytes inhibits serine proteases to modulate integrin/IGF-1-mediated ERK and AKT signaling and thus is a critical factor contributing to adipogenesis. RNA-seq revealed Serpina3c as a critical factor regulating adipogenesis Knockdown of Serpina3c attenuated the mitotic clonal expansion of 3T3-L1 cells Knockdown of Serpina3c leads to the degradation of integrin α5 Serpina3c regulates integrin-mediated IGF-1 signaling and ERK/AKT activation
Collapse
Affiliation(s)
- Yoonjeong Choi
- Department of Biochemistry and Molecular Biology, Chronic Intractable Disease for Systems Medicine Research Center, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, South Korea; Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul 03722, South Korea
| | - Hyeonjin Choi
- Department of Biochemistry and Molecular Biology, Chronic Intractable Disease for Systems Medicine Research Center, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, South Korea
| | - Bo Kyung Yoon
- Department of Biochemistry and Molecular Biology, Chronic Intractable Disease for Systems Medicine Research Center, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, South Korea; Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul 03722, South Korea
| | - Hyemin Lee
- Department of Biochemistry and Molecular Biology, Chronic Intractable Disease for Systems Medicine Research Center, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, South Korea; Department of Integrated OMICS for Biomedical Sciences, Graduate School, Yonsei University, Seoul 03722, South Korea
| | - Jo Woon Seok
- Department of Biochemistry and Molecular Biology, Chronic Intractable Disease for Systems Medicine Research Center, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, South Korea; Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul 03722, South Korea
| | - Hyo Jung Kim
- Department of Biochemistry and Molecular Biology, Chronic Intractable Disease for Systems Medicine Research Center, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, South Korea.
| | - Jae-Woo Kim
- Department of Biochemistry and Molecular Biology, Chronic Intractable Disease for Systems Medicine Research Center, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, South Korea; Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul 03722, South Korea; Department of Integrated OMICS for Biomedical Sciences, Graduate School, Yonsei University, Seoul 03722, South Korea.
| |
Collapse
|
277
|
Genomic regions associated with principal components for growth, visual score and reproductive traits in Nellore cattle. Livest Sci 2020. [DOI: 10.1016/j.livsci.2020.103936] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
278
|
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: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
279
|
Peroxisome proliferator-activated receptor-coactivator 1-beta (PGC-1β) modulates the expression of genes involved in adipogenesis during preadipocyte differentiation in chicken. Gene 2020; 741:144516. [PMID: 32119914 DOI: 10.1016/j.gene.2020.144516] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 02/25/2020] [Accepted: 02/26/2020] [Indexed: 11/21/2022]
Abstract
To study the influence of the PGC-1β gene on chicken adipocyte proliferation and differentiation, we constructed RNA interference (RNAi) vectors that target the PGC-1β gene and transfected these vectors into adipocytes. Oil Red O staining and a CCK-8 cell kit were used to determine cell triglyceride accumulation status and cell proliferation after transfection, respectively. The mRNA abundances of PGC-1β and adipocyte-differentiation-related genes (PPARγ, C/EBPα, SREBP-1c, FAS, and A-FABP) were detected by real-time PCR. The results showed that the mRNA and protein abundances of PGC-1β in PGC-1β-shRNA transfected adipocytes were significantly lower than those in the control. Interference decreased cell differentiation, but did not depress the cell proliferation. PGC-1β interference impeded the triglyceride accumulation, the mRNA expression levels of nuclear receptors PPARγ and SREBP-1c, and fatty acid synthetase (FAS), and both proteins PPARγ and SREBP-1c, and the fatty acids transporting protein A-FABP. Generally, PGC-1β modulated the cell differentiation and triglyceride accumulation in chicken adipocytes.
Collapse
|
280
|
|
281
|
Miao Z, Wang S, Wang Y, Guo L, Zhang J, Liu Y, Yang Q. A Potential Linking between Vitamin D and Adipose Metabolic Disorders. Can J Gastroenterol Hepatol 2020; 2020:2656321. [PMID: 32149047 PMCID: PMC7049848 DOI: 10.1155/2020/2656321] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 11/10/2019] [Accepted: 11/27/2019] [Indexed: 02/06/2023] Open
Abstract
Vitamin D has been discovered centuries ago, and current studies have focused on the biological effects of vitamin D on adipogenesis. Besides its role in calcium homeostasis and energy metabolism, vitamin D is also involved in the regulation of development and process of metabolic disorders. Adipose tissue is a major storage depot of vitamin D. This review summarized studies on the relationship between vitamin D and adipogenesis and furthermore focuses on adipose metabolic disorders. We reviewed the biological roles and functionalities of vitamin D, the correlation between vitamin D and adipose tissue, the effect of vitamin D on adipogenesis, and adipose metabolic diseases. Vitamin D is associated with adipogenesis, and vitamin D supplements can reduce the burden caused by metabolic diseases. The review provides new insights and basis for medical therapy on adipose metabolic diseases.
Collapse
Affiliation(s)
- Zhiguo Miao
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan 453003, China
| | - Shan Wang
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan 453003, China
| | - Yimin Wang
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan 453003, China
| | - Liping Guo
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan 453003, China
| | - Jinzhou Zhang
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan 453003, China
| | - Yang Liu
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, Henan 453003, China
| | - Qiyuan Yang
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| |
Collapse
|
282
|
Dewhurst-Trigg R, Hulston CJ, Markey O. The effect of quantity and quality of dietary fat intake on subcutaneous white adipose tissue inflammatory responses. Proc Nutr Soc 2020; 79:1-15. [PMID: 32063233 DOI: 10.1017/s0029665120000038] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The global prevalence of obesity and obesity-associated cardiometabolic diseases is a significant public health burden. Chronic low-grade inflammation in metabolic tissues such as white adipose tissue (WAT) is linked to obesity and may play a role in disease progression. The overconsumption of dietary fat has been suggested to modulate the WAT inflammatory environment. It is also recognised that fats varying in degree of fatty acid saturation may elicit differential WAT inflammatory responses. This information has originated predominantly from animal or cell models and translation into human participants in vivo remains limited. This review will summarise human intervention studies investigating the effect of dietary fat quantity and quality on subcutaneous WAT inflammation, with a specific focus on the toll-like receptor 4 (TLR4)/NF-κB and nucleotide-binding and oligomerisation domain-like receptor, leucine-rich repeat and pyrin domain-containing 3 (NLRP3) inflammasome molecular signalling pathways. Overall, firm conclusions are hard to draw regarding the effect of dietary fat quantity and quality on WAT inflammatory responses due to the heterogeneity of study designs, diet composition and participant cohorts recruited. Previous studies have predominantly focused on measures of WAT gene expression. It is suggested that future work includes measures of WAT total content and phosphorylation of proteins involved in TLR4/NF-κB and NLRP3 signalling as this is more representative of alterations in WAT physiological function. Understanding pathways linking the intake of total fat and specific fatty acids with WAT metabolic-inflammatory responses may have important implications for public health by informing dietary guidelines aimed at cardiometabolic risk reduction.
Collapse
Affiliation(s)
- R Dewhurst-Trigg
- School of Sport, Exercise and Health Sciences, Loughborough University, LoughboroughLE11 3TU, UK
| | - C J Hulston
- School of Sport, Exercise and Health Sciences, Loughborough University, LoughboroughLE11 3TU, UK
| | - O Markey
- School of Sport, Exercise and Health Sciences, Loughborough University, LoughboroughLE11 3TU, UK
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences, University of Reading, ReadingRG6 6AP, UK
| |
Collapse
|
283
|
King S, Baptiston Tanaka C, Ross D, Kruzic JJ, Levinger I, Klineberg I, Brennan‐Speranza TC. A diet high in fat and fructose adversely affects osseointegration of titanium implants in rats. Clin Exp Dent Res 2020; 6:107-116. [PMID: 32067396 PMCID: PMC7025982 DOI: 10.1002/cre2.255] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 09/02/2019] [Accepted: 09/10/2019] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVES Diet-induced metabolic dysfunction such as type 2 diabetes mellitus increases the risk of implant failure in both dental and orthopaedic settings. We hypothesised that a diet high in fat and fructose would adversely affect peri-implant bone structure and function including osseointegration. MATERIALS AND METHODS Thirty female Sprague-Dawley rats were divided into three groups (n = 10), control group (normal chow) and two intervention groups on a high-fat (60%), high-fructose (20%; HFHF) diet. Titanium implants were placed in the proximal tibial metaphysis in all groups either before commencing the diet (dHFHF group) or 6 weeks after commencing the diet (HFHF group) and observed for an 8-week healing period. Fasting blood glucose levels (fBGLs) were measured weekly. Structural and functional features of the peri-implant bone, including bone-to-implant contact (BIC), were analysed post euthanasia using microcomputed tomography, pull-out tests, and dynamic histomorphometry. RESULTS The fBGLs were unchanged across all groups. Peri-implant trabecular bone volume was reduced in the HFHF group compared with controls (p = .02). Percentage BIC was reduced in both HFHF group (25.42 ± 3.61) and dHFHF group (28.56 ± 4.07) compared with the control group (43.26 ± 3.58, p < .05) and reflected the lower pull-out loads required in those groups. Osteoblast activity was reduced in both intervention groups compared with the control group (p < .05). CONCLUSION The HFHF diet compromised osseointegration regardless of whether the implant was placed before or after the onset of the diet and, despite the absence of elevated fBGLs, confirming that changes in bone cell function affected both the initiation and maintenance of osseointegration independent of blood glucose levels.
Collapse
Affiliation(s)
- Shalinie King
- Sydney Dental School, Faculty of Medicine and HealthThe University of SydneySydneyNew South WalesAustralia
| | | | - Dean Ross
- Department of Physiology and Bosch Institute for Medical ResearchThe University of SydneySydneyNew South WalesAustralia
| | - Jamie J. Kruzic
- School of Mechanical and Manufacturing EngineeringUNSW SydneySydneyNew South WalesAustralia
| | - Itamar Levinger
- Institute for Health and Sport (IHES)Victoria UniversityMelbourneVictoriaAustralia
- Australian Institute for Musculoskeletal Science (AIMSS), Department of Medicine‐Western Health, Melbourne Medical SchoolThe University of MelbourneMelbourneVictoriaAustralia
| | - Iven Klineberg
- Sydney Dental School, Faculty of Medicine and HealthThe University of SydneySydneyNew South WalesAustralia
| | - Tara C. Brennan‐Speranza
- Department of Physiology and Bosch Institute for Medical ResearchThe University of SydneySydneyNew South WalesAustralia
| |
Collapse
|
284
|
Choi DH, Han JH, Yu KH, Hong M, Lee SY, Park KH, Lee SU, Kwon TH. Antioxidant and Anti-Obesity Activities of Polygonum cuspidatum Extract through Alleviation of Lipid Accumulation on 3T3-L1 Adipocytes. J Microbiol Biotechnol 2020; 30:21-30. [PMID: 31838799 PMCID: PMC9728287 DOI: 10.4014/jmb.1910.10040] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Natural products are widely used due to their various biological activities which include antiinflammatory, antioxidant, and anti-obesity effects. In this study, we determined the antioxidative and anti-obesity effects of Polygonum cuspidatum 50% ethanol extract (PEE). The antioxidative effect of PEE was evaluated using its radical scavenging activity, total phenolic content, and reducing power. The anti-obesity effect of PEE was investigated using 3T3-L1 adipocytes. The antioxidative activity of PEE was progressively increased in various concentrations, mainly due to the presence of phenolic compounds. PEE also alleviated lipid accumulation on 3T3-L1 adipocytes and downregulated the mRNA and protein production of adipogenesis-related (SREBP-1c, PPARγ, C/EBPα) and lipogenesis-related (aP2, FAS, ACC) markers. Furthermore, we found that the inhibitory effect on lipid accumulation via PEE was caused by the alleviation of NF-κB, p38 MAPK, ERK1/2, and JNK at the protein level. Taken together, our results imply that PEE is a potential antioxidant that can prevent obesityassociated disorders.
Collapse
Affiliation(s)
- Da-Hye Choi
- Department of Research and Development, Chuncheon Bio-industry Foundation (CBF), Chuncheon 24232, Republic of Korea
| | - Joon-Hee Han
- Department of Research and Development, Chuncheon Bio-industry Foundation (CBF), Chuncheon 24232, Republic of Korea
| | - Keun-Hyung Yu
- Department of Research and Development, Chuncheon Bio-industry Foundation (CBF), Chuncheon 24232, Republic of Korea
| | - Min Hong
- Department of Research and Development, Chuncheon Bio-industry Foundation (CBF), Chuncheon 24232, Republic of Korea
| | - Sun-Yeop Lee
- Department of Research and Development, Chuncheon Bio-industry Foundation (CBF), Chuncheon 24232, Republic of Korea
| | - Ka-Hee Park
- Department of Research and Development, Chuncheon Bio-industry Foundation (CBF), Chuncheon 24232, Republic of Korea
| | - Soo-Ung Lee
- Department of Research and Development, Chuncheon Bio-industry Foundation (CBF), Chuncheon 24232, Republic of Korea
| | - Tae-Hyung Kwon
- Department of Research and Development, Chuncheon Bio-industry Foundation (CBF), Chuncheon 24232, Republic of Korea,Corresponding author Phone: +82-33-258-6972 Fax: +82-33-258-6173 E-mail:
| |
Collapse
|
285
|
Li Y, Li C, Wu J, Liu W, Li D, Xu J. Harmane ameliorates obesity though inhibiting lipid accumulation and inducing adipocyte browning. RSC Adv 2020; 10:4397-4403. [PMID: 35495252 PMCID: PMC9049078 DOI: 10.1039/c9ra09383d] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 01/10/2020] [Indexed: 12/25/2022] Open
Abstract
Harmane, a compound derived from Peganum harmala L., was discovered as a lipid accumulation inhibitor with our triglyceride (TG) assay. To investigate the molecular mechanism underlying the role of harmane in the prevention of lipid accumulation, a number of biological experiments have been designed to determine if harmane reduces lipid accumulation by suppressing adipogenesis, lipogenesis, and regulating a specific signal transduction pathway. Our experimental data show that harmane inhibits TG accumulation though down-regulating the expression of adipogenic and lipogenic factors, up-regulating adipocyte browning markers and activating the SIRT1-LKB1-AMPK pathway. Therefore, harmane ameliorates obesity though inhibiting lipid accumulation and inducing adipocyte browning. Harmane ameliorates obesity though inhibiting lipid accumulation and inducing adipocyte browning.![]()
Collapse
Affiliation(s)
- Yanwen Li
- School of Biotechnology and Health Sciences, Wuyi University Jiangmen 529000 China
| | - Chanjuan Li
- School of Pharmaceutical Sciences, Sun Yat-sen University Guangzhou 510006 China
| | - Jiaqiang Wu
- School of Biotechnology and Health Sciences, Wuyi University Jiangmen 529000 China
| | - Wenfeng Liu
- School of Biotechnology and Health Sciences, Wuyi University Jiangmen 529000 China
| | - Dongli Li
- School of Biotechnology and Health Sciences, Wuyi University Jiangmen 529000 China
| | - Jun Xu
- School of Biotechnology and Health Sciences, Wuyi University Jiangmen 529000 China .,School of Pharmaceutical Sciences, Sun Yat-sen University Guangzhou 510006 China
| |
Collapse
|
286
|
Transcriptome Modifications in the Porcine Intramuscular Adipocytes during Differentiation and Exogenous Stimulation with TNF-α and Serotonin. Int J Mol Sci 2020; 21:ijms21020638. [PMID: 31963662 PMCID: PMC7013444 DOI: 10.3390/ijms21020638] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 01/15/2020] [Accepted: 01/16/2020] [Indexed: 02/07/2023] Open
Abstract
Adipocytes are dynamic cells that have critical functions to maintain body energy homeostasis. Adipocyte physiology is affected by the adipogenic differentiation, cell program, as well as by the exogenous stimulation of biochemical factors, such as serotonin and TNF-α. In this work, we investigated the global transcriptome modifications when porcine intramuscular preadipocyte (PIP) was differentiated into porcine mature adipocyte (pMA). Moreover, we studied transcriptome changes in pMA after stimulation with serotonin or TNF-α by using a microarray approach. Transcriptome analysis revealed that the expression of 270, 261, and 249 genes were modified after differentiation, or after serotonin and TNF-α stimulation, respectively. Expression changes in APP, HNF4A, ESR1, EGR1, SRC, HNF1A, FN1, ALB, STAT3, CBL, CEBPB, AR, FOS, CFTR, PAN2, PTPN6, VDR, PPARG, STAT5A and NCOA3 genes which are enriched in the ‘PPAR signaling’ and ‘insulin resistance’ pathways were found in adipocytes during the differentiation process. Dose-dependent serotonin stimulation resulted in a decreased fat accumulation in pMAs. Serotonin-induced differentially expressed genes in pMAs were found to be involved in the significant enrichment of ′GPCR ligand-binding′, ‘cell chemotaxis’, ‘blood coagulation and complement’, ‘metabolism of lipid and lipoproteins’, ‘regulation of lipid metabolism by PPARA’, and ‘lipid digestion, mobilization and transport’ pathways. TNF-α stimulation also resulted in transcriptome modifications linked with proinflammatory responses in the pMA of intramuscular origin. Our results provide a landscape of transcriptome modifications and their linked-biological pathways in response to adipogenesis, and exogenous stimulation of serotonin- and TNF-α to the pMA of intramuscular origin.
Collapse
|
287
|
Kim JH, Lee S, Kim HY, Cho EJ. Acer okamotoanum inhibits adipocyte differentiation by the regulation of adipogenesis and lipolysis in 3T3‑L1 cells. Int J Mol Med 2020; 45:589-596. [PMID: 31894306 DOI: 10.3892/ijmm.2019.4448] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 10/04/2019] [Indexed: 11/06/2022] Open
Abstract
Acer okamotoanum is reported to have various antioxidant, anti‑inflammatory and beneficial immune system effects. The anti‑adipocyte differentiation effects and mechanisms of the ethyl acetate (EtOAc) fraction of an A. okamotoanum extraction was investigated in 3T3‑L1 adipocyte cells. Treatment with differentiation inducers increased the level of triglycerides (TGs) in 3T3‑L1 adipocyte cells compared with an untreated control. However, the EtOAc fraction of A. okamotoanum significantly decreased TGs. Treatment with 1, 2.5 and 5 µg/ml showed weak activity, but TG production was inhibited at 10 µg/ml compared with the control. In addition, A. okamotoanum caused a significant downregulation of proteins related to adipogenesis, such as γ‑cytidine‑cytidine‑adenosine‑adenosine‑thymidine/enhancer binding protein‑α, ‑β and peroxisome proliferator‑activated receptor‑γ, compared with the untreated control. Furthermore, A. okamotoanum significantly upregulated lipolysis related protein, hormone‑sensitive lipase and the phosphorylation of adenosine monophosphate‑activated protein kinase (AMPK). Therefore, these results indicate that A. okamotoanum suppressed adipogenesis and increased lipolysis and the activation of AMPK, suggesting a protective role in adipocyte differentiation.
Collapse
Affiliation(s)
- Ji Hyun Kim
- Department of Food Science and Nutrition and Kimchi Research Institute, Pusan National University, Busan 46241, Republic of Korea
| | - Sanghyun Lee
- Department of Plant Science and Technology, Chung‑Ang University, Anseong 17546, Republic of Korea
| | - Hyun Young Kim
- Department of Food Science, Gyeongnam National University of Science and Technology, Jinju 52725, Republic of Korea
| | - Eun Ju Cho
- Department of Food Science and Nutrition and Kimchi Research Institute, Pusan National University, Busan 46241, Republic of Korea
| |
Collapse
|
288
|
Kokai LE, Sivak WN, Schilling BK, Karunamurthy A, Egro FM, Schusterman MA, Minteer DM, Simon P, D’Amico RA, Rubin JP. Clinical Evaluation of an Off-the-Shelf Allogeneic Adipose Matrix for Soft Tissue Reconstruction. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2020; 8:e2574. [PMID: 32095393 PMCID: PMC7015604 DOI: 10.1097/gox.0000000000002574] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 09/10/2019] [Indexed: 01/20/2023]
Abstract
Biomaterials derived from human adipose extracellular matrix have shown promise in vitro and in animal studies as an off-the-shelf adipogenic matrix for sustained volume replacement. Herein, we report the results of a randomized prospective study conducted with allograft adipose matrix (AAM) grafted into the pannus of presurgical abdominoplasty patients 3 or 6 months before scheduled surgery. This is the first report of a longitudinal histologic analysis of AAM in clinical use. METHODS Ten healthy patients undergoing elective abdominoplasty were recruited to receive AAM before surgery. Enrolled subjects were randomized into either a 3-month follow-up cohort or a 6-month follow-up cohort. Subjects were monitored for adverse events associated with AAM grafting in addition to undergoing serial biopsy. Following surgical excision of the pannus, representative samples from the AAM surgical sites were stained and evaluated with hematoxylin and eosin for tissue morphology, Masson's trichrome for collagen, and perilipin for adipocytes. RESULTS All subjects tolerated AAM with no severe adverse events reported. At 3 months following implantation, AAM remained visible within the confines of the subjects' native surrounding adipose tissue with sparse adipocytes apparent within the matrix. By 6 months, AAM had remodeled and was primarily composed of perilipin-positive adipocytes. Histologic analysis confirmed tissue remodeling (hematoxylin and eosin), adipogenesis (perilipin), and angiogenesis (Masson's trichrome) occurred with the presence of AAM. CONCLUSIONS AAM is a safe, allogeneic, off-the-shelf regenerative matrix that is adipogenic and noninflammatory and promotes angiogenesis.
Collapse
Affiliation(s)
- Lauren E. Kokai
- From the Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pa
- McGowan Institute for Regenerative Medicine, Pittsburgh, Pa
| | - Wesley N. Sivak
- From the Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pa
| | - Benjamin K. Schilling
- From the Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pa
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pa
| | | | - Francesco M. Egro
- From the Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pa
| | - M. Asher Schusterman
- From the Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pa
| | - Danielle M. Minteer
- From the Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pa
- McGowan Institute for Regenerative Medicine, Pittsburgh, Pa
| | - Patsy Simon
- From the Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pa
- McGowan Institute for Regenerative Medicine, Pittsburgh, Pa
| | - Richard A. D’Amico
- Department of Plastic Surgery, Mount Sinai School of Medicine, New York, N.Y
| | - J. Peter Rubin
- From the Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pa
- McGowan Institute for Regenerative Medicine, Pittsburgh, Pa
- Division of Molecular & Genomic Pathology, Pittsburgh, Pa
| |
Collapse
|
289
|
Zhang Y, Ecelbarger CM, Lesniewski LA, Müller CE, Kishore BK. P2Y 2 Receptor Promotes High-Fat Diet-Induced Obesity. Front Endocrinol (Lausanne) 2020; 11:341. [PMID: 32582029 PMCID: PMC7283874 DOI: 10.3389/fendo.2020.00341] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 04/30/2020] [Indexed: 12/11/2022] Open
Abstract
P2Y2, a G protein-coupled receptor (R), is expressed in all organs involved in the development of obesity and insulin resistance. To explore the role of it in diet-induced obesity, we fed male P2Y2-R whole body knockout (KO) and wild type (WT) mice (B6D2 genetic background) with regular diet (CNT; 10% calories as fat) or high-fat diet (HFD; 60% calories as fat) with free access to food and water for 16 weeks, and euthanized them. Adjusted for body weights (BW), KO mice consumed modestly, but significantly more HFD vs. WT mice, and excreted well-formed feces with no taint of fat or oil. Starting from the 2nd week, HFD-WT mice displayed significantly higher BW with terminal mean difference of 22% vs. HFD-KO mice. Terminal weights of white adipose tissue (WAT) were significantly lower in the HFD-KO vs. HFD-WT mice. The expression of P2Y2-R mRNA in WAT was increased by 2-fold in HFD-fed WT mice. Serum insulin, leptin and adiponectin levels were significantly elevated in the HFD-WT mice, but not in the HFD-KO mice. When induced in vitro, preadipocytes derived from KO mice fed regular diet did not differentiate and mature as robustly as those from the WT mice, as assessed by cellular expansion and accumulation of lipid droplets. Blockade of P2Y2-R by AR-C118925 in preadipocytes derived from WT mice prevented differentiation and maturation. Under basal conditions, KO mice had significantly higher serum triglycerides and showed slightly impaired lipid tolerance as compared to the WT mice. HFD-fed KO mice had significantly better glucose tolerance (GTT) as compared to HFD-fed WT mice. Whole body insulin sensitivity and mRNA expression of insulin receptor, IRS-1 and GLUT4 in WAT was significantly higher in HFD-fed KO mice vs. HFD-fed WT mice. On the contrary, the expression of pro-inflammatory molecules MCP-1, CCR2, CD68, and F4/80 were significantly higher in the WAT of HFD-fed WT vs. HFD-fed KO mice. These data suggest that P2Y2-R plays a significant role in the development of diet-induced obesity by promoting adipogenesis and inflammation, and altering the production of adipokines and lipids and their metabolism in adipose tissue, and thereby facilitates HFD-induced insulin resistance.
Collapse
Affiliation(s)
- Yue Zhang
- Department of Veterans Affairs Salt Lake City Health Care System, Nephrology Research, Salt Lake City, UT, United States
- Departments of Internal Medicine, University of Utah Health, Salt Lake City, UT, United States
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Carolyn M. Ecelbarger
- Division of Endocrinology and Metabolism, Department of Medicine, Center for the Study of Sex Differences in Health, Aging, and Disease, Georgetown University, Washington, DC, United States
| | - Lisa A. Lesniewski
- Departments of Internal Medicine, University of Utah Health, Salt Lake City, UT, United States
- Department of Veterans Affairs Salt Lake City Health Care System, Geriatric Research, Education and Clinical Center, Salt Lake City, UT, United States
- Department of Nutrition and Integrative Physiology, University of Utah Health, Salt Lake City, UT, United States
- Center on Aging, University of Utah Health, Salt Lake City, UT, United States
| | - Christa E. Müller
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical and Medicinal Chemistry, University of Bonn, Bonn, Germany
| | - Bellamkonda K. Kishore
- Department of Veterans Affairs Salt Lake City Health Care System, Nephrology Research, Salt Lake City, UT, United States
- Departments of Internal Medicine, University of Utah Health, Salt Lake City, UT, United States
- Department of Nutrition and Integrative Physiology, University of Utah Health, Salt Lake City, UT, United States
- Center on Aging, University of Utah Health, Salt Lake City, UT, United States
- *Correspondence: Bellamkonda K. Kishore
| |
Collapse
|
290
|
Jain S, Yassin MA, Fuoco T, Liu H, Mohamed-Ahmed S, Mustafa K, Finne-Wistrand A. Engineering 3D degradable, pliable scaffolds toward adipose tissue regeneration; optimized printability, simulations and surface modification. J Tissue Eng 2020; 11:2041731420954316. [PMID: 32983402 PMCID: PMC7498972 DOI: 10.1177/2041731420954316] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 08/11/2020] [Indexed: 01/17/2023] Open
Abstract
We present a solution to regenerate adipose tissue using degradable, soft, pliable 3D-printed scaffolds made of a medical-grade copolymer coated with polydopamine. The problem today is that while printing, the medical grade copolyesters degrade and the scaffolds become very stiff and brittle, being not optimal for adipose tissue defects. Herein, we have used high molar mass poly(L-lactide-co-trimethylene carbonate) (PLATMC) to engineer scaffolds using a direct extrusion-based 3D printer, the 3D Bioplotter®. Our approach was first focused on how the printing influences the polymer and scaffold's mechanical properties, then on exploring different printing designs and, in the end, on assessing surface functionalization. Finite element analysis revealed that scaffold's mechanical properties vary according to the gradual degradation of the polymer as a consequence of the molar mass decrease during printing. Considering this, we defined optimal printing parameters to minimize material's degradation and printed scaffolds with different designs. We subsequently functionalized one scaffold design with polydopamine coating and conducted in vitro cell studies. Results showed that polydopamine augmented stem cell proliferation and adipogenic differentiation owing to increased surface hydrophilicity. Thus, the present research show that the medical grade PLATMC based scaffolds are a potential candidate towards the development of implantable, resorbable, medical devices for adipose tissue regeneration.
Collapse
Affiliation(s)
- Shubham Jain
- Department of Fibre and Polymer
Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Mohammed Ahmad Yassin
- Tissue Engineering Group, Department of
Clinical Dentistry, Faculty of Medicine, University of Bergen, Hordaland,
Norway
| | - Tiziana Fuoco
- Department of Fibre and Polymer
Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Hailong Liu
- Department of Fibre and Polymer
Technology, KTH Royal Institute of Technology, Stockholm, Sweden
- Department of Solid Mechanics, KTH Royal
Institute of Technology, Stockholm, Sweden
| | - Samih Mohamed-Ahmed
- Tissue Engineering Group, Department of
Clinical Dentistry, Faculty of Medicine, University of Bergen, Hordaland,
Norway
| | - Kamal Mustafa
- Tissue Engineering Group, Department of
Clinical Dentistry, Faculty of Medicine, University of Bergen, Hordaland,
Norway
| | - Anna Finne-Wistrand
- Department of Fibre and Polymer
Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| |
Collapse
|
291
|
The Effect of MicroRNA-331-3p on Preadipocytes Proliferation and Differentiation and Fatty Acid Accumulation in Laiwu Pigs. BIOMED RESEARCH INTERNATIONAL 2019; 2019:9287804. [PMID: 31886267 PMCID: PMC6914919 DOI: 10.1155/2019/9287804] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 10/03/2019] [Accepted: 11/14/2019] [Indexed: 02/06/2023]
Abstract
Objective The proliferation and differentiation of preadipocytes are regulated by microRNAs (miRNAs), hormones, and other factors. This study aimed to investigate the effects of miR-331-3p on the proliferation and differentiation of preadipocytes in addition to fatty acid metabolism. Methods Preadipocytes were transfected with miR-331-3p mimics, miR-NC, or miR-331-3p inhibitor to explore its effect on cell proliferation and fatty acid accumulation. Furthermore, preadipocytes were transfected with pre-miR-331-3p, pcDNA3.1(+), or miR-331-3p inhibitor to explore its effect on differentiation. Results It was observed that miR-331-3p could inhibit preadipocytes proliferation. Furthermore, miR-331-3p was highly expressed during cellular differentiation and appeared to promote the process. In addition, dual fluorescein analysis showed that dihydrolipoamide S-succinyltransferase (DLST) is a target gene of miR-331-3p, and overexpression of miR-331-3p could regulate the metabolism of fatty acids in the citrate pyruvate cycle by targeting DLST expression. Conclusion In summary, these findings indicated that miR-331-3p exerts contrasting effects on the processes of fat deposition.
Collapse
|
292
|
Adiponectin homolog osmotin, a potential anti-obesity compound, suppresses abdominal fat accumulation in C57BL/6 mice on high-fat diet and in 3T3-L1 adipocytes. Int J Obes (Lond) 2019; 43:2422-2433. [PMID: 31164725 DOI: 10.1038/s41366-019-0383-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 03/26/2019] [Accepted: 04/01/2019] [Indexed: 12/12/2022]
Abstract
OBJECTIVES Obesity is characterized by excessive fat accumulation due to an imbalance between energy intake and expenditure. Osmotin, a plant derived natural protein, is a known homolog of adiponectin. To analyze the role of Osmotin in controlling energy metabolism by suppressing abdominal fat accumulation. METHODS We investigated the effects of osmotin in C57BL/6 mice on high-fat diet and in 3T3-L1 adipocytes by Biochemical tests, Immunofluorescence confocal Microscopy, RT-PCR, and Flow cytometry. RESULTS In this study, we investigated the anti-obesity effects of osmotin on adipocyte differentiation and regulation of the related factors lipolysis and glucose uptake in 3T3-L1 cells in vitro. Moreover, we analyzed the role of osmotin in prevention of insulin resistance, excess fat accumulation and metabolic syndrome in high-fat diet mouse model via AMPK and MAPK pathways in vivo. In addition, osmotin caused cell cycle arrest in G0/G1 phase by regulating expression of p21, p27 and CDK2 and improved glucose control, as concluded from glucose and insulin tolerance tests. CONCLUSION These results reveal the role of osmotin in AMPK downstream signaling. These results provide the first indication that osmotin exerts therapeutic effects on obesity, which could promote development of therapeutic aspects for obesity and related diseases.
Collapse
|
293
|
HWANG JS, LEE SB, CHOI MJ, KIM JT, SEO HG. Anti-adipogenic effect of a turmeric extract-loaded nanoemulsion in 3T3-L1 preadipocytes and high fat diet-fed mice. FOOD SCIENCE AND TECHNOLOGY 2019. [DOI: 10.1590/fst.20718] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
|
294
|
Borah AK, Singh A, Yasmin R, Doley R, Mattaparthi VSK, Saha S. 1α, 25-dihydroxy Vitamin D3 containing fractions of Catharanthus roseus leaf aqueous extract inhibit preadipocyte differentiation and induce lipolysis in 3T3-L1 cells. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 19:338. [PMID: 31783835 PMCID: PMC6883588 DOI: 10.1186/s12906-019-2754-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 11/14/2019] [Indexed: 01/12/2023]
Abstract
BACKGROUND To investigate the potential of Catharanthus roseus leaf aqueous crude extract (CRACE) as a regulator of adipocyte development and function. METHODS 3T3-L1 adipogenesis model was used to investigate the effect of CRACE on adipogenesis. 3T3-L1 preadipocytes (for adipogenic differentiation) and mature 3T3-L1 adipocytes (for adipocyte function) were treated with non-toxic doses of CRACE. The outcomes were corroborated by intracellular lipid accumulation, expression of pro-and anti-adipogenic effector molecules. To investigate CRACE mediated lipolysis, cAMP accumulation, glycerol release and phosphorylation of key effector molecules were tested in treated mature adipocytes. Finally, the extract was fractionated to identify the active molecule/s in the extract. RESULTS CRACE significantly reduced adipocyte differentiation by modulating PPARγ expression. At early stage CRACE directly targeted Lipin1 expression and consequently impacted KLF7, subsequently expression of GATA2, CEBPα, SREBP1c were targeted, with PPARγ expression, particularly curtailed. While CRACE significantly reduced several lipogenic genes like FAS and GPD1 in mature adipocytes, concomitantly, it greatly increased lipolysis resulting in decreased lipid accumulation in mature adipocytes. The increase in lipolysis was due to decreased Akt activation, increased cAMP level, and PKA activity. The fractionation of CRACE allowed identification of two fractions with potent anti-adipogenic activity. Both the fractions contained 1α, 25-dihydroxy Vitamin D3 as major component. CONCLUSIONS 1α, 25-dihydroxy Vitamin D3 containing CRACE can be developed into an effective anti-obesity formulation that decreases adipogenesis and increases lipid catabolism.
Collapse
Affiliation(s)
- Anuj Kumar Borah
- Department of Molecular Biology and Biotechnology, Tezpur University, Napaam, Tezpur, Assam 784028 India
| | - Archana Singh
- Department of Molecular Biology and Biotechnology, Tezpur University, Napaam, Tezpur, Assam 784028 India
| | - Rafika Yasmin
- Department of Molecular Biology and Biotechnology, Tezpur University, Napaam, Tezpur, Assam 784028 India
| | - Robin Doley
- Department of Molecular Biology and Biotechnology, Tezpur University, Napaam, Tezpur, Assam 784028 India
| | | | - Sougata Saha
- Department of Biotechnology, National Institute of Technology, Durgapur, West Bengal 713209 India
| |
Collapse
|
295
|
Wang Y, Fu Y, Yan Z, Zhang XB, Pei M. Impact of Fibronectin Knockout on Proliferation and Differentiation of Human Infrapatellar Fat Pad-Derived Stem Cells. Front Bioeng Biotechnol 2019; 7:321. [PMID: 31803729 PMCID: PMC6873900 DOI: 10.3389/fbioe.2019.00321] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 10/28/2019] [Indexed: 12/15/2022] Open
Abstract
Fibronectin plays an essential role in tissue development and regeneration. However, the effects of fibronectin knockout (FN1-KO) on stem cells' proliferation and differentiation remain unknown. In this study, CRISPR/Cas9 generated FN1-KO in human infrapatellar fat pad-derived stem cells (IPFSCs) was evaluated for proliferation ability including cell cycle and surface markers as well as stemness gene expression and for differentiation capacity including chondrogenic and adipogenic differentiation. High passage IPFSCs were also evaluated for proliferation and differentiation capacity after expansion on decellularized ECM (dECM) deposited by FN1-KO cells. Successful FN1-KO in IPFSCs was confirmed by Sanger sequencing and Inference of CRISPR Edits analysis (ICE) as well as immunostaining for fibronectin expression. Compared to the GFP control, FN1-KO cells showed an increase in cell growth, percentage of cells in the S and G2 phases, and CD105 and CD146 expression but a decrease in expression of stemness markers CD73, CD90, SSEA4, and mesenchymal condensation marker CDH2 gene. FN1-KO decreased both chondrogenic and adipogenic differentiation capacity. Interestingly, IPFSCs grown on dECMs deposited by FN1-KO cells exhibited a decrease in cell proliferation along with a decline in CDH2 expression. After induction, IPFSCs plated on dECMs deposited by FN1-KO cells also displayed decreased expression of both chondrogenic and adipogenic capacity. We concluded that FN1-KO increased human IPFSCs' proliferation capacity; however, this capacity was reversed after expansion on dECM deposited by FN1-KO cells. Significance of fibronectin in chondrogenic and adipogenic differentiation was demonstrated in both FN1-KO IPFSCs and FN(-) matrix microenvironment.
Collapse
Affiliation(s)
- Yiming Wang
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, United States
- Department of Orthopaedics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yawen Fu
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Disease Hospital, Tianjin, China
- Department of Medicine, Loma Linda University, Loma Linda, CA, United States
| | - Zuoqin Yan
- Department of Orthopaedics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiao-Bing Zhang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Disease Hospital, Tianjin, China
- Department of Medicine, Loma Linda University, Loma Linda, CA, United States
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, United States
- WVU Cancer Institute, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, United States
| |
Collapse
|
296
|
Gulyaeva O, Nguyen H, Sambeat A, Heydari K, Sul HS. Sox9-Meis1 Inactivation Is Required for Adipogenesis, Advancing Pref-1 + to PDGFRα + Cells. Cell Rep 2019; 25:1002-1017.e4. [PMID: 30355480 PMCID: PMC6903418 DOI: 10.1016/j.celrep.2018.09.086] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 08/27/2018] [Accepted: 09/26/2018] [Indexed: 12/28/2022] Open
Abstract
Adipocytes arise from the commitment and differentiation of adipose precursors in white adipose tissue (WAT). In studying adipogenesis, precursor markers, including Pref-1 and PDGFRα, are used to isolate precursors from stromal vascular fractions of WAT, but the relation among the markers is not known. Here, we used the Pref-1 promoter-rtTA system in mice for labeling Pref-1+ cells and for inducible inactivation of the Pref-1 target Sox9. We show the requirement of Sox9 for the maintenance of Pref-1+ proliferative, early precursors. Upon Sox9 inactivation, these Pref-1+ cells become PDGFRα+ cells that express early adipogenic markers. Thus, we show that Pref-1+ cells precede PDGFRα+ cells in the adipogenic pathway and that Sox9 inactivation is required for WAT growth and expansion. Furthermore, we show that in maintaining early adipose precursors, Sox9 activates Meis1, which prevents adipogenic differentiation. Our study also demonstrates the Pref-1 promoter-rtTA system for inducible gene inactivation in early adipose precursor populations. The relationship among Sox9+, Pref-1+, and PDGFRα+ WAT precursors has not been studied. Gulyaeva et al. show that Pref-1+ cells are early adipose precursors and, upon Sox9 inactivation, they become PDGFRα+ cells at a later stage of the adipogenic pathway. In maintaining Pref-1+ adipose precursors, Sox9 activates Meis1, which prevents adipogenic differentiation.
Collapse
Affiliation(s)
- Olga Gulyaeva
- Endocrinology Program, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Hai Nguyen
- Endocrinology Program, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Audrey Sambeat
- Department of Nutritional Sciences & Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Kartoosh Heydari
- Cancer Research Laboratory, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Hei Sook Sul
- Endocrinology Program, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Nutritional Sciences & Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
297
|
The Power of LC-MS Based Multiomics: Exploring Adipogenic Differentiation of Human Mesenchymal Stem/Stromal Cells. Molecules 2019; 24:molecules24193615. [PMID: 31597247 PMCID: PMC6804244 DOI: 10.3390/molecules24193615] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 09/26/2019] [Accepted: 10/04/2019] [Indexed: 12/12/2022] Open
Abstract
The molecular study of fat cell development in the human body is essential for our understanding of obesity and related diseases. Mesenchymal stem/stromal cells (MSC) are the ideal source to study fat formation as they are the progenitors of adipocytes. In this work, we used human MSCs, received from surgery waste, and differentiated them into fat adipocytes. The combination of several layers of information coming from lipidomics, metabolomics and proteomics enabled network analysis of the biochemical pathways in adipogenesis. Simultaneous analysis of metabolites, lipids, and proteins in cell culture is challenging due to the compound’s chemical difference, so most studies involve separate analysis with unimolecular strategies. In this study, we employed a multimolecular approach using a two–phase extraction to monitor the crosstalk between lipid metabolism and protein-based signaling in a single sample (~105 cells). We developed an innovative analytical workflow including standardization with in-house produced 13C isotopically labeled compounds, hyphenated high-end mass spectrometry (high-resolution Orbitrap MS), and chromatography (HILIC, RP) for simultaneous untargeted screening and targeted quantification. Metabolite and lipid concentrations ranged over three to four orders of magnitude and were detected down to the low fmol (absolute on column) level. Biological validation and data interpretation of the multiomics workflow was performed based on proteomics network reconstruction, metabolic modelling (MetaboAnalyst 4.0), and pathway analysis (OmicsNet). Comparing MSCs and adipocytes, we observed significant regulation of different metabolites and lipids such as triglycerides, gangliosides, and carnitine with 113 fully reprogrammed pathways. The observed changes are in accordance with literature findings dealing with adipogenic differentiation of MSC. These results are a proof of principle for the power of multimolecular extraction combined with orthogonal LC-MS assays and network construction. Considering the analytical and biological validation performed in this study, we conclude that the proposed multiomics workflow is ideally suited for comprehensive follow-up studies on adipogenesis and is fit for purpose for different applications with a high potential to understand the complex pathophysiology of diseases.
Collapse
|
298
|
Holly JMP, Biernacka K, Perks CM. The Neglected Insulin: IGF-II, a Metabolic Regulator with Implications for Diabetes, Obesity, and Cancer. Cells 2019; 8:cells8101207. [PMID: 31590432 PMCID: PMC6829378 DOI: 10.3390/cells8101207] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 09/30/2019] [Accepted: 10/01/2019] [Indexed: 02/07/2023] Open
Abstract
When originally discovered, one of the initial observations was that, when all of the insulin peptide was depleted from serum, the vast majority of the insulin activity remained and this was due to a single additional peptide, IGF-II. The IGF-II gene is adjacent to the insulin gene, which is a result of gene duplication, but has evolved to be considerably more complicated. It was one of the first genes recognised to be imprinted and expressed in a parent-of-origin specific manner. The gene codes for IGF-II mRNA, but, in addition, also codes for antisense RNA, long non-coding RNA, and several micro RNA. Recent evidence suggests that each of these have important independent roles in metabolic regulation. It has also become clear that an alternatively spliced form of the insulin receptor may be the principle IGF-II receptor. These recent discoveries have important implications for metabolic disorders and also for cancer, for which there is renewed acknowledgement of the importance of metabolic reprogramming.
Collapse
Affiliation(s)
- Jeff M P Holly
- Department of Translational Health Science, Bristol Medical School, Faculty of Health Sciences, University of Bristol, Learning & Research Building, Southmead Hospital, Bristol, BS10 5NB, UK.
| | - Kalina Biernacka
- Department of Translational Health Science, Bristol Medical School, Faculty of Health Sciences, University of Bristol, Learning & Research Building, Southmead Hospital, Bristol, BS10 5NB, UK
| | - Claire M Perks
- Department of Translational Health Science, Bristol Medical School, Faculty of Health Sciences, University of Bristol, Learning & Research Building, Southmead Hospital, Bristol, BS10 5NB, UK
| |
Collapse
|
299
|
Lee JH, Park A, Oh KJ, Lee SC, Kim WK, Bae KH. The Role of Adipose Tissue Mitochondria: Regulation of Mitochondrial Function for the Treatment of Metabolic Diseases. Int J Mol Sci 2019; 20:ijms20194924. [PMID: 31590292 PMCID: PMC6801758 DOI: 10.3390/ijms20194924] [Citation(s) in RCA: 150] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 09/29/2019] [Accepted: 09/30/2019] [Indexed: 02/07/2023] Open
Abstract
: Mitochondria play a key role in maintaining energy homeostasis in metabolic tissues, including adipose tissues. The two main types of adipose tissues are the white adipose tissue (WAT) and the brown adipose tissue (BAT). WAT primarily stores excess energy, whereas BAT is predominantly responsible for energy expenditure by non-shivering thermogenesis through the mitochondria. WAT in response to appropriate stimuli such as cold exposure and β-adrenergic agonist undergoes browning wherein it acts as BAT, which is characterized by the presence of a higher number of mitochondria. Mitochondrial dysfunction in adipocytes has been reported to have strong correlation with metabolic diseases, including obesity and type 2 diabetes. Dysfunction of mitochondria results in detrimental effects on adipocyte differentiation, lipid metabolism, insulin sensitivity, oxidative capacity, and thermogenesis, which consequently lead to metabolic diseases. Recent studies have shown that mitochondrial function can be improved by using thiazolidinedione, mitochondria-targeted antioxidants, and dietary natural compounds; by performing exercise; and by controlling caloric restriction, thereby maintaining the metabolic homeostasis by inducing adaptive thermogenesis of BAT and browning of WAT. In this review, we focus on and summarize the molecular regulation involved in the improvement of mitochondrial function in adipose tissues so that strategies can be developed to treat metabolic diseases.
Collapse
Affiliation(s)
- Jae Ho Lee
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea
| | - Anna Park
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea
| | - Kyoung-Jin Oh
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34141, Korea
| | - Sang Chul Lee
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34141, Korea
| | - Won Kon Kim
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea.
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34141, Korea.
| | - Kwang-Hee Bae
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea.
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34141, Korea.
| |
Collapse
|
300
|
Koh YM, Jang SW, Ahn TW. Anti-obesity effect of Yangkyuksanwha-tang in high-fat diet-induced obese mice. Altern Ther Health Med 2019; 19:246. [PMID: 31488172 PMCID: PMC6728965 DOI: 10.1186/s12906-019-2669-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Accepted: 08/30/2019] [Indexed: 01/10/2023]
Abstract
BACKGROUND Yangkyuksanwha-tang (YST) is an herbal medicine based on Sasang constitutional medicine (SCM) and is widely used in Korean traditional medicine. The aim of the study was to evaluate the effect of YST on obesity in high-fat diet (HFD)-induced obese mice. METHODS We induced obesity in C57bl/6 J mice using a HFD, and then orally administered 300 mg/kg YST for 6 weeks. We measured body weight, food efficiency, organ and fat weight, serum biochemical parameters, and obesity-related gene expression, and carried out histological analysis at the end of the experimental period. RESULTS YST significantly reduced the absolute body weight and food efficiency ratio. The serum, aminotransferase, glucose, total cholesterol, triglyceride, and low-density lipoprotein-cholesterol levels were significantly lower in the YST-treated group than in the control group, whereas the high-density lipoprotein-cholesterol level in the YST-treated group was significantly higher. The YST-treated group also showed a significant reduction in regional fatty tissues and the absolute weight of various organs. We also observed a significantly reduced expression of AP2/FABP4, C/EBP-β, leptin, and SREBP1c/ADD1 mRNA, and significantly increased expression of UCP-2 and adiponectin mRNA in adipose tissue in the YST-treated group. YST also decreased the lipid droplet size and lipid accumulation in the liver, as well as adipocyte size in epididymal adipose tissue. At the dose tested, YST was non-toxic to the liver and kidneys of the mice. CONCLUSION The results imply that YST has anti-obesity effects in obesity-induced mice. Although the number of experimental animals was limited and the drug effects concern mice, rather than humans, which have different constitutions, the study has valuable implications with respect to the general effects of YST.
Collapse
|