1
|
Weijie Z, Meng Z, Chunxiao W, Lingjie M, Anguo Z, Yan Z, Xinran C, Yanjiao X, Li S. Obesity-induced chronic low-grade inflammation in adipose tissue: A pathway to Alzheimer's disease. Ageing Res Rev 2024; 99:102402. [PMID: 38977081 DOI: 10.1016/j.arr.2024.102402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 06/19/2024] [Accepted: 06/30/2024] [Indexed: 07/10/2024]
Abstract
Alzheimer's disease (AD) is a leading cause of cognitive impairment worldwide. Overweight and obesity are strongly associated with comorbidities, such as hypertension, diabetes, and insulin resistance (IR), which contribute substantially to the development of AD and subsequent morbidity and mortality. Adipose tissue (AT) is a highly dynamic organ composed of a diverse array of cell types, which can be classified based on their anatomic localization or cellular composition. The expansion and remodeling of AT in the context of obesity involves immunometabolic and functional shifts steered by the intertwined actions of multiple immune cells and cytokine signaling within AT, which contribute to the development of metabolic disorders, IR, and systemic markers of chronic low-grade inflammation. Chronic low-grade inflammation, a prolonged, low-dose stimulation by specific immunogens that can progress from localized sites and affect multiple organs throughout the body, leads to neurodystrophy, increased apoptosis, and disruption of homeostasis, manifesting as brain atrophy and AD-related pathology. In this review, we sought to elucidate the mechanisms by which AT contributes to the onset and progression of AD in obesity through the mediation of chronic low-grade inflammation, particularly focusing on the roles of adipokines and AT-resident immune cells.
Collapse
Affiliation(s)
- Zhai Weijie
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China; Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Zhao Meng
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China; Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Wei Chunxiao
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China; Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Meng Lingjie
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China; Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Zhao Anguo
- Department of Urology, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou 215000 China
| | - Zhang Yan
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China; Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Cui Xinran
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China; Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Xu Yanjiao
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China; Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Sun Li
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China; Cognitive Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China.
| |
Collapse
|
2
|
Ramesh T, Shahid M. Bacoside-A repressed the differentiation and lipid accumulation of 3T3-L1 preadipocytes by modulating the expression of adipogenic genes. Biotechnol Appl Biochem 2024; 71:741-752. [PMID: 38419375 DOI: 10.1002/bab.2573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 02/07/2024] [Accepted: 02/14/2024] [Indexed: 03/02/2024]
Abstract
Obesity is one of the more complicated diseases, it can induce numerous life-threatening diseases mainly diabetes mellitus, cardiovascular disease, hypertension, and certain cancers. In this study, we assessed the efficacy of bacoside-A (a dammarane-type triterpenoid saponin derived from the plant Bacopa monniera Linn.) on the adipogenesis of 3T3-L1 preadipocytes. Results of this study illustrated that bacoside-A decreased the differentiation of 3T3-L1 cell, as evidenced by diminution of lipid droplets, which contains triglycerides and other lipids. During the differentiation process, transcription factors, which are mainly participating in adipogenesis such us CCAAT/enhancer-binding protein α (C/EBPα) and C/EBPβ, peroxisome proliferator-activated receptor-γ (PPARγ), and sterol regulatory element-binding protein-1c (SREBP-1c), expressions were significantly suppressed by bacoside-A. In addition, bacoside-A showed a potent reduction in genes precise to adipocytes such as lipoprotein lipase (LPL), fatty acid synthase (FAS), adipocyte fatty acid-binding protein (FABP4), and leptin expressions. Further, bacoside-A stimulated the phosphorylation of acetyl CoA carboxylase (ACC) and AMP-activated protein kinase (AMPK). These results demonstrated that bacoside-A has anti-adipogenic effects by regulating the transcription factors involved in adipocyte differentiation. Therefore, bacoside-A might be considered as a potent therapeutic agent for alleviating obesity and hyperlipidemia.
Collapse
Affiliation(s)
- Thiyagarajan Ramesh
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj, Kingdom of Saudi Arabia
| | - Mohammad Shahid
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj, Kingdom of Saudi Arabia
| |
Collapse
|
3
|
Yeon J, Kim E, Bazarragchaa B, Kim SY, Huh JY, Park H, Suh SS, Seo JB. Stellera chamaejasme L. extract inhibits adipocyte differentiation through activation of the extracellular signal-regulated kinase pathway. PLoS One 2024; 19:e0300520. [PMID: 38512891 PMCID: PMC10956757 DOI: 10.1371/journal.pone.0300520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 02/23/2024] [Indexed: 03/23/2024] Open
Abstract
Stellera chamaejasme L. (SCL) is a perennial herb with demonstrated bioactivities against inflammation and metabolic dysfunction. Adipocyte differentiation is a critical regulator of metabolic homeostasis and a promising target for the treatment of metabolic diseases, so we examined the effects of SCL on adipogenesis. A methanol extract of SCL dose-dependently suppressed intracellular lipid accumulation in adipocyte precursors cultured under differentiation induction conditions and reduced expression of the adipogenic transcription factors PPARγ and C/EBPα as well as the downstream lipogenic genes fatty acid binding protein 4, adiponectin, fatty acid synthase, and stearoyl-CoA desaturase. The extract also promoted precursor cell proliferation and altered expression of the cell cycle regulators cyclin-dependent kinase 4, cyclin E, and cyclin D1. In addition, SCL extract stimulated extracellular signal-regulated kinase (ERK) phosphorylation, while pharmacological inhibition of ERK effectively blocked the inhibitory effects of SCL extract on preadipocyte differentiation. These results suggest that SCL extract contains bioactive compounds that can suppress adipogenesis through modulation of the ERK pathway.
Collapse
Affiliation(s)
- Jaegoo Yeon
- Department of Biosciences, Mokpo National University, Jeonnam, Republic of Korea
| | - Eunbin Kim
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, Biomedical and Healthcare Research Institute, Mokpo National University, Jeonnam, Republic of Korea
| | | | - Soo-Yong Kim
- International Biological Material Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Jin Young Huh
- Department of Life Science, Sogang University, Seoul, Republic of Korea
| | - Hyuntae Park
- Department of Obstetrics & Gynecology, Korea University College of Medicine, Seoul, Republic of Korea
| | - Sung-Suk Suh
- Department of Biosciences, Mokpo National University, Jeonnam, Republic of Korea
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, Biomedical and Healthcare Research Institute, Mokpo National University, Jeonnam, Republic of Korea
| | - Jong Bae Seo
- Department of Biosciences, Mokpo National University, Jeonnam, Republic of Korea
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, Biomedical and Healthcare Research Institute, Mokpo National University, Jeonnam, Republic of Korea
| |
Collapse
|
4
|
Heimann M, Elashry MI, Klymiuk MC, Eldaey A, Wenisch S, Arnhold S. Optimizing the Adipogenic Induction Protocol Using Rosiglitazone Improves the Physiological Parameters and Differentiation Capacity of Adipose Tissue-Derived Mesenchymal Stem Cells for Horses, Sheep, Dogs, Murines, and Humans. Animals (Basel) 2023; 13:3224. [PMID: 37893949 PMCID: PMC10603751 DOI: 10.3390/ani13203224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 09/28/2023] [Accepted: 10/03/2023] [Indexed: 10/29/2023] Open
Abstract
The investigation of adipose tissue-derived mesenchymal stem cells (ASCs) has received considerable interest in regenerative medicine. A nontoxic adipogenic induction protocol valid for cells of different mammalian species has not been described. This study aims to establish an adipogenic differentiation protocol suitable for horses, sheep, dogs, murines, and human cells. An optimized rosiglitazone protocol, consisting of 5% fetal calf serum in Dulbecco's Modified Eagle's Medium, 10 μg/mL insulin, 0.55 μg/mL transferrin, 6.8 ng sodium selenite, 1 μM dexamethasone, and 1-5 μM of rosiglitazone, is compared to the 3-isobutyl-1-methylxantine (IBMX) protocol, where rosiglitazone was replaced with 0.5 mM IBMX and 0.2 mM indomethacin. Cell viability, cytotoxicity, a morphometric analysis of the lipid, and the expression of adipogenic markers for 14 days were assessed. The data revealed that using 5 µM of rosiglitazone promotes the adipogenic differentiation capacity in horse, sheep, and dog cells compared to IBMX induction. Meanwhile, marked reductions in the cell viability and cell number with the IBMX protocol were detected, and rosiglitazone increased the cell number and lipid droplet size, prevented apoptosis, and upregulated FABP-4 and Leptin expression in the cells of most of the species. Our data revealed that the rosiglitazone protocol improves the adipogenesis of ASCs, together with having less toxicity, and should be considered for cell reproducibility and clinical applications targeting obesity.
Collapse
Affiliation(s)
- Manuela Heimann
- Institute of Veterinary Anatomy, Histology and Embryology, Justus-Liebig-University of Giessen, 35392 Giessen, Germany; (M.H.); (M.C.K.); (S.A.)
| | - Mohamed I. Elashry
- Institute of Veterinary Anatomy, Histology and Embryology, Justus-Liebig-University of Giessen, 35392 Giessen, Germany; (M.H.); (M.C.K.); (S.A.)
| | - Michele C. Klymiuk
- Institute of Veterinary Anatomy, Histology and Embryology, Justus-Liebig-University of Giessen, 35392 Giessen, Germany; (M.H.); (M.C.K.); (S.A.)
| | - Asmaa Eldaey
- Clinic of Small Animals, c/o Institute of Veterinary Anatomy, Histology and Embryology, Justus-Liebig-University of Giessen, 35392 Giessen, Germany; (A.E.); (S.W.)
| | - Sabine Wenisch
- Clinic of Small Animals, c/o Institute of Veterinary Anatomy, Histology and Embryology, Justus-Liebig-University of Giessen, 35392 Giessen, Germany; (A.E.); (S.W.)
| | - Stefan Arnhold
- Institute of Veterinary Anatomy, Histology and Embryology, Justus-Liebig-University of Giessen, 35392 Giessen, Germany; (M.H.); (M.C.K.); (S.A.)
| |
Collapse
|
5
|
Park YK, Jang BC. The Receptor Tyrosine Kinase c-Met Promotes Lipid Accumulation in 3T3-L1 Adipocytes. Int J Mol Sci 2023; 24:ijms24098086. [PMID: 37175792 PMCID: PMC10179087 DOI: 10.3390/ijms24098086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/23/2023] [Accepted: 04/27/2023] [Indexed: 05/15/2023] Open
Abstract
The receptor tyrosine kinase c-Met is elaborated in embryogenesis, morphogenesis, metabolism, cell growth, and differentiation. JNJ38877605 (JNJ) is an inhibitor of c-Met with anti-tumor activity. The c-Met expression and its role in adipocyte differentiation are unknown. Here, we investigated the c-Met expression and phosphorylation, knockdown (KD) effects, and pharmacological inhibition of c-Met by JNJ on fat accumulation in murine preadipocyte 3T3-L1 cells. During 3T3-L1 preadipocyte differentiation, strikingly, c-Met expression at the protein and mRNA levels and the protein phosphorylation on Y1234/1235 and Y1349 is crucial for inducing its kinase catalytic activity and activating a docking site for signal transducers were increased in a time-dependent manner. Of note, JNJ treatment at 20 μM that strongly inhibits c-Met phosphorylation without altering its total expression resulted in less lipid accumulation and triglyceride (TG) content with no cytotoxicity. JNJ further reduced the expression of adipogenic regulators, including CCAAT/enhancer-binding protein-α (C/EBP-α), peroxisome proliferator-activated receptor-γ (PPAR-γ), fatty acid synthase (FAS), acetyl CoA carboxylase (ACC), and perilipin A. Moreover, JNJ treatment increased cAMP-activated protein kinase (AMPK) and liver kinase B-1 (LKB-1) phosphorylation but decreased ATP levels. Significantly, KD of c-Met suppressed fat accumulation and triglyceride (TG) quantity and reduced the expression of C/EBP-α, PPAR-γ, FAS, ACC, and perilipin A. Collectively, the present results demonstrate that c-Met is a novel, highly conserved mediator of adipogenesis regulating lipid accumulation in murine adipocytes.
Collapse
Affiliation(s)
- Yu-Kyoung Park
- Department of Molecular Medicine, College of Medicine, Keimyung University, 1095 Dalgubeoldaero, Dalseo-gu, Daegu 42601, Republic of Korea
- Department of Physiology, Senotherapy-Based Metabolic Disease Control Research Center, College of Medicine, Yeungnam University, 170, Hyeonchung-ro, Nam-gu, Daegu 42415, Republic of Korea
| | - Byeong-Churl Jang
- Department of Molecular Medicine, College of Medicine, Keimyung University, 1095 Dalgubeoldaero, Dalseo-gu, Daegu 42601, Republic of Korea
| |
Collapse
|
6
|
He Y, Liang Z, Wang J, Tang H, Li J, Cai J, Liao Y. Ceiling culture of human mature white adipocytes with a browning agent: A novel approach to induce transdifferentiation into beige adipocytes. Front Bioeng Biotechnol 2022; 10:905194. [PMID: 36046675 PMCID: PMC9420896 DOI: 10.3389/fbioe.2022.905194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 07/21/2022] [Indexed: 11/13/2022] Open
Abstract
Excess and dysfunctional adipose tissue plays an important role in metabolic diseases, including obesity, atherosclerosis and type 2 diabetes mellitus. In mammals, adipose tissue is categorized into two types: white and brown. Adult brown tissue is mainly composed of beige adipocytes, which dispose of stored energy as heat and have become increasingly popular as a therapeutic target for obesity. However, there is still a paucity of cell models that allow transdifferentiation of mature white adipocytes into beige adipocytes, as seen in vivo. Here, we describe a novel, ceiling culture-based model of human mature white adipocytes, which transdifferentiate into beige adipocytes under the mechanical force and hypoxia of ceiling culture. We also show that the use of rosiglitazone and rapamycin can modulate transdifferentiation, up and down regulating expression of beige adipocyte-specific genes, respectively. Rosiglitazone additionally facilitated the upregulation of fatty acid lipolysis and oxidation genes. Finally, these beige adipocytes derived from dedifferentiated adipocytes exhibited a progenitor-specific phenotype, with higher expression of mature adipocyte-specific genes than adipocyte-derived stem cells. Overall, we report a novel approach to conveniently cultivate beige adipocytes from white adipocytes in vitro, suitable for mechanistic studies of adipose biology and development of cell and drug therapies in the future.
Collapse
Affiliation(s)
| | | | | | | | | | - Junrong Cai
- *Correspondence: Junrong Cai, ; Yunjun Liao,
| | - Yunjun Liao
- *Correspondence: Junrong Cai, ; Yunjun Liao,
| |
Collapse
|
7
|
Yang Z, Qi Z, Yang X, Gao Q, Hu Y, Yuan X. Inhibition of RIP3 increased ADSC viability under OGD and modified the competency of adipogenesis, angiogenesis, and inflammation regulation. Biosci Rep 2022; 42:BSR20212808. [PMID: 35302166 PMCID: PMC8965819 DOI: 10.1042/bsr20212808] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 03/03/2022] [Accepted: 03/17/2022] [Indexed: 11/17/2022] Open
Abstract
Adipose-derived stem cells (ADSCs) showed decreased cell viability and increased cell death under oxygen-glucose deprivation (OGD). Meanwhile, vital necroptotic proteins, including receptor-interacting protein kinase (RIP) 3 (RIP3) and mixed lineage kinase domain-like pseudokinase (MLKL), were expressed in the early stage. The present study aims to explore the effect of necroptosis inhibition on ADSCs. ADSCs were obtained from normal human subcutaneous fat and verified by multidirectional differentiation and flow cytometry. By applying cell counting kit-8 (CCK-8), calcein/propidium iodide (PI) staining and immunostaining, we determined the OGD treatment time of 4 h, a timepoint when the cells showed a significant decrease in viability and increased protein expression of RIP3, phosphorylated RIP3 (pRIP3) and phosphorylated MLKL (pMLKL). After pretreatment with the inhibitor of RIP3, necroptotic protein expression decreased under OGD conditions, and cell necrosis decreased. Transwell assays proved that cell migration ability was retained. Furthermore, the expression of the adipogenic transcription factor peroxisome proliferator-activated receptor γ (PPARγ) and quantitative analysis of Oil Red O staining increased in the inhibitor group. The expression of vascular endothelial growth factor-A (VEGFA) and fibroblast growth factor 2 (FGF2) and the migration test suggest that OGD increases the secretion of vascular factors, promotes the migration of human umbilical vein endothelial cells (HUVECs), and forms unstable neovascularization. ELISA revealed that inhibition of RIP3 increased the secretion of the anti-inflammatory factor, interleukin (IL)-10 (IL-10) and reduced the expression of the proinflammatory factor IL-1β. Inhibition of RIP3 can reduce the death of ADSCs, retain their migration ability and adipogenic differentiation potential, reduce unstable neovascularization and inhibit the inflammatory response.
Collapse
Affiliation(s)
- Zhenyu Yang
- Chinese Academy of Medical Sciences and Peking Union Medical College Plastic Surgery Hospital and Institute, Beijing, China
| | - Zuoliang Qi
- Chinese Academy of Medical Sciences and Peking Union Medical College Plastic Surgery Hospital and Institute, Beijing, China
| | - Xiaonan Yang
- Chinese Academy of Medical Sciences and Peking Union Medical College Plastic Surgery Hospital and Institute, Beijing, China
| | - Qiuni Gao
- Chinese Academy of Medical Sciences and Peking Union Medical College Plastic Surgery Hospital and Institute, Beijing, China
| | - Yuling Hu
- Chinese Academy of Medical Sciences and Peking Union Medical College Plastic Surgery Hospital and Institute, Beijing, China
| | - Xihang Yuan
- Chinese Academy of Medical Sciences and Peking Union Medical College Plastic Surgery Hospital and Institute, Beijing, China
| |
Collapse
|
8
|
Lamichhane G, Pandeya PR, Lamichhane R, Rhee SJ, Devkota HP, Jung HJ. Anti-Obesity Potential of Ponciri Fructus: Effects of Extracts, Fractions and Compounds on Adipogenesis in 3T3-L1 Preadipocytes. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27030676. [PMID: 35163941 PMCID: PMC8839251 DOI: 10.3390/molecules27030676] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/11/2022] [Accepted: 01/17/2022] [Indexed: 12/17/2022]
Abstract
BACKGROUND Ponciri Fructus, a crude drug consisting of the dried immature fruits of Poncirus trifoliata (L.) Raf., is a popular folk medicine used for the treatment of allergy and gastrointestinal disorders in Korea and China. In this study, the anti-adipogenic activity of extracts and isolated compounds were evaluated using 3T3-L1 preadipocytes. METHODS Dried immature fruits were extracted and fractionated into n-hexane, ethyl acetate (EtOAc), n-butanol and water-soluble fractions. The ethanol extract and fractions were tested for anti-adipogenic activity in the 3T3-L1 cell line. The active fractions (n-hexane and EtOAc fractions) were further subjected to chromatographic techniques to isolate and identify active compounds. Furthermore, the isolated compounds were evaluated for their anti-adipogenic activity. RESULTS Altogether, seven compounds, including two flavonoids, one phytosteroid and four coumarin derivatives, were isolated. Ethanol extract, n-hexane fraction, EtOAc fraction and three isolated compounds (phellopterin, oxypeucedanin and poncirin) showed significant anti-adipogenic activity as observed by reduced lipid deposition in differentiated 3T3-L1 cells. Further, oxypeucedanin downregulated the key adipogenic markers, such as peroxisome proliferator-activated receptors proteins γ (PPAR-γ), sterol response element binding proteins-1 (SREBP-1), CCAAT/enhancer binding proteins-α (C/EBP-α), adipocyte-specific lipid binding proteins (FABP-4), adipocyte fatty acid binding proteins (aP2), lipoprotein lipase (LPL) and leptin. CONCLUSION This study indicated that the ethanol extract, hexane fraction and ethyl acetate fraction of P. trifoliata fruits possess strong anti-adipogenic activity, containing the active compounds such as phellopterin, oxypeucedanin and poncirin. Further research is recommended to explore their efficacy and safety in animal and clinical models.
Collapse
Affiliation(s)
- Gopal Lamichhane
- Department of Oriental Pharmacy and Wonkwang-Oriental Medicines Research Institute, Wonkwang University, Iksan 570-749, Korea; (G.L.); (P.R.P.); (R.L.)
| | - Prakash Raj Pandeya
- Department of Oriental Pharmacy and Wonkwang-Oriental Medicines Research Institute, Wonkwang University, Iksan 570-749, Korea; (G.L.); (P.R.P.); (R.L.)
| | - Ramakanta Lamichhane
- Department of Oriental Pharmacy and Wonkwang-Oriental Medicines Research Institute, Wonkwang University, Iksan 570-749, Korea; (G.L.); (P.R.P.); (R.L.)
| | - Su-jin Rhee
- Department of Pharmacy, College of Pharmacy, Wonkwang University, Iksan 570-749, Korea;
| | - Hari Prasad Devkota
- Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo ku, Kumamoto 862-0973, Japan;
| | - Hyun-Ju Jung
- Department of Oriental Pharmacy and Wonkwang-Oriental Medicines Research Institute, Wonkwang University, Iksan 570-749, Korea; (G.L.); (P.R.P.); (R.L.)
- Correspondence: ; Tel.: +82-63-850-6814
| |
Collapse
|
9
|
Tang Y, Zhang W, Sheng T, He X, Xiong X. Overview of the molecular mechanisms contributing to the formation of cancer‑associated adipocytes (Review). Mol Med Rep 2021; 24:768. [PMID: 34490479 PMCID: PMC8430316 DOI: 10.3892/mmr.2021.12408] [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: 05/10/2021] [Accepted: 08/24/2021] [Indexed: 12/30/2022] Open
Abstract
Adipocytes are the main stromal cells in the tumor microenvironment. In addition to serving as energy stores for triglycerides, adipocytes may function as an active endocrine organ. The crosstalk between adipocytes and cancer cells was shown to promote the migration, invasion and proliferation of cancer cells and to cause phenotypic and functional changes in adipocytes. Tumor-derived soluble factors, such as TNF-α, plasminogen activator inhibitor 1, Wnt3a, IL-6, and exosomal microRNAs (miRNA/miRs), including miR-144, miR-126, miR-155, as well as other miRNAs, have been shown to act on adipocytes at the tumor invasion front, resulting in the formation of cancer-associated adipocytes (CAAs) with diminished reduced terminal differentiation markers and a dedifferentiated phenotype. In addition, the number and size of CAA lipid droplets have been found to be significantly reduced compared with those of mature adipocytes, whereas inflammatory cytokines and proteases are overexpressed. The aim of the present review was to summarize the latest findings on the biological changes of CAAs and the potential role of tumor-adipocyte crosstalk in the formation of CAAs, in the hope of providing novel perspectives for breast cancer treatment.
Collapse
Affiliation(s)
- Yunpeng Tang
- Second Clinical Medical School, School of Basic Medical Sciences, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Wenkai Zhang
- Second Clinical Medical School, School of Basic Medical Sciences, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Tianqiang Sheng
- Second Clinical Medical School, School of Basic Medical Sciences, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xi He
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xiangyang Xiong
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
10
|
Pomegranate flower extract bidirectionally regulates the proliferation, differentiation and apoptosis of 3T3-L1 cells through regulation of PPARγ expression mediated by PI3K-AKT signaling pathway. Biomed Pharmacother 2020; 131:110769. [DOI: 10.1016/j.biopha.2020.110769] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 08/31/2020] [Accepted: 09/17/2020] [Indexed: 12/13/2022] Open
|
11
|
DeBari MK, Abbott RD. Adipose Tissue Fibrosis: Mechanisms, Models, and Importance. Int J Mol Sci 2020; 21:ijms21176030. [PMID: 32825788 PMCID: PMC7503256 DOI: 10.3390/ijms21176030] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 08/15/2020] [Accepted: 08/17/2020] [Indexed: 02/06/2023] Open
Abstract
Increases in adipocyte volume and tissue mass due to obesity can result in inflammation, further dysregulation in adipose tissue function, and eventually adipose tissue fibrosis. Like other fibrotic diseases, adipose tissue fibrosis is the accumulation and increased production of extracellular matrix (ECM) proteins. Adipose tissue fibrosis has been linked to decreased insulin sensitivity, poor bariatric surgery outcomes, and difficulty in weight loss. With the rising rates of obesity, it is important to create accurate models for adipose tissue fibrosis to gain mechanistic insights and develop targeted treatments. This article discusses recent research in modeling adipose tissue fibrosis using in vivo and in vitro (2D and 3D) methods with considerations for biomaterial selections. Additionally, this article outlines the importance of adipose tissue in treating other fibrotic diseases and methods used to detect and characterize adipose tissue fibrosis.
Collapse
Affiliation(s)
- Megan K. DeBari
- Department of Materials Science and Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA;
| | - Rosalyn D. Abbott
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
- Correspondence:
| |
Collapse
|
12
|
Malodobra-Mazur M, Cierzniak A, Dobosz T. Oleic acid influences the adipogenesis of 3T3-L1 cells via DNA Methylation and may predispose to obesity and obesity-related disorders. Lipids Health Dis 2019; 18:230. [PMID: 31883537 PMCID: PMC6935146 DOI: 10.1186/s12944-019-1173-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 12/09/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Adipogenesis is the process of adipocytes formation from unspecialized progenitor cells called mesenchymal stromal cells. Numerous mechanisms including epigenetic regulation modulate the correct progress of this process. Dietary exposures occurring over a specific period of time might cause long-lasting and even permanent changes in gene expression regulated by epigenetic mechanisms. For that reason, we investigated the adipogenesis of 3 T3-L1 cells with the excess of saturated and monounsaturated fatty acids and their influence on global and site-specific DNA methylation in these cells. MATERIALS AND METHODS 3T3-L1 cells were cultured in vitro to obtain 100% of confluence, then the adipogenesis was induced by a differentiation cocktail with the addition of the excess of 0.25 mM and 0.5 mM of palmitic (16:0), stearic (18:0) and oleic (18:1n-9) acids. DNA and RNA were extracted at five-time points to assess the adipogenesis process. The phenotype of mature adipocytes (insulin sensitivity, adipokines secretion, fat content) was estimated in fully mature adipocytes. DNA methylation was investigated both during adipogenesis and in mature adipocytes. RESULTS Oleic acids stimulated expression of C/ebpα and Pparγ, which was correlated with lower methylation levels at promoters sites. Furthermore, cells cultured with an excess of oleic acid were characterized by higher lipid accumulation rate, higher leptin, and lower adiponectin secretion. Moreover, in all experimental cells, insulin signaling and glucose utilization were impaired. CONCLUSION Oleic acid affected the methylation of Pparγ and C/ebpα promoters, what correlated with higher expression. Furthermore, examined free fatty acids influenced the phenotype of mature adipocytes, especially insulin signaling pathway and adipokine secretion.
Collapse
Affiliation(s)
- Malgorzata Malodobra-Mazur
- Department of Forensic Medicine, Molecular Techniques Unit, Wroclaw Medical University, Sklodowskiej-Curie 52, 50-369, Wroclaw, Poland.
| | - Aneta Cierzniak
- Department of Forensic Medicine, Molecular Techniques Unit, Wroclaw Medical University, Sklodowskiej-Curie 52, 50-369, Wroclaw, Poland
| | - Tadeusz Dobosz
- Department of Forensic Medicine, Molecular Techniques Unit, Wroclaw Medical University, Sklodowskiej-Curie 52, 50-369, Wroclaw, Poland
| |
Collapse
|
13
|
Morita-Takemura S, Wanaka A. Blood-to-brain communication in the hypothalamus for energy intake regulation. Neurochem Int 2019; 128:135-142. [DOI: 10.1016/j.neuint.2019.04.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 04/08/2019] [Accepted: 04/11/2019] [Indexed: 01/03/2023]
|
14
|
Duru K, Kovaleva E, Danilova I, van der Bijl P, Belousova A. The potential beneficial role of isoflavones in type 2 diabetes mellitus. Nutr Res 2018; 59:1-15. [DOI: 10.1016/j.nutres.2018.06.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 06/22/2018] [Accepted: 06/24/2018] [Indexed: 01/07/2023]
|
15
|
Lv Z, Xing K, Li G, Liu D, Guo Y. Dietary Genistein Alleviates Lipid Metabolism Disorder and Inflammatory Response in Laying Hens With Fatty Liver Syndrome. Front Physiol 2018; 9:1493. [PMID: 30405443 PMCID: PMC6207982 DOI: 10.3389/fphys.2018.01493] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 10/03/2018] [Indexed: 01/21/2023] Open
Abstract
This study investigated the molecular mechanism underlying the effect of dietary genistein (GEN) on fatty liver syndrome (FLS) in laying hens. Hens in the control group (CG) were fed a high-energy and low-choline (HELC) diet to establish the FLS model. The livers of the FLS hens were friable and swollen from hemorrhage. Hepatic steatosis and inflammatory cell infiltration were present around the liver blood vessels. Hens in the low-genistein (LGE) and high-genistein (he) groups were fed GEN at 40 and 400 mg/kg doses, respectively, as supplements to the HELC diet. GEN at 40 mg/kg significantly increased gonadotropin-releasing hormone (GnRH) mRNA expression in the hypothalamus, the serum estrogen (E2) level, and the laying rate, whereas 400 mg/kg of GEN decreased GnRH expression and the laying rate without significantly affecting E2, suggesting that high-dose GEN adversely affected the reproductive performance. Either high- or low-dose GEN treatment could alleviate metabolic disorders and inflammatory responses in FLS hens. GEN significantly decreased the serum ALT, creatinine, triglyceride (TG), total cholesterol (TC), and free fatty acid (FFA) levels. Accordingly, the TG and long-chain fatty acid (LCFA) levels, including long-chain saturated fatty acids (LSFAs) and monounsaturated fatty acids (MUFAs), and the n-6:n-3 polyunsaturated fatty acid (PUFA) ratio in the liver were reduced after the GEN treatments, whereas the levels of C22:0, n-3 family fatty acids, C20:3n6, and C20:4n6 were increased. These results indicated that dietary GEN downregulated the expression of genes related to fatty acid synthesis [sterol regulatory element-binding protein 1 (SREBP1c), liver X receptor alpha (LXRα), fatty acid synthase (FAS), and acetyl coenzyme A synthetase (ACC)] and the fatty acid transporter (FAT). Furthermore, GEN treatments upregulated the transcription of genes related to fatty acid β-oxidation [peroxisome proliferator-activated receptor (PPAR)α, PPARδ, ACOT8, ACAD8, and ACADs] in the liver and reduced PPARγ and AFABP expression in abdominal fat. Dietary GEN alleviated inflammatory cell infiltration in the livers of FLS hens and downregulated TNF-α, IL-6, and IL-1β expression. Moreover, GEN treatment increased SOD activity and decreased malondialdehyde activity in the liver. In conclusion, GEN supplementation in the feed inhibited fatty acid synthesis and enhanced β-oxidation in the liver through the PPAR-ACAD/ACOT and PPAR-LXRα-SREBP1c-ACC/FAS/FAT pathways. Dietary GEN alleviated metabolic disorder and inflammation in the FLS hens by improving the antioxidant capacity and fatty acid profile.
Collapse
Affiliation(s)
- Zengpeng Lv
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Kun Xing
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Guang Li
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Dan Liu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yuming Guo
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
16
|
Shafei AES, Nabih ES, Shehata KA, Abd Elfatah ESM, Sanad ABA, Marey MY, Hammouda AAMA, Mohammed MMM, Mostafa R, Ali MA. Prenatal Exposure to Endocrine Disruptors and Reprogramming of Adipogenesis: An Early-Life Risk Factor for Childhood Obesity. Child Obes 2018; 14:18-25. [PMID: 29019419 DOI: 10.1089/chi.2017.0180] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Obesity is a global health problem. It is characterized by excess adipose tissue that results from either increase in the number of adipocytes or increase in adipocytes size. Adipocyte differentiation is a highly regulated process that involves the activation of several transcription factors culminating in the removal of adipocytes from the cell cycle and induction of highly specific proteins. Several other factors, including hormones, genes, and epigenetics, are among the most important triggers of the differentiation process. Although the main contributing factors to obesity are high caloric intake, a sedentary lifestyle, and genetic predisposition, strong evidence supports a role for life exposure to environmental pollutants. Endocrine-disrupting chemicals are exogenous, both natural and man-made, chemicals that disrupt the body signaling processes, thus interfering with the endocrine system. Several studies have shown that prenatal exposure to endocrine disruptors modulates the mechanisms, by which multipotent mesenchymal stem cells differentiate into adipocytes. This review discusses adipocytes differentiation and highlights the possible mechanisms of prenatal exposure to endocrine disruptors in reprogramming of adipogenesis and induction of obesity later in life. Therefore, this review provides knowledge that reduction of early life exposure to these chemicals could open the door for new strategies in the prevention of obesity, especially during childhood.
Collapse
Affiliation(s)
- Ayman El-Sayed Shafei
- 1 Department of Biomedical Research, Armed Forces College of Medicine , Cairo, Egypt
| | - Enas Samir Nabih
- 2 Department of Medical Biochemistry, Faculty of Medicine, Ain Shams University , Cairo, Egypt
| | | | | | | | | | | | | | - Randa Mostafa
- 1 Department of Biomedical Research, Armed Forces College of Medicine , Cairo, Egypt
| | - Mahmoud A Ali
- 1 Department of Biomedical Research, Armed Forces College of Medicine , Cairo, Egypt
| |
Collapse
|
17
|
Park YK, Obiang-Obounou BW, Lee J, Lee TY, Bae MA, Hwang KS, Lee KB, Choi JS, Jang BC. Anti-Adipogenic Effects on 3T3-L1 Cells and Zebrafish by Tanshinone IIA. Int J Mol Sci 2017; 18:ijms18102065. [PMID: 28953247 PMCID: PMC5666747 DOI: 10.3390/ijms18102065] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 09/15/2017] [Accepted: 09/22/2017] [Indexed: 12/22/2022] Open
Abstract
Tanshinone IIA is a diterpene quinone isolated from the roots of Salviamiltiorrhiza bunge that has traditionally been used in China for the treatment of cardiovascular and cerebrovascular disorders. Although there is recent evidence showing that tanshinone IIA has an anti-obesity effect, its underlying mechanism of anti-obesity effect is poorly understood. Here, we investigated the effect of tanshinone IIA on lipid accumulation in 3T3-L1 preadipocytes and zebrafish. Notably, tanshinone IIA at 10 μM concentration greatly reduced lipid accumulation and triglyceride (TG) contents during 3T3-L1 preadipocyte differentiation, suggesting its anti-adipogenic effect. On mechanistic levels, tanshinone IIA reduced the expression levels of CCAAT/enhancer-binding protein-α (C/EBP-α), peroxisome proliferator-activated receptor-γ (PPAR-γ), fatty acid synthase (FAS), and perilipin A but also the phosphorylation levels of signal transducer and activator of transcription-3/5 (STAT-3/5) in differentiating 3T3-L1 cells. In addition, tanshinone IIA strongly inhibited leptin and resistin mRNA expression in differentiating 3T3-L1 cells. Importantly, the tanshinone IIA's lipid-reducing effect was also seen in zebrafish. In sum, these findings demonstrate that tanshinone IIA has anti-adipogenic effects on 3T3-L1 cells and zebrafish, and its anti-adipogenic effect on 3T3-L1 cells is largely attributable to the reduced expression and/or phosphorylation levels of C/EBP-α, PPAR-γ, FAS, perilipin A, and STAT-3/5.
Collapse
Affiliation(s)
- Yu-Kyoung Park
- Department of Molecular Medicine, College of Medicine, Keimyung University, 1095 Dalgubeoldaero, Dalseo-gu, Daegu 42601, Korea.
| | - Brice Wilfried Obiang-Obounou
- Department of Food Nutrition, College of Natural Sciences, Keimyung University, 1095 Dalgubeoldaero, Dalseo-gu, Daegu 42601, Korea.
| | - Jinho Lee
- Department of Chemistry, College of Natural Sciences, Keimyung University, 1095 Dalgubeoldaero, Dalseo-gu, Daegu 42601, Korea.
| | - Tae-Yun Lee
- Department of Microbiology, College of Medicine, Yeungnam University, 170 Hyeonchung-ro, Nam-gu, Daegu 42415, Korea.
| | - Myung-Ae Bae
- Bio & Drug Discovery Division, Korea Research Institute of Chemical Technology, 141 Gajeong-ro Yuseong-gu, Daejeon 34114, Korea.
| | - Kyu-Seok Hwang
- Bio & Drug Discovery Division, Korea Research Institute of Chemical Technology, 141 Gajeong-ro Yuseong-gu, Daejeon 34114, Korea.
| | - Kyung-Bok Lee
- Biological Disaster Analysis Group, Division of Convergence Biotechnology, Korea Basic Science Institute, 169-148 Gwahak-ro, Yuseong-gu, Daejeon 34133, Korea.
| | - Jong-Soon Choi
- Biological Disaster Analysis Group, Division of Convergence Biotechnology, Korea Basic Science Institute, 169-148 Gwahak-ro, Yuseong-gu, Daejeon 34133, Korea.
| | - Byeong-Churl Jang
- Department of Molecular Medicine, College of Medicine, Keimyung University, 1095 Dalgubeoldaero, Dalseo-gu, Daegu 42601, Korea.
| |
Collapse
|
18
|
Mo D, Yu K, Chen H, Chen L, Liu X, He Z, Cong P, Chen Y. Transcriptome Landscape of Porcine Intramuscular Adipocytes during Differentiation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2017; 65:6317-6328. [PMID: 28673084 DOI: 10.1021/acs.jafc.7b02039] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The adipocyte differentiation process, controlled by a tightly regulated transcriptional cascade, contributes partly to determine intramuscular adipose tissue (IMAT) mass, which is associated with meat quality in food animals, as well as obesity and related metabolic complications in human. Thus, this study aimed to characterize genes critical for intramuscular preadipocyte differentiation. Primary intramuscular preadipocytes were isolated from pigs, and mRNA profiles were performed at several key points (0 h, 4 h, 8 h, 1 day, 2 days, and 6 days) during adipogenesis using microarrays. By gene functional analysis, we identified numerous differentially expressed genes among distinct stages of intramuscular preadipocyte differentiation, which included numbers of transcription factors in the early stages. We obtained 4 clusters of differential gene expression pattern, including crucial candidate genes associated with adipogenesis of intramuscular adipocytes. Further, we demonstrated that POSTN and FGFR4 suppressed, whereas AKR1CL1 promoted, the expression of adipogenic marker PPARγ and C/EBPα. Taken together, our data delineated the transcriptome landscape during porcine intramuscular preadipocyte differentiation, which provided a valuable resource for finding the genes responsible for IMAT formation.
Collapse
Affiliation(s)
- Delin Mo
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University , Guangzhou 510006, China
| | - Kaifan Yu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University , Guangzhou 510006, China
| | - Hu Chen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University , Guangzhou 510006, China
| | - Luxi Chen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University , Guangzhou 510006, China
| | - Xiaohong Liu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University , Guangzhou 510006, China
| | - Zuyong He
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University , Guangzhou 510006, China
| | - Peiqing Cong
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University , Guangzhou 510006, China
| | - Yaosheng Chen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University , Guangzhou 510006, China
| |
Collapse
|
19
|
Hosney M, Sabet S, El-Shinawi M, Gaafar KM, Mohamed MM. Leptin is overexpressed in the tumor microenvironment of obese patients with estrogen receptor positive breast cancer. Exp Ther Med 2017; 13:2235-2246. [PMID: 28565832 PMCID: PMC5443182 DOI: 10.3892/etm.2017.4291] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 01/26/2017] [Indexed: 12/11/2022] Open
Abstract
The present study aimed to investigate the potential role of leptin in the progression of breast cancer and the associated cell proliferation signalling pathway(s). A total of 44 female patients diagnosed with breast cancer and 24 healthy donors from Ain Shams University Hospitals (Cairo, Egypt) were enrolled in the present study. The present study assessed leptin expression in breast cancer tissues at the gene and protein level using reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and immunohistochemistry. The results demonstrate that the expression of leptin was significantly higher in tissue of breast cancer samples from obese patients than overweight and control samples (P<0.001). ELISA results indicated a significant increase (P<0.001) of leptin expression in obese patients. To investigate whether there is any difference in leptin expression between the peripheral and tumor microenvironment blood of patients with breast cancer, the concentration of leptin was assessed in plasma from both using ELISA assays. The results demonstrated a statistically significant increase in the level of leptin in plasma samples from the tumor microenvironment of obese patients with estrogen receptor positive (ER+) breast cancer, compared with peripheral plasma samples. Furthermore, the leptin gene was overexpressed in obese ER+ breast cancer tissue. RT-qPCR was also performed to assess the expression of genes involved in proliferation pathways including leptin receptor (LEPR), aromatase, mitogen activated protein kinase (MAPK) and signal transducer and activator of transcription-3 (STAT3). A positive association between leptin expression, LEPR, aromatase, MAPK and STAT3 was detected in tissue samples of patients with breast cancer. The current study concluded that leptin may enhance breast cancer progression by inducing the expression of JAK/STAT3, ERK1/2 and estrogen pathways in obese patients breast cancer.
Collapse
Affiliation(s)
- Mohamed Hosney
- Department of Zoology, Cancer Biology Research Laboratory (CBRL), Faculty of Science, Cairo University, Giza 12613, Egypt
| | - Salwa Sabet
- Department of Zoology, Cancer Biology Research Laboratory (CBRL), Faculty of Science, Cairo University, Giza 12613, Egypt
| | - Mohamed El-Shinawi
- Department of General Surgery, Faculty of Medicine, Ain Shams University, Cairo 11566, Egypt
| | - Khadiga M Gaafar
- Department of Zoology, Cancer Biology Research Laboratory (CBRL), Faculty of Science, Cairo University, Giza 12613, Egypt
| | - Mona M Mohamed
- Department of Zoology, Cancer Biology Research Laboratory (CBRL), Faculty of Science, Cairo University, Giza 12613, Egypt
| |
Collapse
|
20
|
Kobayashi S, Kawasaki Y, Takahashi T, Maeno H, Nomura M. Mechanisms for the anti-obesity actions of bofutsushosan in high-fat diet-fed obese mice. Chin Med 2017; 12:8. [PMID: 28360931 PMCID: PMC5369197 DOI: 10.1186/s13020-017-0129-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 03/04/2017] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND The Kampo medicine bofutsushosan (BTS; Pulvis ledebouriellae compositae; Fang Feng Tong Sheng San) has been used as an anti-obesity treatment in overweight patients. In this study, we assessed the underlying physiological changes induced by BTS in obese mice maintained on a high-fat diet. METHODS Male ICR mice were fed a 60% kcal fat diet for 5 weeks starting at 4 weeks of age and then fed the same diet with administration of water (control) or aqueous BTS extract (1.0-2.0 g/kg) for 25 days. Body weight, wet weight of isolated white adipose tissue, and obesity-related serum parameters (glucose, lipids, leptin, adiponectin) were measured after treatment. The mRNA expression levels of leptin, adiponectin, and UCP1 in the adipose tissues were determined by quantitative real-time polymerase chain reaction after the first 5 days of treatment. RESULTS Bofutsushosan (1.5-2.0 g/kg) significantly decreased total body weight and total wet weight of white adipose tissue isolated from subcutaneous (retroperitoneal) and visceral regions (epididymal, mesenteric, and perirenal). At 2.0 g/kg, BTS also decreased total fat mass, visceral fat mass, and ratio of fat mass to body weight as measured by computed tomography, and significantly decreased epididymal adipocyte size after 14 and 25 days' treatment. Twenty-five days' treatment lowered serum glucose, insulin, leptin, and triglycerides, and reduced homeostasis model assessment-insulin resistance. Alternatively, 2.0 g/kg BTS significantly increased mRNA levels of adiponectin, leptin, and UCP1 in interscapular brown adipose tissue but not epididymal white adipose tissue after 5 days' administration. CONCLUSION In the early administration period, BTS increased mRNA expression levels of leptin, adiponectin, and UCP1 in brown adipose tissues. With longer administration, BTS improved insulin resistance, and subsequently reduced serum levels of leptin and triglyceride in parallel with decreased visceral white adipose tissue volume and adipocyte size.
Collapse
Affiliation(s)
- Shinjiro Kobayashi
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Hokuriku University, Kanazawa, 920-1181 Japan
| | - Yuki Kawasaki
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Hokuriku University, Kanazawa, 920-1181 Japan
| | - Tatsuo Takahashi
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Hokuriku University, Kanazawa, 920-1181 Japan
| | - Hironori Maeno
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Hokuriku University, Kanazawa, 920-1181 Japan
| | - Masaaki Nomura
- Center of Clinical Pharmacy Education, Faculty of Pharmaceutical Sciences, Hokuriku University, Kanazawa, 920-1181 Japan
| |
Collapse
|
21
|
Henriksson I, Gatenholm P, Hägg DA. Increased lipid accumulation and adipogenic gene expression of adipocytes in 3D bioprinted nanocellulose scaffolds. Biofabrication 2017; 9:015022. [PMID: 28140346 DOI: 10.1088/1758-5090/aa5c1c] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Compared to standard 2D culture systems, new methods for 3D cell culture of adipocytes could provide more physiologically accurate data and a deeper understanding of metabolic diseases such as diabetes. By resuspending living cells in a bioink of nanocellulose and hyaluronic acid, we were able to print 3D scaffolds with uniform cell distribution. After one week in culture, cell viability was 95%, and after two weeks the cells displayed a more mature phenotype with larger lipid droplets than standard 2D cultured cells. Unlike cells in 2D culture, the 3D bioprinted cells did not detach upon lipid accumulation. After two weeks, the gene expression of the adipogenic marker genes PPARγ and FABP4 was increased 2.0- and 2.2-fold, respectively, for cells in 3D bioprinted constructs compared with 2D cultured cells. Our 3D bioprinted culture system produces better adipogenic differentiation of mesenchymal stem cells and a more mature cell phenotype than conventional 2D culture systems.
Collapse
Affiliation(s)
- I Henriksson
- 3D Bioprinting Center, Department of Chemistry and Chemical Engineering, Chalmers University of Technology, Göteborg, Sweden. Cellink, 470 Ramona Street, Palo Alto 94391, CA, United States of America
| | | | | |
Collapse
|
22
|
Wu M, Liu D, Zeng R, Xian T, Lu Y, Zeng G, Sun Z, Huang B, Huang Q. Epigallocatechin-3-gallate inhibits adipogenesis through down-regulation of PPARγ and FAS expression mediated by PI3K-AKT signaling in 3T3-L1 cells. Eur J Pharmacol 2016; 795:134-142. [PMID: 27940057 DOI: 10.1016/j.ejphar.2016.12.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 11/30/2016] [Accepted: 12/05/2016] [Indexed: 01/09/2023]
Abstract
Epigallocatechin-3-gallate (EGCG), a major component in green tea, functions as extensive bioactivities including anti-inflammation, anti-oxidation, and anti-cancer. However, little is known about its anti-adipogenesis and underlying mechanisms. The purport of this study sought to investigate effects of EGCG on 3T3-L1 preadipocyte differentiation and to explore its possible mechanisms. The 3T3-L1 cells were induced to differentiate under the condition of pro-adipogenic cocktail with or without indicated EGCG concentrations (10, 50, 100, 200µM) for 2, 4, 6 and 8 days, respectively. Also, another batch of 3T3-L1 cells was induced under the optimal EGCG concentration (100µM) with or without SC3036 (PI3K activator, 10µM) or SC79 (AKT activator, 0.5µM) for 8 days. Subsequently, the cell viability was examined by MTT assay and the cell morphology was visualized by Oil red O staining. Finally, the mRNA levels including peroxisome proliferator activated receptor γ (PPARγ) and fatty acid synthase (FAS) were detected by quantitative real time PCR, while the protein levels of PPARγ, FAS, phosphatidylinositol 3 kinase (PI3K), insulin receptor substrate1(IRS1), AKT, and p-AKT were measured by immunoblotting analysis. Our results showed that EGCG inhibited adipogenesis of 3T3-L1 preadipocyte in a concentration-dependent manner. Moreover, the inhibitory effects were reversed by SC3036 or SC79, suggesting that the inhibitory effects of EGCG are mediated by PI3K-AKT signaling to down-regulate PPARγ and FAS expression levels. The findings shed light on EGCG anti-adipogenic effects and its underlying mechanism and provide a novel preventive-therapeutic potential for obesity subjects as a compound from Chinese green tea.
Collapse
Affiliation(s)
- Mengqing Wu
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, Jiangxi Province, P.R. China; Department of Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang 330006, Jiangxi Province, P.R. China
| | - Dan Liu
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, Jiangxi Province, P.R. China; Department of Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang 330006, Jiangxi Province, P.R. China
| | - Rong Zeng
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, Jiangxi Province, P.R. China; Department of Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang 330006, Jiangxi Province, P.R. China
| | - Tao Xian
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, Jiangxi Province, P.R. China; Department of Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang 330006, Jiangxi Province, P.R. China
| | - Yi Lu
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, Jiangxi Province, P.R. China; Department of Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang 330006, Jiangxi Province, P.R. China
| | - Guohua Zeng
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, Jiangxi Province, P.R. China; Department of Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang 330006, Jiangxi Province, P.R. China
| | - Zhangzetian Sun
- Jiangxi Medical School, Nanchang University, Nanchang 330006, Jiangxi Province, P.R. China
| | - Bowei Huang
- Jiangxi Medical School, Nanchang University, Nanchang 330006, Jiangxi Province, P.R. China
| | - Qiren Huang
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, Jiangxi Province, P.R. China; Department of Pharmacology, School of Pharmaceutical Science, Nanchang University, Nanchang 330006, Jiangxi Province, P.R. China.
| |
Collapse
|
23
|
Choi J, Lee SY, Yoo YM, Kim CH. Maturation of Adipocytes is Suppressed by Fluid Shear Stress. Cell Biochem Biophys 2016; 75:87-94. [PMID: 27830366 DOI: 10.1007/s12013-016-0771-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 10/31/2016] [Indexed: 10/20/2022]
Abstract
Preadipocytes are mechano-responsive cells and their differentiation to adipocytes may be regulated by various types of physical stimulation. Understanding the mechanism of differentiation, which increases the number of adipocytes and lipid accumulation is important in the study of obesity-related diseases. In this study, we investigated the effects of physical stimulation at different stages of adipogenic differentiation using physiological levels of fluid shear stress. Preadipocytes were treated with dexamethasone, 3-isobutyl-1-methylxanthine and insulin for 3 days (induction period) and incubated for additional 6 days for maturation. Fluid shear stress of 1 Pa at 1 Hz was applied for 1 h at different stages of differentiation. Fluid shear stress applied at the maturation period significantly reduced the expressions of C/enhancer binding protein (EBP)α and peroxisome proliferator-activated receptor (PPAR)γ2 leading to reduced lipid accumulation. Fluid shear stress applied at the early or late stages of the induction period only decreased peroxisome proliferator-activated receptor γ2 expression without any significant changes in lipid accumulation. Stimulation at multiple days during the induction period did not result in changes in lipid accumulation compared to stimulation at a single day. These results suggest that lipid droplet accumulation is effectively decreased by fluid shear stress applied during the cell maturation period. Understanding the cellular response to physical stimulation throughout the entire adipocyte differentiation period may be important in controlling adipogenesis by physical stimulation.
Collapse
Affiliation(s)
- Jongyun Choi
- Department of Biomedical Engineering, College of Health Science, Yonsei University, Wonju, Gangwon-do, 26493, Republic of Korea
| | - Sei Young Lee
- Department of Biomedical Engineering, College of Health Science, Yonsei University, Wonju, Gangwon-do, 26493, Republic of Korea
| | - Yeong-Min Yoo
- Department of Biomedical Engineering, College of Health Science, Yonsei University, Wonju, Gangwon-do, 26493, Republic of Korea
| | - Chi Hyun Kim
- Department of Biomedical Engineering, College of Health Science, Yonsei University, Wonju, Gangwon-do, 26493, Republic of Korea.
| |
Collapse
|
24
|
Yoo SR, Lee MY, Kang BK, Shin HK, Jeong SJ. Soshiho-Tang Aqueous Extract Exerts Antiobesity Effects in High Fat Diet-Fed Mice and Inhibits Adipogenesis in 3T3-L1 Adipocytes. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2016; 2016:2628901. [PMID: 27777595 PMCID: PMC5061987 DOI: 10.1155/2016/2628901] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 08/23/2016] [Accepted: 09/01/2016] [Indexed: 01/22/2023]
Abstract
Soshiho-tang (SST; sho-saiko-to in Japanese; xiaochaihu-tang in Chinese) has generally been used to improve liver fibrosis- and cirrhosis-related symptoms in traditional Korean medicine. Although many studies have investigated the pharmacological properties of SST, its antiobesity effect has not been elucidated. Thus, our present study was carried out to evaluate the antiobesity effect of SST using a high fat diet- (HFD) induced obese mouse model and 3T3-L1 adipose cells. C57BL/6J mice were randomly divided into four groups (n = 6/group), normal diet (ND), HFD-fed group, and HFD- and SST-fed groups (S200: 200 mg/kg of SST; S600: 600 mg/kg of SST) and given HFD with or without SST extract for 8 weeks. 3T3-L1 preadipocytes were differentiated into adipocytes for 8 days with or without SST. In the HFD-fed obese mice, body weight and fat accumulation in adipose tissue were significantly reduced by SST administration. Compared with control-differentiated adipocytes, SST significantly inhibited lipid accumulation by decreasing the triglyceride (TG) content and leptin concentration in 3T3-L1 adipocytes. SST also decreased the expression of adipogenesis-related genes including lipoprotein lipase (LPL), fatty acid binding protein 4 (FABP4), CCAAT/enhancer-binding protein-alpha (C/EBP-α), and peroxisome proliferator-activated receptor-gamma (PPAR-γ). Our findings suggest that SST has potential as a nontoxic antiobesity medication.
Collapse
Affiliation(s)
- Sae-Rom Yoo
- K-Herb Research Center, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea
| | - Mee-young Lee
- K-Herb Research Center, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea
| | - Byoung-Kab Kang
- KM Fundamental Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea
| | - Hyeun-Kyoo Shin
- K-Herb Research Center, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea
| | - Soo-Jin Jeong
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea
- Korean Medicine Life Science, University of Science & Technology, Daejeon 34113, Republic of Korea
| |
Collapse
|
25
|
Ilavenil S, Kim DH, Vijayakumar M, Srigopalram S, Roh SG, Arasu MV, Lee JS, Choi KC. Potential role of marine algae extract on 3T3-L1 cell proliferation and differentiation: an in vitro approach. Biol Res 2016; 49:38. [PMID: 27604997 PMCID: PMC5013630 DOI: 10.1186/s40659-016-0098-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 08/22/2016] [Indexed: 01/08/2023] Open
Abstract
Background From ancient times, marine algae have emerged as alternative medicine and foods, contains the rich source of natural products like proteins, vitamins, and secondary metabolites, especially Chlorella vulgaris (C. vulgaris) contains numerous anti-inflammatory, antioxidants and wound healing substances. Type 2 diabetes mellitus is closely associated with adipogenesis and their factors. Hence, we aimed to investigate the chemical constituents and adipogenic modulatory properties of C. vulgaris in 3T3-L1 pre-adipocytes. Results We analysed chemical constituents in ethanolic extract of C. vulgaris (EECV) by LC–MS. Results revealed that the EECV contains few triterpenoids and saponin compounds. Further, the effect of EECV on lipid accumulation along with genes and proteins expressions which are associated with adipogenesis and lipogenesis were evaluated using oil red O staining, qPCR and western blot techniques. The data indicated that that EECV treatment increased differentiation and lipid accumulation in 3T3-L1 cells, which indicates positive regulation of adipogenic and lipogenic activity. These increases were associated with up-regulation of PPAR-γ2, C/EBP-α, adiponectin, FAS, and leptin mRNA and protein expressions. Also, EECV treatments increased the concentration of glycerol releases as compared with control cells. Troglitazone is a PPAR-γ agonist that stimulates the PPAR-γ2, adiponectin, and GLUT-4 expressions. Similarly, EECV treatments significantly upregulated PPAR-γ2, adiponectin, GLUT-4 expressions and glucose utilization. Further, EECV treatment decreased AMPK-α expression as compared with control and metformin treated cells. Conclusion The present research findings confirmed that the EECV effectively modulates the lipid accumulation and differentiation in 3T3-L1 cells through AMPK-α mediated signalling pathway.
Collapse
Affiliation(s)
- Soundharrajan Ilavenil
- Grassland and Forage Division, National Institute of Animal Science, RDA, Seonghwan-Eup, Cheonan-Si, Chungnam, 330-801, Republic of Korea
| | - Da Hye Kim
- The United Graduate School of Agricultural Sciences, Tottori University, Tottori-Shi, 680-8553, Japan
| | - Mayakrishnan Vijayakumar
- Grassland and Forage Division, National Institute of Animal Science, RDA, Seonghwan-Eup, Cheonan-Si, Chungnam, 330-801, Republic of Korea
| | - Srisesharam Srigopalram
- Grassland and Forage Division, National Institute of Animal Science, RDA, Seonghwan-Eup, Cheonan-Si, Chungnam, 330-801, Republic of Korea
| | - Sang Gun Roh
- Laboratory of Animal Physiology, Graduate School of Agricultural Science, Tohoku University, Aoba, Sendai, Japan
| | - Mariadhas Valan Arasu
- Department of Botany and Microbiology, Addiriyah Chair for Environmental Studies, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Jong Suk Lee
- Bio-center, Gyeonggi Institute of Science and Technology, Suwon, Republic of Korea
| | - Ki Choon Choi
- Grassland and Forage Division, National Institute of Animal Science, RDA, Seonghwan-Eup, Cheonan-Si, Chungnam, 330-801, Republic of Korea.
| |
Collapse
|
26
|
Kim EJ, Kim YK, Kim MK, Kim S, Kim JY, Lee DH, Chung JH. UV-induced inhibition of adipokine production in subcutaneous fat aggravates dermal matrix degradation in human skin. Sci Rep 2016; 6:25616. [PMID: 27161953 PMCID: PMC4861907 DOI: 10.1038/srep25616] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 04/20/2016] [Indexed: 12/18/2022] Open
Abstract
Ultraviolet (UV) exposure to the human skin reduces triglycerides contents and lipid synthesis in the subcutaneous (SC) fat. Because adiponectin and leptin are the most abundant adipokines from the SC fat, we aim to investigate how they interact with UV exposure and skin aging. The expressions of adiponectin and leptin were significantly decreased in SC fat of sun-exposed forearm skin, in comparison with that of sun-protected buttock skin of the same elderly individuals, indicating that chronic UV exposure decreases both adipokines. Acute UV irradiation also decreased the expressions of adiponectin and leptin in SC fat. The expressions of adiponectin receptor 1/2 and leptin receptor were significantly decreased in the dermis as well as in SC fat. Moreover, while exogenous adiponectin and leptin administration prevented UV- and TNF-α induced matrix metalloproteinase (MMP)-1 expression, they also increased UV- and TNF-α induced reduction of type 1 procollagen production. Silencing of adiponectin, leptin or their receptors led to an increased MMP-1 and a decreased type 1 procollagen expression, which was reversed by treatment with recombinant human adiponectin or leptin. In conclusion, UV exposure decreases the expression of adiponectin and leptin, leading to the exacerbation of photoaging by stimulating MMP-1 expression and inhibiting procollagen synthesis.
Collapse
Affiliation(s)
- Eun Ju Kim
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea.,Laboratory of Cutaneous Aging Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea.,Institute of Human-Environment Interface Biology, Seoul National University, Seoul, Republic of Korea
| | - Yeon Kyung Kim
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea.,Laboratory of Cutaneous Aging Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea.,Institute of Human-Environment Interface Biology, Seoul National University, Seoul, Republic of Korea
| | - Min-Kyoung Kim
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea.,Laboratory of Cutaneous Aging Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea.,Institute of Human-Environment Interface Biology, Seoul National University, Seoul, Republic of Korea
| | - Sungsoo Kim
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea.,Laboratory of Cutaneous Aging Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea.,Institute of Human-Environment Interface Biology, Seoul National University, Seoul, Republic of Korea
| | - Jin Yong Kim
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea.,Laboratory of Cutaneous Aging Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea.,Institute of Human-Environment Interface Biology, Seoul National University, Seoul, Republic of Korea
| | - Dong Hun Lee
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea.,Laboratory of Cutaneous Aging Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea.,Institute of Human-Environment Interface Biology, Seoul National University, Seoul, Republic of Korea
| | - Jin Ho Chung
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea.,Laboratory of Cutaneous Aging Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea.,Institute of Human-Environment Interface Biology, Seoul National University, Seoul, Republic of Korea.,Institute on Aging, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
27
|
Inhibiting DNA methylation switches adipogenesis to osteoblastogenesis by activating Wnt10a. Sci Rep 2016; 6:25283. [PMID: 27136753 PMCID: PMC4853709 DOI: 10.1038/srep25283] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 04/14/2016] [Indexed: 01/07/2023] Open
Abstract
Both adipocytes and osteoblasts share the mesodermal lineage that derives from mesenchymal stem cells. Most studies investigating the mechanisms underlying the regulation of adipogenic or osteoblastogenic development focus on transcriptional pathways; little is known about the epigenetic mechanisms in this process. We thus determined the role of 5-aza-2′-deoxycytidine (5-Aza-dC), an inhibitor of DNA methylation, in the lineage determination between adipogenesis and osteoblastogenesis. Inhibiting DNA methylation in 3T3-L1 preadipocytes by 5-Aza-dC significantly inhibited adipogenesis whereas promoted osteoblastogenesis. This dual effect of 5-Aza-dC was associated with up-regulation of Wnt10a, a key factor determining the fate of the mesenchymal lineage towards osteoblasts. Consistently, IWP-2, an inhibitor of Wnt proteins, was found to prevent the anti-adipogenic effect of 5-Aza-dC in 3T3-L1 preadipocytes and block the osteoblastogenic effect of 5-Aza-dC in ST2 mesenchymal stem cell line. Finally, the Wnt10a 5′-region is enriched with CpG sites, whose methylation levels were markedly reduced by 5-Aza-dC. Thus we conclude that inhibiting DNA methylation by 5-Aza-dC mutual-exclusively regulates the lineage determination of adipogenesis and osteoblastogenesis by demethylating Wnt10a gene and upregulating its expression. Our study defines DNA methylation as a novel mechanism underlying adipocyte and bone cell development.
Collapse
|
28
|
Kim JA, Karadeniz F, Ahn BN, Kwon MS, Mun OJ, Bae MJ, Seo Y, Kim M, Lee SH, Kim YY, Mi-Soon J, Kong CS. Bioactive quinone derivatives from the marine brown alga Sargassum thunbergii induce anti-adipogenic and pro-osteoblastogenic activities. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2016; 96:783-90. [PMID: 25720987 DOI: 10.1002/jsfa.7148] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Revised: 01/05/2015] [Accepted: 02/19/2015] [Indexed: 05/12/2023]
Abstract
BACKGROUND Health problems related to the lack of bone formation are a major problem for ageing populations in the modern world. As a part of the ongoing trend to develop natural substances that attenuate bone loss in osteoporosis, the effects of the edible brown alga Sargassum thunbergii and its active contents on adipogenic differentiation in 3T3-L1 fibroblasts and osteoblast differentiation in MC3T3-E1 pre-osteoblasts were evaluated. RESULTS Treatment with S. thunbergii significantly reduced lipid accumulation and expression of adipogenic differentiation markers such as peroxisome proliferator-activated receptor γ, CCAAT/enhancer-binding protein α and sterol regulatory element binding protein 1c. In addition, S. thunbergii successfully enhanced osteoblast differentiation as indicated by increased alkaline phosphatase activity along raised levels of osteoblastogenesis indicators, namely bone morphogenetic protein-2, osteocalcin and collagen type I. Two compounds, sargaquinoic and sargahydroquinoic acid, were isolated from active extract and shown to be active by means of osteogenesis inducement. CONCLUSION S. thunbergii could be a source for functional food ingredients for improved treatment of osteoporosis and obesity.
Collapse
Affiliation(s)
- Jung-Ae Kim
- Department of Food and Nutrition, College of Medical and Life Science, Silla University, Busan 617-736, Republic of Korea
- Marine Biotechnology Center for Pharmaceuticals and Foods, Silla University, Busan 617-736, Republic of Korea
| | - Fatih Karadeniz
- Marine Biotechnology Center for Pharmaceuticals and Foods, Silla University, Busan 617-736, Republic of Korea
| | - Byul-Nim Ahn
- Department of Organic Material Science and Engineering, Pusan National University, Busan, Republic of Korea
| | - Myeong Sook Kwon
- Department of Food and Nutrition, College of Medical and Life Science, Silla University, Busan 617-736, Republic of Korea
| | - Ok-Ju Mun
- Department of Food and Nutrition, College of Medical and Life Science, Silla University, Busan 617-736, Republic of Korea
| | - Min Joo Bae
- Department of Food and Nutrition, College of Medical and Life Science, Silla University, Busan 617-736, Republic of Korea
| | - Youngwan Seo
- Marine Environment and Bioscience, Korea Maritime University, Busan 606-791, Republic of Korea
- Ocean Science & Technology School, Korea Marine University, Busan 606-791, Republic of Korea
| | - Mihyang Kim
- Department of Food and Nutrition, College of Medical and Life Science, Silla University, Busan 617-736, Republic of Korea
| | - Sang-Hyeon Lee
- Bioscience and Biotechnology Department, Graduate School, Silla University, Busan 617-736, Republic of Korea
| | - Yuck Yong Kim
- IS Food Co., Marine Bio-industry Department Center, Busan 619-912, Republic of Korea
| | - Jang Mi-Soon
- National Fisheries Research & Development Institute, Food and Safety Research Center, Busan 619-705, Republic of Korea
| | - Chang-Suk Kong
- Department of Food and Nutrition, College of Medical and Life Science, Silla University, Busan 617-736, Republic of Korea
| |
Collapse
|
29
|
Park YK, Hong VS, Lee TY, Lee J, Choi JS, Park DS, Park GY, Jang BC. The novel anti-adipogenic effect and mechanisms of action of SGI-1776, a Pim-specific inhibitor, in 3T3-L1 adipocytes. Int J Mol Med 2015; 37:157-64. [PMID: 26719859 DOI: 10.3892/ijmm.2015.2415] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 10/27/2015] [Indexed: 11/06/2022] Open
Abstract
The proviral integration site for moloney murine leukemia virus (Pim) kinases, consisting of Pim-1, Pim-2 and Pim-3, belongs to a family of serine/threonine kinases that are involved in controlling cell growth and differentiation. Pim kinases are emerging as important mediators of adipocyte differentiation. SGI-1776, an inhibitor of Pim kinases, is widely used to assess the physiological roles of Pim kinases, particularly cell functions. In the present study, we examined the effects of SGI-1776 on adipogenesis. The anti‑adipogenic effect of SGI‑1776 was measured by Oil Red O staining and AdipoRed assays. The effect of SGI‑1776 on the growth of 3T3‑L1 adipocytes was determined by cell count analysis. The effects of SGI‑1776 on the protein and mRNA expression of adipogenesis-related proteins and adipokines in 3T3‑L1 adipocytes were also evaluated by western blot analysis and RT‑PCR, respectively. Notably, SGI-1776 markedly inhibited lipid accumulation during the differentiation of 3T3-L1 preadipocytes into adipocytes. On a mechanistic level, SGI-1776 inhibited not only the expression of CCAAT/enhancer-binding protein-α (C/EBP-α), peroxisome proliferator-activated receptor-γ (PPAR-γ) and fatty acid synthase (FAS), but also the phosphorylation of signal transducer and activator of transcription-3 (STAT-3). Moreover, SGI-1776 decreased the expression of adipokines, including the expression of leptin and regulated on activation, normal T cell expressed and secreted (RANTES) during adipocyte differentiation. These findings demonstrate that SGI-1776 inhibits adipogenesis by downregulating the expression and/or phosphorylation levels of C/EBP-α, PPAR-γ, FAS and STAT-3.
Collapse
Affiliation(s)
- Yu-Kyoung Park
- Department of Molecular Medicine, College of Medicine, Keimyung University, Daegu 704-701, Republic of Korea
| | - Victor Sukbong Hong
- Department of Chemistry, College of Natural Sciences, Keimyung University, Daegu 704-701, Republic of Korea
| | - Tae-Yoon Lee
- Department of Microbiology, College of Medicine, Yeungnam University, Daegu 705‑717, Republic of Korea
| | - Jinho Lee
- Department of Chemistry, College of Natural Sciences, Keimyung University, Daegu 704-701, Republic of Korea
| | - Jong-Soon Choi
- Division of Life Science, Korea Basic Science Institute, Daejeon 305-333, Republic of Korea
| | - Dong-Soon Park
- Department of Rehabilitation Medicine, Catholic University of Daegu School of Medicine, Daegu 705-718, Republic of Korea
| | - Gi-Young Park
- Department of Rehabilitation Medicine, Catholic University of Daegu School of Medicine, Daegu 705-718, Republic of Korea
| | - Byeong-Churl Jang
- Department of Molecular Medicine, College of Medicine, Keimyung University, Daegu 704-701, Republic of Korea
| |
Collapse
|
30
|
Leptin produced by obese adipose stromal/stem cells enhances proliferation and metastasis of estrogen receptor positive breast cancers. Breast Cancer Res 2015; 17:112. [PMID: 26286584 PMCID: PMC4541745 DOI: 10.1186/s13058-015-0622-z] [Citation(s) in RCA: 150] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 07/23/2015] [Indexed: 02/06/2023] Open
Abstract
Introduction The steady increase in the incidence of obesity among adults has been paralleled with higher levels of obesity-associated breast cancer. While recent studies have suggested that adipose stromal/stem cells (ASCs) isolated from obese women enhance tumorigenicity, the mechanism(s) by which this occurs remains undefined. Evidence suggests that increased adiposity results in increased leptin secretion from adipose tissue, which has been shown to increased cancer cell proliferation. Previously, our group demonstrated that ASCs isolated from obese women (obASCs) also express higher levels of leptin relative to ASCs isolated from lean women (lnASCs) and that this obASC-derived leptin may account for enhanced breast cancer cell growth. The current study investigates the impact of inhibiting leptin expression in lnASCs and obASCs on breast cancer cell (BCC) growth and progression. Methods Estrogen receptor positive (ER+) BCCs were co-cultured with leptin shRNA lnASCs or leptin shRNA obASCs and changes in the proliferation, migration, invasion, and gene expression of BCCs were investigated. To assess the direct impact of leptin inhibition in obASCs on BCC proliferation, MCF7 cells were injected alone or mixed with control shRNA obASCs or leptin shRNA obASCs into SCID/beige mice. Results ER+ BCCs were responsive to obASCs during direct co-culture, whereas lnASCs were unable to increase ER+ BCC growth. shRNA silencing of leptin in obASCs negated the enhanced proliferative effects of obASC on BCCs following direct co-culture. BCCs co-cultured with obASCs demonstrated enhanced expression of epithelial-to-mesenchymal transition (EMT) and metastasis genes (SERPINE1, MMP-2, and IL-6), while BCCs co-cultured with leptin shRNA obASCs did not display similar levels of gene induction. Knockdown of leptin significantly reduced tumor volume and decreased the number of metastatic lesions to the lung and liver. These results correlated with reduced expression of both SERPINE1 and MMP-2 in tumors formed with MCF7 cells mixed with leptin shRNA obASCs, when compared to tumors formed with MCF7 cells mixed with control shRNA obASCs. Conclusion This study provides mechanistic insight as to how obesity enhances the proliferation and metastasis of breast cancer cells; specifically, obASC-derived leptin contributes to the aggressiveness of breast cancer in obese women. Electronic supplementary material The online version of this article (doi:10.1186/s13058-015-0622-z) contains supplementary material, which is available to authorized users.
Collapse
|
31
|
Cohen DM, Won KJ, Nguyen N, Lazar MA, Chen CS, Steger DJ. ATF4 licenses C/EBPβ activity in human mesenchymal stem cells primed for adipogenesis. eLife 2015; 4:e06821. [PMID: 26111340 PMCID: PMC4501333 DOI: 10.7554/elife.06821] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 06/25/2015] [Indexed: 12/24/2022] Open
Abstract
A well-established cascade of transcription factor (TF) activity orchestrates adipogenesis in response to chemical cues, yet how cell-intrinsic determinants of differentiation such as cell shape and/or seeding density inform this transcriptional program remain enigmatic. Here, we uncover a novel mechanism licensing transcription in human mesenchymal stem cells (hMSCs) adipogenically primed by confluence. Prior to adipogenesis, confluency promotes heterodimer recruitment of the bZip TFs C/EBPβ and ATF4 to a non-canonical C/EBP DNA sequence. ATF4 depletion decreases both cell-density-dependent transcription and adipocyte differentiation. Global profiling in hMSCs and a novel cell-free assay reveals that ATF4 requires C/EBPβ for genomic binding at a motif distinct from that bound by the C/EBPβ homodimer. Our observations demonstrate that C/EBPβ bridges the transcriptional programs in naïve, confluent cells and early differentiating pre-adipocytes. Moreover, they suggest that homo- and heterodimer formation poise C/EBPβ to execute diverse and stage-specific transcriptional programs by exploiting an expanded motif repertoire.
Collapse
Affiliation(s)
- Daniel M Cohen
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| | - Kyoung-Jae Won
- The Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| | - Nha Nguyen
- The Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| | - Mitchell A Lazar
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| | - Christopher S Chen
- Department of Biomedical Engineering, Boston University, Boston, United States
| | - David J Steger
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| |
Collapse
|
32
|
Tougaard P, Skov S, Pedersen AE, Krych L, Nielsen DS, Bahl MI, Christensen EG, Licht TR, Poulsen SS, Metzdorff SB, Hansen AK, Hansen CHF. TL1A regulates TCRγδ+intraepithelial lymphocytes and gut microbial composition. Eur J Immunol 2014; 45:865-75. [DOI: 10.1002/eji.201444528] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Revised: 09/22/2014] [Accepted: 11/13/2014] [Indexed: 01/03/2023]
Affiliation(s)
- Peter Tougaard
- Department of Veterinary Disease Biology; Section of Experimental Animal Models; Faculty of Health and Medical Sciences; University of Copenhagen; Copenhagen Denmark
| | - S. Skov
- Department of Veterinary Disease Biology; Section of Experimental Animal Models; Faculty of Health and Medical Sciences; University of Copenhagen; Copenhagen Denmark
| | - A. E. Pedersen
- Department of International Health; Immunology and Microbiology; Faculty of Health and Medical Sciences; University of Copenhagen; Copenhagen Denmark
| | - L. Krych
- Department of Food Science; Faculty of Science; University of Copenhagen; Copenhagen Denmark
| | - D. S. Nielsen
- Department of Food Science; Faculty of Science; University of Copenhagen; Copenhagen Denmark
| | - M. I. Bahl
- National Food Institute; Division of Microbiology and Risk Assessment; Technical University of Denmark; Moerkhoej Denmark
| | - E. G. Christensen
- National Food Institute; Division of Microbiology and Risk Assessment; Technical University of Denmark; Moerkhoej Denmark
| | - T. R. Licht
- National Food Institute; Division of Microbiology and Risk Assessment; Technical University of Denmark; Moerkhoej Denmark
| | - S. S. Poulsen
- Department of Biomedical Sciences; Faculty of Health and Medical Sciences; University of Copenhagen; Copenhagen Denmark
| | - S. B. Metzdorff
- Department of Veterinary Disease Biology; Section of Experimental Animal Models; Faculty of Health and Medical Sciences; University of Copenhagen; Copenhagen Denmark
| | - A. K. Hansen
- Department of Veterinary Disease Biology; Section of Experimental Animal Models; Faculty of Health and Medical Sciences; University of Copenhagen; Copenhagen Denmark
| | - C. H. F. Hansen
- Department of Veterinary Disease Biology; Section of Experimental Animal Models; Faculty of Health and Medical Sciences; University of Copenhagen; Copenhagen Denmark
| |
Collapse
|
33
|
Identification of KMU-3, a novel derivative of gallic acid, as an inhibitor of adipogenesis. PLoS One 2014; 9:e109344. [PMID: 25285517 PMCID: PMC4186852 DOI: 10.1371/journal.pone.0109344] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Accepted: 09/08/2014] [Indexed: 12/23/2022] Open
Abstract
Differentiation of preadipocyte, also called adipogenesis, leads to the phenotype of mature adipocyte. Excessive adipogenesis, however, is largely linked to the development of obesity. Herein we investigated a library of 53 novel chemicals, generated from a number of polyphenolic natural compounds, on adipogenesis. Strikingly, among the chemicals tested, KMU-3, a derivative of gallic acid, strongly suppressed lipid accumulation during the differentiation of 3T3-L1 preadipocytes into adipocytes. On mechanistic levels, KMU-3 inhibited expressions of CCAAT/enhancer-binding protein-α (C/EBP-α), peroxisome proliferator-activated receptor-γ (PPAR-γ), and fatty acid synthase (FAS) during adipocyte differentiation. Moreover, KMU-3 reduced expressions of adipokines, including retinol binding protein-4 (RBP-4), leptin, and regulated on activation, normal T cell expressed and secreted (RANTES) during adipocyte differentiation. Of further note, KMU-3 rapidly blocked the phosphorylation of signal transducer and activator of transcription-3 (STAT-3) during the early stage of adipogenesis. Importantly, pharmacological inhibition studies revealed that AG490, a JAK-2/STAT-3 inhibitor suppressed adipogenesis and STAT-3 phosphorylation, implying that early blockage of STAT-3 activity is crucial for the KMU-3-mediated anti-adipogenesis. These findings demonstrate firstly that KMU-3 inhibits adipogenesis by down-regulating STAT-3, PPAR-γ, C/EBP-α, and FAS. This work shows that KMU-3 is an inhibitor of adipogenesis and thus may have therapeutic potential against obesity.
Collapse
|
34
|
Inhibition of adipogenesis and leptin production in 3T3-L1 adipocytes by a derivative of meridianin C. Biochem Biophys Res Commun 2014; 452:1078-83. [DOI: 10.1016/j.bbrc.2014.09.050] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 09/12/2014] [Indexed: 01/24/2023]
|
35
|
|
36
|
Abstract
Since individual cells from freshly isolated white adipose tissue (WAT) exhibit variable levels of fat accumulation, we attempted to determine which factor(s) cause this variation. We used primary WAT cells from adult mice and the mouse 3T3-L1 cell-line of preadipocytes for these studies. Cells were labeled with BODIPY (boron-dipyrromethene) lipid probe, a marker for fat accumulation in live cells, and sorted on a fluorescence-activated cell sorter into two populations exhibiting low or high BODIPY fluorescence intensity. After more than 12 doublings as dedifferentiated cells in growth medium, the sorted populations were exposed to adipogenic medium for 7 days and analyzed for BODIPY accumulation and mRNA expression of adipogenic markers. WAT-derived cells initially sorted to have low or high BODIPY fluorescence intensity maintained a similar low or high lipid phenotype after redifferentiation. Cell surface TSH receptor expression, which is known to increase when preadipocytes are differentiated, correlated with BODIPY staining in all states. mRNA levels of Pparγ, Srebp1c, aP2, and Pref1, key regulators of adipogenesis, and leptin, Glut4, Fasn, and Tshr, markers of adipocyte differentiation, correlated with the levels of fat accumulation. Overexpression of Pparγ in 3T3-L1 cells, as expected, caused cells from low- and high-BODIPY populations to accumulate more fat. More importantly, prior to differentiation, the endogenous Pparγ promoter exhibited higher levels of acetylated histone H3, an activatory modification, in high-BODIPY- compared with low-BODIPY-derived populations. We conclude that fat accumulation is a heritable trait in WAT and that epigenetic modification on the Pparγ promoter contributes to this heritability.
Collapse
Affiliation(s)
- Liora S Katz
- Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Elizabeth Geras-Raaka
- Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Marvin C Gershengorn
- Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
37
|
Krontiras P, Gatenholm P, Hägg DA. Adipogenic differentiation of stem cells in three-dimensional porous bacterial nanocellulose scaffolds. J Biomed Mater Res B Appl Biomater 2014; 103:195-203. [DOI: 10.1002/jbm.b.33198] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 03/26/2014] [Accepted: 04/24/2014] [Indexed: 11/08/2022]
Affiliation(s)
- Panagiotis Krontiras
- Department of Chemical and Biological Engineering; Chalmers University of Technology; Gothenburg SE-412 96 Sweden
| | - Paul Gatenholm
- Department of Chemical and Biological Engineering; Chalmers University of Technology; Gothenburg SE-412 96 Sweden
| | - Daniel A Hägg
- Department of Chemical and Biological Engineering; Chalmers University of Technology; Gothenburg SE-412 96 Sweden
| |
Collapse
|
38
|
Lim JM, Wollaston-Hayden EE, Teo CF, Hausman D, Wells L. Quantitative secretome and glycome of primary human adipocytes during insulin resistance. Clin Proteomics 2014; 11:20. [PMID: 24948903 PMCID: PMC4055909 DOI: 10.1186/1559-0275-11-20] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 02/04/2014] [Indexed: 01/04/2023] Open
Abstract
Adipose tissue is both an energy storage depot and an endocrine organ. The impaired regulation of the secreted proteins of adipose tissue, known as adipocytokines, observed during obesity contributes to the onset of whole-body insulin resistance and the pathobiology of type 2 diabetes mellitus (T2DM). In addition, the global elevation of the intracellular glycosylation of proteins by O-linked β-N-acetylglucosamine (O-GlcNAc) via either genetic or pharmacological methods is sufficient to induce insulin resistance in both cultured cells and animal models. The elevation of global O-GlcNAc levels is associated with the altered expression of many adipocytokines. We have previously characterized the rodent adipocyte secretome during insulin sensitive and insulin resistant conditions. Here, we characterize and quantify the secretome and glycome of primary human adipocytes during insulin responsive and insulin resistant conditions generated by the classical method of hyperglycemia and hyperinsulinemia or by the pharmacological manipulation of O-GlcNAc levels. Using a proteomic approach, we identify 190 secreted proteins and report a total of 20 up-regulated and 6 down-regulated proteins that are detected in both insulin resistant conditions. Moreover, we apply glycomic techniques to examine (1) the sites of N-glycosylation on secreted proteins, (2) the structures of complex N- and O-glycans, and (3) the relative abundance of complex N- and O-glycans structures in insulin responsive and insulin resistant conditions. We identify 91 N-glycosylation sites derived from 51 secreted proteins, as well as 155 and 29 released N- and O-glycans respectively. We go on to quantify many of the N- and O-glycan structures between insulin responsive and insulin resistance conditions demonstrating no significant changes in complex glycosylation in the time frame for the induction of insulin resistance. Thus, our data support that the O-GlcNAc modification is involved in the regulation of adipocytokine secretion upon the induction of insulin resistance in human adipocytes.
Collapse
Affiliation(s)
- Jae-Min Lim
- Complex Carbohydrate Research Center, The University of Georgia, 315 Riverbend Road, 30602-4712 Athens, Georgia ; Department of Chemistry, The University of Georgia, 30602 Athens, Georgia ; Department of Chemistry, Changwon National University, Changwon, Gyeongnam 641-773, South Korea
| | - Edith E Wollaston-Hayden
- Complex Carbohydrate Research Center, The University of Georgia, 315 Riverbend Road, 30602-4712 Athens, Georgia ; Department of Biochemistry and Molecular Biology, The University of Georgia, 30602 Athens, Georgia
| | - Chin Fen Teo
- Complex Carbohydrate Research Center, The University of Georgia, 315 Riverbend Road, 30602-4712 Athens, Georgia ; Department of Biochemistry and Molecular Biology, The University of Georgia, 30602 Athens, Georgia
| | - Dorothy Hausman
- Department of Foods and Nutrition, The University of Georgia, 30602 Athens, Georgia
| | - Lance Wells
- Complex Carbohydrate Research Center, The University of Georgia, 315 Riverbend Road, 30602-4712 Athens, Georgia ; Department of Chemistry, The University of Georgia, 30602 Athens, Georgia ; Department of Biochemistry and Molecular Biology, The University of Georgia, 30602 Athens, Georgia
| |
Collapse
|
39
|
Ilavenil S, Arasu MV, Lee JC, Kim DH, Roh SG, Park HS, Choi GJ, Mayakrishnan V, Choi KC. Trigonelline attenuates the adipocyte differentiation and lipid accumulation in 3T3-L1 cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2014; 21:758-765. [PMID: 24369814 DOI: 10.1016/j.phymed.2013.11.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Revised: 10/10/2013] [Accepted: 11/29/2013] [Indexed: 06/03/2023]
Abstract
Trigonelline is a natural alkaloid mainly found in Trigonella Foenum Graecum (fenugreek) Fabaceae and other edible plants with a variety of medicinal applications. Therefore, we investigated the molecular mechanism of trigonelline (TG) on the inhibition of adipocyte differentiation and lipid accumulation in 3T3-L1 cells. Trigonelline suppressed lipid droplet accumulation in a concentration (75 and 100 μM) dependent manner. Treatment of adipocyte with of TG down regulates the peroxisome proliferator-activated receptor (PPARγ) and CCAAT element binding protein (C/EBP-α) mRNA expression, which leads to further down regulation of other gene such as adiponectin, adipogenin, leptin, resistin and adipocyte fatty acid binding protein (aP2) as compared with respective control cells on 5th and 10th day of differentiation. Further, addition of triognelline along with troglitazone to the adipocyte attenuated the troglitazone effects on PPARγ mediated differentiation and lipid accumulation in 3T3-L1 cells. Trigonelline might compete against troglitazone for its binding to the PPARγ. In addition, adipocyte treated with trigonelline and isoproterenol separately. Isoproterenol, a lipolytic agent which inhibits the fatty acid synthase and GLUT-4 transporter expression via cAMP mediated pathway, we found that similar magnitude response of fatty acid synthase and GLUT-4 transporter expression in trigonelline treated adipocyte. These results suggest that the trigonelline inhibits the adipogenesis by its influences on the expression PPARγ, which leads to subsequent down regulation of PPAR-γ mediated pathway during adipogenesis. Our findings provide key approach to the mechanism underlying the anti-adipogenic activity of trigonelline.
Collapse
Affiliation(s)
- Soundharrajan Ilavenil
- Grassland and Forage Division, National Institute of Animal Science, RDA, Seonghwan-Eup, Cheonan-Si, Chungnam 330-801, Republic of Korea
| | - Mariadhas Valan Arasu
- Grassland and Forage Division, National Institute of Animal Science, RDA, Seonghwan-Eup, Cheonan-Si, Chungnam 330-801, Republic of Korea
| | - Jeong-Chae Lee
- Research Center of Bioactive Materials, Chonbuk National University, Jeonju 561-756, Republic of Korea
| | - Da Hye Kim
- Faculty of Life and Environmental Science, Shimane University, Matsue, Japan; The United Graduate School of Agricultural Sciences, Tottori University, Tottori-Shi 680-8553, Japan
| | - Sang Gun Roh
- Laboratory of Animal Physiology, Graduate School of Agricultural Science, Tohoku University, Aoba, Sendai, Japan
| | - Hyung Su Park
- Grassland and Forage Division, National Institute of Animal Science, RDA, Seonghwan-Eup, Cheonan-Si, Chungnam 330-801, Republic of Korea
| | - Gi Jun Choi
- Grassland and Forage Division, National Institute of Animal Science, RDA, Seonghwan-Eup, Cheonan-Si, Chungnam 330-801, Republic of Korea
| | - Vijayakumar Mayakrishnan
- Mushroom Research Centre, Institute of Biological Science, Faculty of Science, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Ki Choon Choi
- Grassland and Forage Division, National Institute of Animal Science, RDA, Seonghwan-Eup, Cheonan-Si, Chungnam 330-801, Republic of Korea.
| |
Collapse
|
40
|
Dib LH, Ortega MT, Fleming SD, Chapes SK, Melgarejo T. Bone marrow leptin signaling mediates obesity-associated adipose tissue inflammation in male mice. Endocrinology 2014; 155:40-6. [PMID: 24169547 PMCID: PMC3868799 DOI: 10.1210/en.2013-1607] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Obesity is characterized by an increased recruitment of proinflammatory macrophages to the adipose tissue (AT), leading to systemic inflammation and metabolic disease. The pathogenesis of this AT inflammation, however, remains to be elucidated. The circulating adipokine leptin is increased in obesity and is involved in immune cell function and activation. In the present study, we investigated the role of leptin in the induction of obesity-associated inflammation. We generated radiation chimeric C57BL/6J mice reconstituted with either leptin receptor-deficient (db/db) or wild-type (WT) bone marrow and challenged them with a high-fat diet (HFD) for 16 weeks. Mice reconstituted with db/db bone marrow (WT/db), had significantly lower body weight and adiposity compared with mice with WT bone marrow (WT/WT). Gonadal AT in WT/db mice displayed a 2-fold lower expression of the inflammatory genes Tnfa, Il6, and Ccl2. In addition, gonadal fat of WT/db mice contained significantly fewer crown-like structures compared with WT/WT mice, and most of their AT macrophages expressed macrophage galactose-type C type lectin 1 (MGL1) and were C-C chemokine receptor type 2 (CCR2)-negative, indicative of an anti-inflammatory phenotype. Moreover, WT/db mice exhibited greater insulin sensitivity compared with WT/WT mice. These data show that disrupted leptin signaling in bone marrow-derived cells attenuates the proinflammatory conditions that mediate many of the metabolic complications that characterize obesity. Our findings establish a novel mechanism involved in the regulation of obesity-associated systemic inflammation and support the hypothesis that leptin is a proinflammatory cytokine.
Collapse
|
41
|
Wollaston-Hayden EE, Harris RBS, Liu B, Bridger R, Xu Y, Wells L. Global O-GlcNAc Levels Modulate Transcription of the Adipocyte Secretome during Chronic Insulin Resistance. Front Endocrinol (Lausanne) 2014; 5:223. [PMID: 25657638 PMCID: PMC4302944 DOI: 10.3389/fendo.2014.00223] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Accepted: 12/05/2014] [Indexed: 01/06/2023] Open
Abstract
Increased flux through the hexosamine biosynthetic pathway and the corresponding increase in intracellular glycosylation of proteins via O-linked β-N-acetylglucosamine (O-GlcNAc) is sufficient to induce insulin resistance (IR) in multiple systems. Previously, our group used shotgun proteomics to identify multiple rodent adipocytokines and secreted proteins whose levels are modulated upon the induction of IR by indirectly and directly modulating O-GlcNAc levels. We have validated the relative levels of several of these factors using immunoblotting. Since adipocytokines levels are regulated primarily at the level of transcription and O-GlcNAc alters the function of many transcription factors, we hypothesized that elevated O-GlcNAc levels on key transcription factors are modulating secreted protein expression. Here, we show that upon the elevation of O-GlcNAc levels and the induction of IR in mature 3T3-F442a adipocytes, the transcript levels of multiple secreted proteins reflect the modulation observed at the protein level. We validate the transcript levels in male mouse models of diabetes. Using inguinal fat pads from the severely IR db/db mouse model and the mildly IR diet-induced mouse model, we have confirmed that the secreted proteins regulated by O-GlcNAc modulation in cell culture are likewise modulated in the whole animal upon a shift to IR. By comparing the promoters of similarly regulated genes, we determine that Sp1 is a common cis-acting element. Furthermore, we show that the LPL and SPARC promoters are enriched for Sp1 and O-GlcNAc modified proteins during insulin resistance in adipocytes. Thus, the O-GlcNAc modification of proteins bound to promoters, including Sp1, is linked to adipocytokine transcription during insulin resistance.
Collapse
Affiliation(s)
- Edith E. Wollaston-Hayden
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA, USA
| | - Ruth B. S. Harris
- Department of Physiology, Georgia Health Sciences University, Augusta, GA, USA
| | - Bingqiang Liu
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA, USA
| | - Robert Bridger
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
| | - Ying Xu
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA, USA
| | - Lance Wells
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA, USA
- *Correspondence: Lance Wells, Department of Biochemistry and Molecular Biology, Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Road, Athens, GA 30602, USA e-mail:
| |
Collapse
|
42
|
p300-dependent acetylation of activating transcription factor 5 enhances C/EBPβ transactivation of C/EBPα during 3T3-L1 differentiation. Mol Cell Biol 2013; 34:315-24. [PMID: 24216764 DOI: 10.1128/mcb.00956-13] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Adipogenesis is a multistep process by which 3T3-L1 preadipocytes differentiate into mature adipocytes through mitotic clonal expansion (MCE) and terminal differentiation. The CCAAT/enhancer-binding protein β (C/EBPβ) is an important transcription factor that takes part in both of these processes. C/EBPβ not only transactivates C/EBPα and the peroxisome proliferator-activated receptor γ (PPARγ), which cause 3T3-L1 preadipocytes to enter terminal adipocyte differentiation, but also is required to activate cell cycle genes necessary for MCE. The identification of potential cofactors of C/EBPβ will help to explain how C/EBPβ undertakes these specialized roles during the different stages of adipogenesis. In this study, we found that activating transcription factor 5 (ATF5) can bind to the promoter of C/EBPα via its direct interaction with C/EBPβ (which is mediated via the p300-dependent acetylation of ATF5), leading to enhanced C/EBPβ transactivation of C/EBPα. We also show that p300 is important for the interaction of ATF5 with C/EBPβ as well as for the binding activity of this complex on the C/EBPα promoter. Consistent with these findings, overexpression of ATF5 and an acetylated ATF5 mimic both promoted 3T3-L1 adipocyte differentiation, whereas short interfering RNA-mediated ATF5 downregulation inhibited this process. Furthermore, we show that the elevated expression of ATF5 is correlated with an obese phenotype in both mice and humans. In summary, we have identified ATF5 as a new cofactor of C/EBPβ and examined how C/EBPβ and ATF5 (acetylated by a p300-dependent mechanism) regulate the transcription of C/EBPα.
Collapse
|
43
|
Wittmann K, Storck K, Muhr C, Mayer H, Regn S, Staudenmaier R, Wiese H, Maier G, Bauer-Kreisel P, Blunk T. Development of volume-stable adipose tissue constructs using polycaprolactone-based polyurethane scaffolds and fibrin hydrogels. J Tissue Eng Regen Med 2013; 10:E409-E418. [PMID: 24170732 DOI: 10.1002/term.1830] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Revised: 07/25/2013] [Accepted: 08/30/2013] [Indexed: 01/07/2023]
Abstract
Adipose tissue engineering aims at the restoration of soft tissue defects and the correction of contour deformities. It is therefore crucial to provide functional adipose tissue implants with appropriate volume stability. Here, we investigate two different fibrin formulations, alone or in combination with biodegradable polyurethane (PU) scaffolds as additional support structures, with regard to their suitability to generate volume-stable adipose tissue constructs. Human adipose-derived stem cells (ASCs) were incorporated in a commercially available fibrin sealant as well as a stable fibrin hydrogel previously developed by our group. The composite constructs made from the commercially available fibrin and porous poly(ε-caprolactone)-based polyurethane scaffolds exhibited increased volume stability as compared to fibrin gels alone; however, only constructs using the stable fibrin gels completely maintained their size and weight for 21 days. Adipogenesis of ASCs was not impaired by the additional PU scaffold. After induction with a common hormonal cocktail, for constructs with either fibrin formulation, strong adipogenic differentiation of ASCs was observed after 21 days in vitro. Furthermore, upregulation of adipogenic marker genes was demonstrated at mRNA (PPARγ, C/EBPα, GLUT4 and aP2; qRT-PCR) and protein (leptin; ELISA) levels. Stable fibrin/PU constructs were further evaluated in a pilot in vivo study, resulting in areas of well-vascularized adipose tissue within the implants after only 5 weeks. Copyright © 2013 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Katharina Wittmann
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, University of Würzburg, Germany
| | - Katharina Storck
- Department of ENT, Head and Neck Surgery, Technical University of Munich, Germany
| | - Christian Muhr
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, University of Würzburg, Germany
| | - Helena Mayer
- Department of ENT, Head and Neck Surgery, Technical University of Munich, Germany
| | - Sybille Regn
- Department of ENT, Head and Neck Surgery, Technical University of Munich, Germany
| | - Rainer Staudenmaier
- Department of ENT, Head and Neck Surgery, Technical University of Munich, Germany
| | | | | | - Petra Bauer-Kreisel
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, University of Würzburg, Germany
| | - Torsten Blunk
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, University of Würzburg, Germany.
| |
Collapse
|
44
|
Cignarelli A, Giorgino F, Vettor R. Pharmacologic agents for type 2 diabetes therapy and regulation of adipogenesis. Arch Physiol Biochem 2013; 119:139-50. [PMID: 23724947 DOI: 10.3109/13813455.2013.796996] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The close link between type 2 diabetes and excess body weight highlights the need to consider the effects on weight of different treatments used for correction of hyperglycaemia. Indeed, specific currently available diabetes therapies can cause weight gain, including insulin and its analogues, sulphonylureas, and thiazolidinediones, while others, such as metformin and the GLP-1 receptor agonists, can promote weight loss. Excess body weight in patients with diabetes is largely due to expansion of adipose tissue, and these drugs could interfere with the mechanisms underlying the expansion and differentiation of adipocyte precursors. Almost all anti-diabetes drugs could also potentially affect adipocyte metabolism directly, by modulating lipogenesis, lipolysis, and fat oxidation. This review will examine the available evidence for specific effects of various anti-diabetes drugs on adipose tissue development and function with the ultimate goal of increasing our understanding of how pharmacological agents can modulate energy balance and body fat.
Collapse
Affiliation(s)
- A Cignarelli
- Department of Emergency and Organ Transplantation, Section of Internal Medicine, Endocrinology, Andrology, and Metabolic Diseases, University of Bari "Aldo Moro" , Bari , Italy and
| | | | | |
Collapse
|
45
|
Neo PY, See EYS, Toh SL, Goh JCH. Temporal profiling of the growth and multi-lineage potentiality of adipose tissue-derived mesenchymal stem cells cell-sheets. J Tissue Eng Regen Med 2013; 10:564-79. [PMID: 23784965 DOI: 10.1002/term.1776] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Revised: 04/11/2013] [Accepted: 04/16/2013] [Indexed: 12/24/2022]
Abstract
Cell-sheet tissue engineering retains the benefits of an intact extracellular matrix (ECM) and can be used to produce scaffold-free constructs. Adipose tissue-derived stem cells (ASCs) are multipotent and more easily obtainable than the commonly used bone marrow-derived stem cells (BMSCs). Although BMSC cell sheets have been previously reported to display multipotentiality, a detailed study of the development and multilineage potential of ASC cell sheets (ASC-CSs) is non-existent in the literature. The aims of this study were to temporally profile: (a) the effect of hyperconfluent culture duration on ASC-CSs development; and (b) the multipotentiality of ASC-CSs by differentiation into the osteogenic, adipogenic and chondrogenic lineages. Rabbit ASCs were first isolated and cultured until confluence (day 0). The confluent cells were then cultured in ascorbic acid-supplemented medium for 3 weeks to study cell metabolic activity, cell sheet thickness and early differentiation gene expressions at weekly time points. ASC-CSs and ASCs were then differentiated into the three lineages, using established protocols, and assessed by RT-PCR and histology at multiple time points. ASC-CSs remained healthy up to 3 weeks of hyperconfluent culture. One week-old cell sheets displayed upregulation of early differentiation gene markers (Runx2 and Sox9); however, subsequent differentiation results indicated that they did not necessarily translate to an improved phenotype. ASCs within the preformed cell sheet groups did not differentiate as efficiently as the non-hyperconfluent ASCs, which were directly differentiated. Although ASCs within the cell sheets retained their differentiation capacity and remained viable under prolonged hyperconfluent conditions, future applications of ASC-CSs in tissue engineering should be considered with care. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Puay Yong Neo
- Department of Bioengineering, Faculty of Engineering, National University of Singapore
| | - Eugene Yong-Shun See
- Department of Bioengineering, Faculty of Engineering, National University of Singapore
| | - Siew Lok Toh
- Department of Bioengineering, Faculty of Engineering, National University of Singapore.,Department of Mechanical Engineering, Faculty of Engineering, National University of Singapore
| | - James Cho-Hong Goh
- Department of Bioengineering, Faculty of Engineering, National University of Singapore.,Department of Orthopaedic Surgery, Yong Lin Loo School of Medicine, National University of Singapore
| |
Collapse
|
46
|
Bae KH, Kim WK, Lee SC. Involvement of protein tyrosine phosphatases in adipogenesis: new anti-obesity targets? BMB Rep 2013; 45:700-6. [PMID: 23261055 PMCID: PMC4133817 DOI: 10.5483/bmbrep.2012.45.12.235] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Obesity is a worldwide epidemic as well as being a major risk factor for diabetes, cardiovascular diseases and several types of cancers. Obesity is mainly due to the overgrowth of adipose tissue arising from an imbalance between energy intake and energy expenditure. Adipose tissue, primarily composed of adipocytes, plays a key role in maintaining whole body energy homeostasis. In view of the treatment of obesity and obesity-related diseases, it is critical to understand the detailed signal transduction mechanisms of adipogenic differentiation. Adipogenic differentiation is tightly regulated by many key signal cascades, including insulin signaling. These signal cascades generally transfer or amplify the signal by using serial tyrosine phosphorylations. Thus, protein tyrosine kinases and protein tyrosine phosphatases are closely related to adipogenic differentiation. Compared to protein tyrosine kinases, protein tyrosine phosphatases have received little attention in adipogenic differentiation. This review aims to highlight the involvement of protein tyrosine phosphatases in adipogenic differentiation and the possibility of protein tyrosine phosphatases as drugs to target obesity. [BMB Reports 2012; 45(12): 700-706]
Collapse
Affiliation(s)
- Kwang-Hee Bae
- Medical Proteomics Research Center, KRIBB, Daejeon 305-806, Korea.
| | | | | |
Collapse
|
47
|
Bahar B, O’Doherty JV, O’Doherty AM, Sweeney T. Chito-oligosaccharide inhibits the de-methylation of a 'CpG' island within the leptin (LEP) promoter during adipogenesis of 3T3-L1 cells. PLoS One 2013; 8:e60011. [PMID: 23544120 PMCID: PMC3609775 DOI: 10.1371/journal.pone.0060011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Accepted: 02/20/2013] [Indexed: 11/29/2022] Open
Abstract
Chito-oligosaccharide (COS) is a natural bioactive compound, which has been shown to suppress lipid metabolic genes and lipid accumulation in differentiating adipocytes. Leptin has been identified as a key regulator of energy homeostasis and is known to be under epigenetic regulation during adipogenesis. Hence, the first objective of this experiment was to compare leptin gene (LEP) expression and leptin secretion during the different stages of adipogenesis and to investigate the effect of COS on these processes. As COS inhibited LEP expression during adipogenesis, the second aim was to investigate the methylation dynamics of a ‘CpG’ island in the proximal region of the LEP promoter during adipogenesis and to determine the effect of COS on this process. Mouse 3T3-L1 cells were stimulated to differentiate in the absence or presence of COS and the levels of leptin mRNA and protein were evaluated on days 0, 2, 4 and 6 post-induction of differentiation (PID). The extent of de-methylation of six CpG sites was evaluated. LEP mRNA transcript and protein could not be detected on either day 0PID or 2PID. In contrast, both were detected on day 4PID (P<0.05) and 6PID (P<0.001) and both were inhibited by COS (P<0.001). Of the six CpG sites analyzed, CpG_52, CpG_62 and CpG_95 became 11.5, 5.0 and 5.0% de-methylated between day 2PID and 6PID, respectively. COS blocked this de-methylation event at CpG_52 (P<0.001), CpG_62 (P<0.01) and CpG_95 (P<0.01) on day 6PID. These data suggest that COS can have an epigenetic effect on differentiating adipocytes, a novel biological function of COS which has potential applications for the manipulation of leptin gene expression, adipogenesis, and conditions within the metabolic syndrome spectrum.
Collapse
Affiliation(s)
- Bojlul Bahar
- School of Agriculture and Food Science, University College Dublin, Dublin, Ireland
- School of Veterinary Medicine, University College Dublin, Dublin, Ireland
| | - John V. O’Doherty
- School of Agriculture and Food Science, University College Dublin, Dublin, Ireland
| | - Alan M. O’Doherty
- School of Agriculture and Food Science, University College Dublin, Dublin, Ireland
| | - Torres Sweeney
- School of Veterinary Medicine, University College Dublin, Dublin, Ireland
- * E-mail:
| |
Collapse
|
48
|
Bellas E, Marra KG, Kaplan DL. Sustainable three-dimensional tissue model of human adipose tissue. Tissue Eng Part C Methods 2013; 19:745-54. [PMID: 23373822 DOI: 10.1089/ten.tec.2012.0620] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The need for physiologically relevant sustainable human adipose tissue models is crucial for understanding tissue development, disease progression, in vitro drug development and soft tissue regeneration. The coculture of adipocytes differentiated from human adipose-derived stem cells, with endothelial cells, on porous silk protein matrices for at least 6 months is reported, while maintaining adipose-like outcomes. Cultures were assessed for structure and morphology (Oil Red O content and CD31 expression), metabolic functions (leptin, glycerol production, gene expression for GLUT4, and PPARγ) and cell replication (DNA content). The cocultures maintained size and shape over this extended period in static cultures, while increasing in diameter by 12.5% in spinner flask culture. Spinner flask cultures yielded improved adipose tissue outcomes overall, based on structure and function, when compared to the static cultures. This work establishes a tissue model system that can be applied to the development of chronic metabolic dysfunction systems associated with human adipose tissue, such as obesity and diabetes, due to the long term sustainable functions demonstrated here.
Collapse
Affiliation(s)
- Evangelia Bellas
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, USA
| | | | | |
Collapse
|
49
|
Kleiman A, Keats EC, Chan NG, Khan ZA. Elevated IGF2 prevents leptin induction and terminal adipocyte differentiation in hemangioma stem cells. Exp Mol Pathol 2012; 94:126-36. [PMID: 23047069 DOI: 10.1016/j.yexmp.2012.09.023] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Revised: 09/11/2012] [Accepted: 09/28/2012] [Indexed: 10/27/2022]
Abstract
Infantile hemangioma is a benign vascular tumor that exhibits a unique yet predictable lifecycle of rapid proliferation followed by spontaneous regression. Recent studies have identified that insulin-like growth factor-2 (IGF2), a fetal mitogen, is highly expressed during the proliferative phase of hemangioma growth. Since hemangiomas arise from CD133+ stem cells, high levels of IGF2 may regulate the activity of the stem cells and therefore, hemangioma growth. The aim of this study was to understand the functional significance of elevated IGF2 in hemangiomas. We show that IGF2 localizes to the CD133+ cells in hemangioma specimens. We, therefore, hypothesized that IGF2 may be regulating the plasticity of hemangioma stem cells. To test our hypothesis, we used CD133-selected cells from hemangiomas to knockdown the expression of IGF2. We found that IGF2 is a mitogen for hemangioma stem cells and prevents leptin induction and full terminal differentiation of hemangioma stem cells into adipocytes. We also show that IGF2 does not alter the initial commitment phase. These findings implicate an important role of IGF2 in expanding hemangioma stem cells and preventing terminal adipocyte differentiation.
Collapse
Affiliation(s)
- Alexandra Kleiman
- Department of Pathology, University of Western Ontario, London ON, Canada
| | | | | | | |
Collapse
|
50
|
Liang G, Gao W, Liang A, Ye W, Peng Y, Zhang L, Sharma S, Su P, Huang D. Normal leptin expression, lower adipogenic ability, decreased leptin receptor and hyposensitivity to Leptin in Adolescent Idiopathic Scoliosis. PLoS One 2012; 7:e36648. [PMID: 22615788 PMCID: PMC3352937 DOI: 10.1371/journal.pone.0036648] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Accepted: 04/11/2012] [Indexed: 01/16/2023] Open
Abstract
Leptin has been suggested to play a role in the etiology of Adolescent Idiopathic Scoliosis (AIS), however, the leptin levels in AIS girls are still a discrepancy, and no in vitro study of leptin in AIS is reported. We took a series of case-control studies, trying to understand whether Leptin gene polymorphisms are involved in the etiology of the AIS or the change in leptin level is a secondary event, to assess the level of leptin receptor, and to evaluate the differences of response to leptin between AIS cases and controls. We screened all exons of Leptin gene in 45 cases and 45 controls and selected six tag SNPs to cover all the observed variations. Association analysis in 446 AIS patients and 550 healthy controls showed no association between the polymorphisms of Leptin gene and susceptibility/severity to AIS. Moreover, adipogenesis assay of bone mesenchymal stem cells (MSCs) suggested that the adipogenic ability of MSCs from AIS girls was lower than controls. After adjusting the differentiation rate, expressions of leptin and leptin receptor were similar between two groups. Meanwhile, osteogenesis assay of MSC showed the leptin level was similar after adjusting the differentiation rate, but the leptin receptor level was decreased in induced AIS osteoblasts. Immunocytochemistry and western blot analysis showed less leptin receptors expressed in AIS group. Furthermore, factorial designed studies with adipogenesis and osteogenesis revealed that the MSCs from patients have no response to leptin treatment. Our results suggested that Leptin gene variations are not associated with AIS and low serum leptin probably is a secondary outcome which may be related to the low capability of adipogenesis in AIS. The decreased leptin receptor levels may lead to the hyposensitivity to leptin. These findings implied that abnormal peripheral leptin signaling plays an important role in the pathological mechanism of AIS.
Collapse
Affiliation(s)
- Guoyan Liang
- Department of Orthopedics, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Wenjie Gao
- Department of Orthopedics, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Anjing Liang
- Department of Orthopedics, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Wei Ye
- Department of Orthopedics, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yan Peng
- Department of Orthopedics, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Liangming Zhang
- Department of Orthopedics, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Swarkar Sharma
- Seay Center for Musculoskeletal Research, Texas Scottish Rite Hospital for Children, Dallas, Texas, United States of America
| | - Peiqiang Su
- Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
- * E-mail: (DH); (PS)
| | - Dongsheng Huang
- Department of Orthopedics, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
- * E-mail: (DH); (PS)
| |
Collapse
|