1
|
Uramaru N, Kawashima A, Osabe M, Higuchi T. Rhododendrol, a reductive metabolite of raspberry ketone, suppresses the differentiation of 3T3‑L1 cells into adipocytes. Mol Med Rep 2023; 27:51. [PMID: 36633126 PMCID: PMC9879071 DOI: 10.3892/mmr.2023.12938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 12/21/2022] [Indexed: 01/13/2023] Open
Abstract
Obesity is a serious medical condition worldwide, and a major risk factor for type 2 diabetes, metabolic syndrome, cancer and cardiovascular disease. In addition to changes in dietary habits and physical activity, consuming supplements to maintain good health and prevent obesity is important in modern society. Raspberry ketone (RK) is a natural phenolic ketone found in the European red raspberry (Rubus idaeus L.) and is hypothesized to prevent obesity when administered orally. The present study found that RK was reduced to rhododendrol (ROH) in human liver microsomes and cytosol. The present study investigated whether the metabolite ROH had anti‑adipogenic effects using mouse 3T3‑L1 cells. The effects of ROH or RK on lipid accumulation during differentiation of 3T3‑L1 pre‑adipocyte into adipocyte were determined using Oil Red O staining. CCAAT enhancer‑binding protein α (C/EBPα) and peroxisome proliferator‑activated receptor γ (PPARγ) mRNA and protein expression were examined using reverse transcription‑quantitative PCR and western blotting analysis, respectively. The present study revealed that ROH suppressed lipid accumulation in the cells, similar to RK. In addition, ROH suppressed the mRNA expression levels of C/EBPα and PPARγ in 3T3‑L1 adipocytes. Furthermore, ROH suppressed PPARγ protein expression in 3T3‑L1 adipocytes. These findings suggested that ROH is an active metabolite with an anti‑adipogenic effect, which may contribute to the anti‑obesity effect of orally administered RK. The present study indicated that it is important to understand the biological activity of the metabolites of orally administered compounds.
Collapse
Affiliation(s)
- Naoto Uramaru
- Division of Pharmaceutical Health Biosciences, Nihon Pharmaceutical University, Saitama 362-0806, Japan
| | - Azusa Kawashima
- Division of Pharmaceutical Health Biosciences, Nihon Pharmaceutical University, Saitama 362-0806, Japan
| | - Makoto Osabe
- Division of Pharmaceutical Health Biosciences, Nihon Pharmaceutical University, Saitama 362-0806, Japan
| | - Toshiyuki Higuchi
- Division of Pharmaceutical Health Biosciences, Nihon Pharmaceutical University, Saitama 362-0806, Japan,Correspondence to: Professor Toshiyuki Higuchi, Division of Pharmaceutical Health Biosciences, Nihon Pharmaceutical University, 10281 Komuro, Ina-machi, Kitaadachi-gun, Saitama 362-0806, Japan, E-mail:
| |
Collapse
|
2
|
Senoo H, Murata D, Wai M, Arai K, Iwata W, Sesaki H, Iijima M. KARATE: PKA-induced KRAS4B-RHOA-mTORC2 supercomplex phosphorylates AKT in insulin signaling and glucose homeostasis. Mol Cell 2021; 81:4622-4634.e8. [PMID: 34551282 DOI: 10.1016/j.molcel.2021.09.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 06/06/2021] [Accepted: 08/30/2021] [Indexed: 01/22/2023]
Abstract
AKT is a serine/threonine kinase that plays an important role in metabolism, cell growth, and cytoskeletal dynamics. AKT is activated by two kinases, PDK1 and mTORC2. Although the regulation of PDK1 is well understood, the mechanism that controls mTORC2 is unknown. Here, by investigating insulin receptor signaling in human cells and biochemical reconstitution, we found that insulin induces the activation of mTORC2 toward AKT by assembling a supercomplex with KRAS4B and RHOA GTPases, termed KARATE (KRAS4B-RHOA-mTORC2 Ensemble). Insulin-induced KARATE assembly is controlled via phosphorylation of GTP-bound KRAS4B at S181 and GDP-bound RHOA at S188 by protein kinase A. By developing a KARATE inhibitor, we demonstrate that KRAS4B-RHOA interaction drives KARATE formation. In adipocytes, KARATE controls insulin-dependent translocation of the glucose transporter GLUT4 to the plasma membrane for glucose uptake. Thus, our work reveals a fundamental mechanism that activates mTORC2 toward AKT in insulin-regulated glucose homeostasis.
Collapse
Affiliation(s)
- Hiroshi Senoo
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Daisuke Murata
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - May Wai
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Kenta Arai
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Wakiko Iwata
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Hiromi Sesaki
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Miho Iijima
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
3
|
Zhang Q, Wu S, Xiao Q, Kang C, Hu H, Hou X, Wei X, Hao W. Effects of 4-nonylphenol on adipogenesis in 3T3-L1 preadipocytes and C3H/10T1/2 mesenchymal stem cells. J Appl Toxicol 2021; 42:588-599. [PMID: 34553387 DOI: 10.1002/jat.4241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 09/01/2021] [Accepted: 09/04/2021] [Indexed: 11/06/2022]
Abstract
Obesogens are a subset of endocrine disruptor chemicals (EDCs) that cause obesity. The typical EDC 4-nonylphenol (4-NP) has been identified as an obesogen. However, the in vitro effects of 4-NP on adipogenesis remain unclear. In this study, 3T3-L1 preadipocytes and C3H/10T1/2 mesenchymal stem cells (MSCs) were used to investigate the influence of 4-NP on adipogenesis. The differentiation protocols for 3T3-L1 preadipocytes and C3H/10T1/2 MSCs took 8 and 12 days, respectively, beginning at Day 0. In differentiated 3T3-L1 preadipocytes, 20 μM 4-NP decreased cell viability on Days 4 and 8. Exposure to 4-NP inhibited triglyceride (TG) accumulation and adipogenic marker expression on Days 0-8, but the inhibitory effects were weaker on Days 2-8. The protein expression of pSTAT3 or STAT3 decreased on Days 0-8 and 2-8. Conversely, 4-NP promoted TG accumulation and the adipogenic marker expression in C3H/10T1/2 adipocytes. The opposing effects were attributed to physiological differences between the two cell lines. The 3T3-L1 preadipocytes are dependent on mitotic clonal expansion (MCE) to drive differentiation, while C3H/10T1/2MSCs and human preadipocytes are not. Additionally, 4-NP downregulated β-catenin expression in C3H/10T1/2 adipocytes. Accordingly, we hypothesized that 4-NP promotes adipogenesis. The role of the canonical Wnt pathway in the promotion of adipogenesis by 4-NP requires further validation. This study provides new insights into the mechanisms and appropriate risk management of 4-NP.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Toxicology, School of Public Health, Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing, China
| | - Shuang Wu
- Department of Toxicology, School of Public Health, Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing, China
| | - Qianqian Xiao
- Department of Toxicology, School of Public Health, Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing, China
| | - Chenping Kang
- Department of Toxicology, School of Public Health, Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing, China
| | - Hong Hu
- Department of Toxicology, School of Public Health, Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing, China
| | - Xiaohong Hou
- Department of Toxicology, School of Public Health, Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing, China
| | - Xuetao Wei
- Department of Toxicology, School of Public Health, Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing, China
| | - Weidong Hao
- Department of Toxicology, School of Public Health, Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing, China
| |
Collapse
|
4
|
Wongtrakul-Kish K, Herbert BR, Packer NH. Bisecting GlcNAc Protein N-Glycosylation Is Characteristic of Human Adipogenesis. J Proteome Res 2020; 20:1313-1327. [PMID: 33383989 DOI: 10.1021/acs.jproteome.0c00702] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Human adipose tissue contains a major source of adipose-derived stem cells (ADSCs) that have the ability to differentiate into various cell types: in vitro, ADSCs can differentiate into mesenchymal lineages including adipocytes, while in vivo, ADSCs become mature adipocytes. Protein glycosylation has been shown to change in stem cell differentiation, and while ADSCs have been acknowledged for their therapeutic potential, little is known about protein glycosylation during human ADSC adipogenic differentiation. In the present study, the global membrane protein glycosylation of native adipocytes was compared to ADSCs from the same individuals as a model of in vivo adipogenesis. For in vitro adipogenesis, ADSCs were adipogenically differentiated in cell culture using an optimized, large-scale differentiation procedure. The membrane glycome of the differentiated ADSCs (dADSCs) was compared with mature adipocytes and the progenitor ADSCs. A total of 137 glycan structures were characterized across the three cell types using PGC-LC coupled with negative-ion electrospray ionization mass spectrometry (ESI-MS)/MS. Significantly higher levels of bisecting GlcNAc-type N-glycans were detected in mature adipocytes (32.1% of total glycans) and in in vitro dADSC progeny (1.9% of total glycans) compared to ADSCs. This was further correlated by the mRNA expression of the MGAT3 gene responsible for the enzymatic synthesis of this structural type. The bisecting GlcNAc structures were found on the majority of human native adipocyte membrane proteins, suggesting an important role in human adipocyte biology. Core fucosylation was also significantly increased during in vivo adipogenesis but did not correlate with an increase in Fut8 gene transcript. Unexpectedly, low abundance structures carrying rare β-linked Gal-Gal termini were also detected. Overall, the N-glycan profiles of the in vitro differentiated progeny did not reflect native adipocytes, and the results show that bisecting GlcNAc structures are a characteristic feature of human adipocyte membrane protein N-glycosylation. Raw MS files are available on GlycoPOST (ID: GPST000153 https://glycopost.glycosmos.org/).
Collapse
Affiliation(s)
- Katherine Wongtrakul-Kish
- Biomolecular Discovery Research Centre, Department of Molecular Sciences, Macquarie University, Sydney, NSW 2109, Australia.,ARC Centre for Nanoscale BioPhotonics, Department of Molecular Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Benjamin R Herbert
- Biomolecular Discovery Research Centre, Department of Molecular Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Nicolle H Packer
- Biomolecular Discovery Research Centre, Department of Molecular Sciences, Macquarie University, Sydney, NSW 2109, Australia.,ARC Centre for Nanoscale BioPhotonics, Department of Molecular Sciences, Macquarie University, Sydney, NSW 2109, Australia
| |
Collapse
|
5
|
Larsen MC, Almeldin A, Tong T, Rondelli CM, Maguire M, Jaskula-Sztul R, Jefcoate CR. Cytochrome P4501B1 in bone marrow is co-expressed with key markers of mesenchymal stem cells. BMS2 cell line models PAH disruption of bone marrow niche development functions. Toxicol Appl Pharmacol 2020; 401:115111. [PMID: 32553695 PMCID: PMC7293885 DOI: 10.1016/j.taap.2020.115111] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 05/27/2020] [Accepted: 06/07/2020] [Indexed: 12/13/2022]
Abstract
Polycyclic aromatic hydrocarbons (PAHs) are ubiquitous pollutants that are metabolized to carcinogenic dihydrodiol epoxides (PAHDE) by cytochrome P450 1B1 (CYP1B1). This metabolism occurs in bone marrow (BM) mesenchymal stem cells (MSC), which sustain hematopoietic stem and progenitor cells (HSPC). In BM, CYP1B1-mediated metabolism of 7, 12-dimethylbenz[a]anthracene (DMBA) suppresses HSPC colony formation within 6 h, whereas benzo(a)pyrene (BP) generates protective cytokines. MSC, enriched from adherent BM cells, yielded the bone marrow stromal, BMS2, cell line. These cells express elevated basal CYP1B1 that scarcely responds to Ah receptor (AhR) inducers. BMS2 cells exhibit extensive transcriptome overlap with leptin receptor positive mesenchymal stem cells (Lepr+ MSC) that control the hematopoietic niche. The overlap includes CYP1B1 and the expression of HSPC regulatory factors (Ebf3, Cxcl12, Kitl, Csf1 and Gas6). MSC are large, adherent fibroblasts that sequester small HSPC and macrophage in the BM niche (Graphic abstract). High basal CYP1B1 expression in BMS2 cells derives from interactions between the Ah-receptor enhancer and proximal promoter SP1 complexes, boosted by autocrine signaling. PAH effects on BMS2 cells model Lepr+MSC niche activity. CYP1B1 metabolizes DMBA to PAHDE, producing p53-mediated mRNA increases, long after the in vivo HSPC suppression. Faster, direct p53 effects, favored by stem cells, remain possible PAHDE targets. However, HSPC regulatory factors remained unresponsive. BP is less toxic in BMS2 cells, but, in BM, CYP1A1 metabolism stimulates macrophage cytokines (Il1b > Tnfa> Ifng) within 6 h. Although absent from BMS2 and Lepr+MSC, their receptors are highly expressed. The impact of this cytokine signaling in MSC remains to be determined. BMS2 and Lepr+MSC cells co-express CYP1B1 and 12 functional niche activity markers. CYP1B1 mRNA in BMS2 cells depends on activation of SP1 coupled to an AhR enhancer unit. DMBA metabolism by CYP1B1 activates p53 gene targets in BMS2 cells far more than BP. HSPC suppression by CYP1B1 generation of PAHDE requires rapid, non-genomic targets. BMS2 and Lepr+MSC share receptors activated by BP stimulation of macrophage cytokines.
Collapse
Affiliation(s)
- Michele Campaigne Larsen
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, WI 53705, United States of America
| | - Ahmed Almeldin
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, WI 53705, United States of America; Physiology Department, Faculty of Medicine, Tanta University, Egypt
| | - Tiegang Tong
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, WI 53705, United States of America
| | - Catherine M Rondelli
- Molecular and Environmental Toxicology Center, University of Wisconsin, Madison, WI 53705, United States of America
| | - Meghan Maguire
- Endocrinology and Reproductive Physiology Program, University of Wisconsin, Madison, WI 53705, United States of America
| | - Renata Jaskula-Sztul
- Molecular and Environmental Toxicology Center, University of Wisconsin, Madison, WI 53705, United States of America
| | - Colin R Jefcoate
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, WI 53705, United States of America; Molecular and Environmental Toxicology Center, University of Wisconsin, Madison, WI 53705, United States of America; Endocrinology and Reproductive Physiology Program, University of Wisconsin, Madison, WI 53705, United States of America.
| |
Collapse
|
6
|
Aulthouse AL, Freeh E, Newstead S, Stockert AL. Part 1: A Novel Model for Three-Dimensional Culture of 3T3-L1 Preadipocytes Stimulates Spontaneous Cell Differentiation Independent of Chemical Induction Typically Required in Monolayer. Nutr Metab Insights 2019; 12:1178638819841399. [PMID: 31001061 PMCID: PMC6454649 DOI: 10.1177/1178638819841399] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 02/08/2019] [Indexed: 12/13/2022] Open
Abstract
Differences in monolayer and three-dimensional (3D) culture systems have been recognized for several years. Despite the recognized importance of 3D systems, low cost and convenience of monolayer culture are still readily used for metabolic and nutritional studies. Here, we present part 1 of a 2-part series that will highlight (1) a novel and cost-effective model for culturing 3T3-L1 preadipocytes in 3D agarose as well as (2) an initial study showing the successful use of this 3D model for experimental analysis of these cells treated with cinnamon extract while suspended in agarose. In part 1, we provide a full characterization of the model system for the 3T3-L1 cells that demonstrate the functionality and convenience of this system. Importantly, we note spontaneous differentiation to adipocytes while cultured under these methods, independent of chemical induction. We present a 2.5-week time course with rounded cells forming vacuoles as early as 24 hours and accumulation of lipid detectable by Oil Red O stain at 0.5 weeks. Serum selection, lipid volume determination, and cell size are characterized. We conclusively demonstrate adipogenesis based on a peroxisome proliferator-activated receptor γ (PPARγ) detection using immunohistochemistry (IHC) of sections from these 3D cultures. Methods, materials and recommendations are described as well as proposed benefits to the use of this culture system for 3T3-L1 cells.
Collapse
Affiliation(s)
- Amy L Aulthouse
- Department of Biological and Allied
Health Sciences, Ohio Northern University, Ada, OH, USA
| | - Ellen Freeh
- Department of Biological and Allied
Health Sciences, Ohio Northern University, Ada, OH, USA
| | - Sabrina Newstead
- Department of Biological and Allied
Health Sciences, Ohio Northern University, Ada, OH, USA
| | - Amy L Stockert
- Department of Pharmaceutical and
Biomedical Sciences, Ohio Northern University, Ada, OH, USA
| |
Collapse
|
7
|
Seo SH, Jo SM, Kim J, Lee M, Lee Y, Kang I. Peanut Sprout Extracts Attenuate Triglyceride Accumulation by Promoting Mitochondrial Fatty Acid Oxidation in Adipocytes. Int J Mol Sci 2019; 20:ijms20051216. [PMID: 30862029 PMCID: PMC6429123 DOI: 10.3390/ijms20051216] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 03/05/2019] [Accepted: 03/06/2019] [Indexed: 02/03/2023] Open
Abstract
Peanut sprouts (PS), which are germinated peanut seeds, have recently been reported to have anti-oxidant, anti-inflammatory, and anti-obesity effects. However, the underlying mechanisms by which PS modulates lipid metabolism are largely unknown. To address this question, serial doses of PS extract (PSE) were added to 3T3-L1 cells during adipocyte differentiation. PSE (25 µg/mL) significantly attenuated adipogenesis by inhibiting lipid accumulation in addition to reducing the level of adipogenic protein and gene expression with the activation of AMP-activated protein kinase (AMPK). Other adipocyte cell models such as mouse embryonic fibroblasts C3H10T1/2 and primary adipocytes also confirmed the anti-adipogenic properties of PSE. Next, we investigated whether PSE attenuated lipid accumulation in mature adipocytes. We found that PSE significantly suppressed lipogenic gene expression, while fatty acid (FA) oxidation genes were upregulated. Augmentation of FA oxidation by PSE in mature 3T3-L1 adipocytes was confirmed via a radiolabeled-FA oxidation rate experiment by measuring the conversion of [3H]-oleic acid (OA) to [3H]-H2O. Furthermore, PSE enhanced the mitochondrial oxygen consumption rate (OCR), especially maximal respiration, and beige adipocyte formation in adipocytes. In summary, PSE was effective in reducing lipid accumulation in 3T3-L1 adipocytes through mitochondrial fatty acid oxidation involved in AMPK and mitochondrial activation.
Collapse
Affiliation(s)
- Seok Hee Seo
- Department of Marine Life Science, Jeju National University, Jeju 63243, Korea.
| | - Sang-Mi Jo
- Department of Food Science and Nutrition, Jeju National University, Jeju 63243, Korea.
| | - Jiyoung Kim
- Department of Food and Nutrition, Kyungnam College of Information & Technology, Pusan 47011, Korea.
| | - Myoungsook Lee
- Department of Food and Nutrition, Sungshin Women's University, Seoul 01133, Korea.
| | - Yunkyoung Lee
- Department of Food Science and Nutrition, Jeju National University, Jeju 63243, Korea.
| | - Inhae Kang
- Department of Food Science and Nutrition, Jeju National University, Jeju 63243, Korea.
| |
Collapse
|
8
|
Raciti GA, Fiory F, Campitelli M, Desiderio A, Spinelli R, Longo M, Nigro C, Pepe G, Sommella E, Campiglia P, Formisano P, Beguinot F, Miele C. Citrus aurantium L. dry extracts promote C/ebpβ expression and improve adipocyte differentiation in 3T3-L1 cells. PLoS One 2018; 13:e0193704. [PMID: 29596447 PMCID: PMC5875749 DOI: 10.1371/journal.pone.0193704] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 02/19/2018] [Indexed: 12/17/2022] Open
Abstract
Metabolic and/or endocrine dysfunction of the white adipose tissue (WAT) contribute to the development of metabolic disorders, such as Type 2 Diabetes (T2D). Therefore, the identification of products able to improve adipose tissue function represents a valuable strategy for the prevention and/or treatment of T2D. In the current study, we investigated the potential effects of dry extracts obtained from Citrus aurantium L. fruit juice (CAde) on the regulation of 3T3-L1 cells adipocyte differentiation and function in vitro. We found that CAde enhances terminal adipocyte differentiation of 3T3-L1 cells raising the expression of CCAAT/enhancer binding protein beta (C/Ebpβ), peroxisome proliferator activated receptor gamma (Pparγ), glucose transporter type 4 (Glut4) and fatty acid binding protein 4 (Fabp4). CAde improves insulin-induced glucose uptake of 3T3-L1 adipocytes, as well. A focused analysis of the phases occurring in the pre-adipocytes differentiation to mature adipocytes furthermore revealed that CAde promotes the early differentiation stage by up-regulating C/ebpβ expression at 2, 4 and 8 h post the adipogenic induction and anticipating the 3T3-L1 cell cycle entry and progression during mitotic clonal expansion (MCE). These findings provide evidence that the exposure to CAde enhances in vitro fat cell differentiation of pre-adipocytes and functional capacity of mature adipocytes, and pave the way to the development of products derived from Citrus aurantium L. fruit juice, which may improve WAT functional capacity and may be effective for the prevention and/or treatment of T2D.
Collapse
Affiliation(s)
- Gregory Alexander Raciti
- URT of the Institute of Experimental Endocrinology and Oncology “G. Salvatore”, National Council of Research, Naples, Italy
- Department of Translational Medical Sciences, “Federico II” University of Naples, Naples, Italy
- * E-mail: (GAR); (CM)
| | - Francesca Fiory
- URT of the Institute of Experimental Endocrinology and Oncology “G. Salvatore”, National Council of Research, Naples, Italy
- Department of Translational Medical Sciences, “Federico II” University of Naples, Naples, Italy
| | - Michele Campitelli
- URT of the Institute of Experimental Endocrinology and Oncology “G. Salvatore”, National Council of Research, Naples, Italy
- Department of Translational Medical Sciences, “Federico II” University of Naples, Naples, Italy
| | - Antonella Desiderio
- URT of the Institute of Experimental Endocrinology and Oncology “G. Salvatore”, National Council of Research, Naples, Italy
- Department of Translational Medical Sciences, “Federico II” University of Naples, Naples, Italy
| | - Rosa Spinelli
- URT of the Institute of Experimental Endocrinology and Oncology “G. Salvatore”, National Council of Research, Naples, Italy
- Department of Translational Medical Sciences, “Federico II” University of Naples, Naples, Italy
| | - Michele Longo
- URT of the Institute of Experimental Endocrinology and Oncology “G. Salvatore”, National Council of Research, Naples, Italy
- Department of Translational Medical Sciences, “Federico II” University of Naples, Naples, Italy
| | - Cecilia Nigro
- URT of the Institute of Experimental Endocrinology and Oncology “G. Salvatore”, National Council of Research, Naples, Italy
- Department of Translational Medical Sciences, “Federico II” University of Naples, Naples, Italy
| | - Giacomo Pepe
- Department of Pharmacy, School of Pharmacy, University of Salerno University of Salerno, Fisciano, Italy
| | - Eduardo Sommella
- Department of Pharmacy, School of Pharmacy, University of Salerno University of Salerno, Fisciano, Italy
| | - Pietro Campiglia
- Department of Pharmacy, School of Pharmacy, University of Salerno University of Salerno, Fisciano, Italy
- European Biomedical Research Institute of Salerno, Salerno, Italy
| | - Pietro Formisano
- URT of the Institute of Experimental Endocrinology and Oncology “G. Salvatore”, National Council of Research, Naples, Italy
- Department of Translational Medical Sciences, “Federico II” University of Naples, Naples, Italy
| | - Francesco Beguinot
- URT of the Institute of Experimental Endocrinology and Oncology “G. Salvatore”, National Council of Research, Naples, Italy
- Department of Translational Medical Sciences, “Federico II” University of Naples, Naples, Italy
| | - Claudia Miele
- URT of the Institute of Experimental Endocrinology and Oncology “G. Salvatore”, National Council of Research, Naples, Italy
- Department of Translational Medical Sciences, “Federico II” University of Naples, Naples, Italy
- * E-mail: (GAR); (CM)
| |
Collapse
|
9
|
Jung MS, Lee SJ, Song Y, Jang SH, Min W, Won CK, Kim HD, Kim TH, Cho JH. Rubus crataegifolius Bunge regulates adipogenesis through Akt and inhibits high-fat diet-induced obesity in rats. Nutr Metab (Lond) 2016; 13:29. [PMID: 27123039 PMCID: PMC4847245 DOI: 10.1186/s12986-016-0091-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Accepted: 04/19/2016] [Indexed: 01/09/2023] Open
Abstract
Background Obesity is one of the greatest public health problems and major risk factors for serious metabolic diseases and significantly increases the risk of premature death. The aim of this study was to determine the inhibitory effects of Rubus crataegifolius Bunge (RCB) on adipocyte differentiation in 3 T3-L1 cells and its anti-obesity properties in high fat diet (HFD)-induced obese rats. Methods 3 T3-L1 adipocytes and HFD-induced obese rats were treated with RCB, and its effect on gene expression was analyzed using RT-PCR and Western blotting experiments. Results RCB treatment significantly inhibited adipocyte differentiation by suppressing the expression of C/EBPβ, C/EBPα, and PPARγ in the 3 T3-L1 adipocytes. Subsequently, the expression of the PPARγ target genes aP2 and fatty acid synthase (FAS) decreased following RCB treatment during adipocyte differentiation. In uncovering the specific mechanism that mediates the effects of RCB, we demonstrated that the insulin-stimulated phosphorylation of Akt strongly decreased and that its downstream substrate phospho-GSK3β was downregulated following RCB treatment in the 3 T3-L1 adipocytes. Moreover, LY294002, an inhibitor of Akt phosphorylation, exerted stronger inhibitory effects on RCB-mediated suppression of adipocyte differentiation, leading to the inhibition of adipocyte differentiation through the downregulation of Akt signaling. An HFD-induced obesity rat model was used to determine the inhibitory effects of RCB on obesity. Body weight gain and fat accumulation in adipose tissue were significantly reduced by the supplementation of RCB. Moreover, RCB treatment caused a significant decrease in adipocyte size, associated with a decrease in epididymal fat weight. The serum total cholesterol (TC) and triglyceride (TG) levels decreased in response to RCB treatment, whereas HDL cholesterol (HDL-C) increased, indicating that RCB attenuated lipid accumulation in adipose tissue in HFD-induced obese rats. Conclusion Our results demonstrate an inhibitory effect of RCB on adipogenesis through the reduction of the adipogenic factors PPARγ, C/EBPα, and phospho-Akt. RCB had a potent anti-obesity effect, reducing body weight gain in HFD-induced obese rats. Electronic supplementary material The online version of this article (doi:10.1186/s12986-016-0091-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Min-Sup Jung
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, 660-701 Korea
| | - Soo-Jung Lee
- Department of Foods and Nutrition, Gyeongsang National University, Jinju, 660-701 Korea
| | - Yuno Song
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, 660-701 Korea
| | - Sun-Hee Jang
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, 660-701 Korea
| | - Wongi Min
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, 660-701 Korea
| | - Chung-Kil Won
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, 660-701 Korea
| | - Hong-Duck Kim
- Department of Environmental Health Science, New York Medical College, Valhalla, NY 10595 USA
| | - Tae Hoon Kim
- Department of Food Science and Biotechnology, Daegu University, Gyeongsan, 38453 Korea
| | - Jae-Hyeon Cho
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, 660-701 Korea.,College of Veterinary Medicine, Gyeongsang National University, Jiju Daero 501, Jinju-city, Gyeongsangnamdo 660-701 Korea
| |
Collapse
|
10
|
Church C, Brown M, Rodeheffer MS. Conditional immortalization of primary adipocyte precursor cells. Adipocyte 2015; 4:203-11. [PMID: 26257993 DOI: 10.1080/21623945.2014.995510] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 12/01/2014] [Accepted: 12/02/2014] [Indexed: 12/17/2022] Open
Abstract
The production of new adipocytes requires the differentiation of adipocyte precursor (AP) cells residing within the adipose tissue stromal-vascular compartment. The objective was to obtain an immortalized primary adipogenic cell line derived from FACS isolated committed APs using the conditional expression of SV40 T antigen. Adipocyte precursors were isolated from white adipose tissue (WAT) using FACS to remove non-adipogenic cell populations from mice expressing a conditionally regulated SV40 T antigen. APs were maintained by continuous culture and induced to undergo adipogenic differentiation. Adipogenesis, determined by Oil Red O staining, was assessed with each passage and compared to wildtype controls. Adipogenic capability was rapidly lost with increased passage number in committed APs with concurrent reduction in cell proliferation and expression of essential late adipogenic genes, including Pparγ and C/ebpα. Thus, FACS purified committed APs have limited capability to undergo expansion and subsequent adipogenic differentiation in vitro even if they are immortalized with the SV40 T antigen.
Collapse
|
11
|
Park KS. Raspberry ketone, a naturally occurring phenolic compound, inhibits adipogenic and lipogenic gene expression in 3T3-L1 adipocytes. PHARMACEUTICAL BIOLOGY 2015; 53:870-875. [PMID: 25429790 DOI: 10.3109/13880209.2014.946059] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
CONTEXT Raspberry ketone (RK) is a natural phenolic compound of red raspberry. The dietary intake of RK has been reported to exert anti-obese actions and alter the lipid metabolism in vivo and human studies. OBJECTIVE To elucidate a possible mechanism for anti-obese actions of RK, the effects of RK on the adipogenic and lipogenic gene expression in 3T3-L1 adipocytes were investigated. MATERIALS AND METHODS 3T3-L1 maturing pre-adipocytes were treated from day 2 to day 8 of differentiation and mature adipocytes for 24 h on day 12 with 1, 10, 20, and 50 μM of RK. Triacylglycerols were assessed by spectrophotometry and gene expression by quantitative real-time polymerase chain reaction (qRT-PCR). RESULTS Treatment of adipocytes with RK suppressed adipocyte differentiation and fat accumulation in a concentration-dependent manner. RK suppressed the expression of major genes involved in the adipogenesis pathway including peroxisome proliferator-activated receptor-γ (PPARγ) and CCAAT enhancer binding protein-α (C/EBPα), which led to further down-regulation of adipocyte fatty acid-binding protein-2 (aP2). In addition, treatment with 10 μM of RK also reduced mRNA levels of lipogenic genes such as acetyl-CoA carboxylase-1 (ACC1), fatty acid synthase (FASN), and stearoyl-CoA desaturase-1 (SCD1). In mature adipocytes, RK increased the transcriptional activities of genes involved in lipolysis and the oxidative pathways including adipose triglyceride lipase (ATGL), hormone sensitive lipase (HSL), and carnitine palmitoyl transferase-1B (CPT1B). DISCUSSION AND CONCLUSION These findings suggest that RK holds great promise for an herbal medicine with the biological activities altering the lipid metabolism in 3T3-L1 adipocytes.
Collapse
Affiliation(s)
- Kyoung Sik Park
- Department of Biomedical Science, College of Science and Engineering, Cheongju University , Chungbuk , Korea
| |
Collapse
|
12
|
Chang WT, Wu CH, Hsu CL. Diallyl trisulphide inhibits adipogenesis in 3T3-L1 adipocytes through lipogenesis, fatty acid transport, and fatty acid oxidation pathways. J Funct Foods 2015. [DOI: 10.1016/j.jff.2015.05.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
13
|
Hallenborg P, Petersen RK, Feddersen S, Sundekilde U, Hansen JB, Blagoev B, Madsen L, Kristiansen K. PPARγ ligand production is tightly linked to clonal expansion during initiation of adipocyte differentiation. J Lipid Res 2014; 55:2491-500. [PMID: 25312885 DOI: 10.1194/jlr.m050658] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Adipocyte differentiation is orchestrated by the ligand-activated nuclear receptor PPARγ. Endogenous ligands comprise oxidized derivatives of arachidonic acid and structurally similar PUFAs. Although expression of PPARγ peaks in mature adipocytes, ligands are produced primarily at the onset of differentiation. Concomitant with agonist production, murine fibroblasts undergo two rounds of mitosis referred to as mitotic clonal expansion. Here we show that mouse embryonic fibroblasts deficient in either of two cell cycle inhibitors, the transcription factor p53 or its target gene encoding the cyclin-dependent kinase inhibitor p21, exhibit increased adipogenic potential. The antiadipogenic effect of p53 relied on its transcriptional activity and p21 expression but was circumvented by administration of an exogenous PPARγ agonist suggesting a linkage between cell cycling and PPARγ ligand production. Indeed, cell cycle inhibitory compounds decreased PPARγ ligand production in differentiating 3T3-L1 preadipocytes. Furthermore, these inhibitors abolished the release of arachidonic acid induced by the hormonal cocktail initiating adipogenesis. Collectively, our results suggest that murine fibroblasts require clonal expansion for PPARγ ligand production at the onset of adipocyte differentiation.
Collapse
Affiliation(s)
- Philip Hallenborg
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | | | - Søren Feddersen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Ulrik Sundekilde
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Jacob B Hansen
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Blagoy Blagoev
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Lise Madsen
- Department of Biology, University of Copenhagen, Copenhagen, Denmark National Institute of Nutrition and Seafood Research, Bergen, Norway
| | | |
Collapse
|
14
|
Guo B, Chatterjee S, Li L, Kim JM, Lee J, Yechoor VK, Minze LJ, Hsueh W, Ma K. The clock gene, brain and muscle Arnt-like 1, regulates adipogenesis via Wnt signaling pathway. FASEB J 2012; 26:3453-63. [PMID: 22611086 PMCID: PMC6137895 DOI: 10.1096/fj.12-205781] [Citation(s) in RCA: 133] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Accepted: 05/01/2012] [Indexed: 12/22/2022]
Abstract
Circadian clocks in adipose tissue are known to regulate adipocyte biology. Although circadian dysregulation is associated with development of obesity, the underlying mechanism has not been established. Here we report that disruption of the clock gene, brain and muscle Arnt-like 1 (Bmal1), in mice led to increased adipogenesis, adipocyte hypertrophy, and obesity, compared to wild-type (WT) mice. This is due to its cell-autonomous effect, as Bmal1 deficiency in embryonic fibroblasts, as well as stable shRNA knockdown (KD) in 3T3-L1 preadipocyte and C3H10T1/2 mesenchymal stem cells, promoted adipogenic differentiation. We demonstrate that attenuation of Bmal1 function resulted in down-regulation of genes in the canonical Wnt pathway, known to suppress adipogenesis. Promoters of these genes (Wnt10a, β-catenin, Dishevelled2, TCF3) displayed Bmal1 occupancy, indicating direct circadian regulation by Bmal1. As a result, Wnt signaling activity was attenuated by Bmal1 KD and augmented by its overexpression. Furthermore, stabilizing β-catenin through Wnt ligand or GSK-3β inhibition achieved partial restoration of blunted Wnt activity and suppression of increased adipogenesis induced by Bmal1 KD. Taken together, our study demonstrates that Bmal1 is a critical negative regulator of adipocyte development through transcriptional control of components of the canonical Wnt signaling cascade, and provides a mechanistic link between circadian disruption and obesity.
Collapse
Affiliation(s)
- Bingyan Guo
- Center for Diabetes Research, Department of Medicine, The Methodist Hospital Research Institute, Houston, Texas, USA
- Department of Cardiovascular Medicine, Second Affiliated Hospital, Hebei Medical University, Shijiazhuang, Hebei, China; and
| | - Somik Chatterjee
- Center for Diabetes Research, Department of Medicine, The Methodist Hospital Research Institute, Houston, Texas, USA
| | - Lifei Li
- Center for Diabetes Research, Department of Medicine, The Methodist Hospital Research Institute, Houston, Texas, USA
| | - Ji M. Kim
- Center for Diabetes Research, Department of Medicine, The Methodist Hospital Research Institute, Houston, Texas, USA
| | - Jeongkyung Lee
- Diabetes and Endocrinology Research Center, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Vijay K. Yechoor
- Diabetes and Endocrinology Research Center, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Laurie J. Minze
- Center for Diabetes Research, Department of Medicine, The Methodist Hospital Research Institute, Houston, Texas, USA
| | - Willa Hsueh
- Center for Diabetes Research, Department of Medicine, The Methodist Hospital Research Institute, Houston, Texas, USA
| | - Ke Ma
- Center for Diabetes Research, Department of Medicine, The Methodist Hospital Research Institute, Houston, Texas, USA
| |
Collapse
|
15
|
Liu XF, Bera TK, Kahue C, Escobar T, Fei Z, Raciti GA, Pastan I. ANKRD26 and its interacting partners TRIO, GPS2, HMMR and DIPA regulate adipogenesis in 3T3-L1 cells. PLoS One 2012; 7:e38130. [PMID: 22666460 PMCID: PMC3364200 DOI: 10.1371/journal.pone.0038130] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Accepted: 05/04/2012] [Indexed: 01/04/2023] Open
Abstract
Partial inactivation of the Ankyrin repeat domain 26 (Ankrd26) gene causes obesity and diabetes in mice and increases spontaneous and induced adipogenesis in mouse embryonic fibroblasts. However, it is not yet known how the Ankrd26 protein carries out its biological functions. We identified by yeast two-hybrid and immunoprecipitation assays the triple functional domain protein (TRIO), the G protein pathway suppressor 2 (GPS2), the delta-interacting protein A (DIPA) and the hyaluronan-mediated motility receptor (HMMR) as ANKRD26 interacting partners. Adipogenesis of 3T3-L1 cells was increased by selective down-regulation of Ankrd26, Trio, Gps2, Hmmr and Dipa. Furthermore, GPS2 and DIPA, which are normally located in the nucleus, were translocated to the cytoplasm, when the C-terminus of ANKRD26 was introduced into these cells. These findings provide biochemical evidence that ANKRD26, TRIO, GPS2 and HMMR are novel and important regulators of adipogenesis and identify new targets for the modulation of adipogenesis.
Collapse
Affiliation(s)
- Xiu-Fen Liu
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Tapan K. Bera
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Charissa Kahue
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Thelma Escobar
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Zhaoliang Fei
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Gregory A. Raciti
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Ira Pastan
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
16
|
Shin D, Howng SYB, Ptáček LJ, Fu YH. miR-32 and its target SLC45A3 regulate the lipid metabolism of oligodendrocytes and myelin. Neuroscience 2012; 213:29-37. [PMID: 22521588 DOI: 10.1016/j.neuroscience.2012.03.054] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Revised: 03/15/2012] [Accepted: 03/19/2012] [Indexed: 01/07/2023]
Abstract
Oligodendrocytes generate large amounts of myelin by extension of their cell membranes. Though lipid is the major component of myelin, detailed lipid metabolism in the maintenance of myelin is not understood. We reported previously that miR-32 might be involved in myelin maintenance (Shin et al., 2009). Here we demonstrate a novel role for miR-32 in oligodendrocyte function and development through the regulation of SLC45A3 (solute carrier family 45, member 3) and other downstream targets such as CLDN-11. miR-32 is highly expressed in the myelin-enriched regions of the brain and mature oligodendrocytes, and it promotes myelin protein expression. We found that miR-32 directly regulates the expression of SLC45A3 by binding to the complementary sequence on the 3'UTR of cldn11 and slc45a3. As a myelin-enriched putative sugar transporter, SLC45A3 enhances intracellular glucose levels and the synthesis of long-chain fatty acids. Therefore, overexpression of SLC45A3 triggers neutral lipid accumulation. Interestingly, both overexpression and suppression of SLC45A3 reduces myelin protein expression in mature oligodendrocytes and alters oligodendrocyte morphology, indicating that tight regulation of SLC45A3 expression is necessary for the proper maintenance of myelin proteins and structure. Taken together, our data suggest that miR-32 and its downstream target SLC45A3 play important roles in myelin maintenance by modulating glucose and lipid metabolism and myelin protein expression in oligodendrocytes.
Collapse
Affiliation(s)
- D Shin
- Department of Neurology, University of California San Francisco, 1550 Fourth Street, San Francisco, CA 94158, USA
| | | | | | | |
Collapse
|
17
|
Kim EJ, Lee DH, Kim HJ, Lee SJ, Ban JO, Cho MC, Jeong HS, Yang Y, Hong JT, Yoon DY. Thiacremonone, a sulfur compound isolated from garlic, attenuates lipid accumulation partially mediated via AMPK activation in 3T3-L1 adipocytes. J Nutr Biochem 2012; 23:1552-8. [PMID: 22405697 DOI: 10.1016/j.jnutbio.2011.10.008] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Revised: 10/01/2011] [Accepted: 10/19/2011] [Indexed: 12/24/2022]
Abstract
Garlic extracts exert anti-cancer and anti-inflammatory effects. However, the anti-adipogenic effect of garlic-derived compounds remains unclear. In this study, we examined the effect of thiacremonone, a sulfur compound isolated from garlic, on adipocyte differentiation using 3T3-L1 cells. We found that thiacremonone significantly inhibited 3T3-L1 differentiation via down-regulation of adipogenesis-related transcription factors and adipogenic markers. The inhibitory effect mainly occurred at the early phase of differentiation in 3T3-L1 cells. There was no cytotoxic effect of thiacremonone in 3T3-L1 cells and treatment of differentiating 3T3-L1 cells with thiacremonone resulted in AMPK activation, which led to an attenuated expression of acetyl CoA carboxylase-1 (ACC-1), an essential enzyme for the synthesis and usage of fatty acids. Moreover, thiacremonone enhanced the mRNA level of carnitine palmitoyltransferase (CPT-1). The modulating effect of thiacremonone on expressions of genes involved in lipolysis was partially abrogated by treatment with compound C, an AMPK inhibitor. Taken together, these results indicated that thiacremonone-induced AMPK activation, inhibition of ACC-1 expression and concomitant recovery of CPT-1 expression resulted in the suppression of intracellular lipid droplet levels, suggesting that thiacremonone may induce reduction of lipid synthesis and increases in fatty acid oxidation partially mediated via AMPK activation. Thiacremonone may be a promising compound for the treatment of obesity.
Collapse
Affiliation(s)
- Eun Jin Kim
- Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University, Hwayang-dong 1, Gwangjin-gu, Seoul 143-701, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Muruganandan S, Parlee SD, Rourke JL, Ernst MC, Goralski KB, Sinal CJ. Chemerin, a novel peroxisome proliferator-activated receptor gamma (PPARgamma) target gene that promotes mesenchymal stem cell adipogenesis. J Biol Chem 2011; 286:23982-95. [PMID: 21572083 DOI: 10.1074/jbc.m111.220491] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Chemerin is an adipocyte-secreted protein that regulates adipogenesis and the metabolic function of mature adipocytes via activation of chemokine-like receptor 1 (CMKLR1). Herein we report the interaction of peroxisome proliferator-activated receptor γ (PPARγ) and chemerin in the context of adipogenesis. Knockdown of chemerin or CMKLR1 expression or antibody neutralization of secreted chemerin protein arrested adipogenic clonal expansion of bone marrow mesenchymal stem cells (BMSCs) by inducing a loss of G(2)/M cyclins (cyclin A2/B2) but not the G(1)/S cyclin D2. Forced expression of PPARγ in BMSCs did not completely rescue this loss of clonal expansion and adipogenesis following chemerin or CMKLR1 knockdown. However, forced expression and/or activation of PPARγ in BMSCs as well as non-adipogenic cell types such as NIH-3T3 embryonic fibroblasts and MCA38 colon carcinoma cells significantly induced chemerin expression and secretion. Sequence analysis revealed a putative PPARγ response element (PPRE) sequence within the chemerin promoter. This PPRE was able to confer PPARγ responsiveness on a heterologous promoter, and mutation of this sequence abolished activation of the chemerin promoter by PPARγ. Chromatin immunoprecipitation confirmed the direct association of PPARγ with this PPRE. Treatment of mice with rosiglitazone elevated chemerin mRNA levels in adipose tissue and bone marrow coincident with an increase in circulating chemerin levels. Together, these findings support a fundamental role for chemerin/CMKLR1 signaling in clonal expansion during adipocyte differentiation as well as a role for PPARγ in regulating chemerin expression.
Collapse
|
19
|
Marchildon F, St-Louis C, Akter R, Roodman V, Wiper-Bergeron NL. Transcription factor Smad3 is required for the inhibition of adipogenesis by retinoic acid. J Biol Chem 2010; 285:13274-84. [PMID: 20179325 DOI: 10.1074/jbc.m109.054536] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The process of adipocyte differentiation is driven by a highly coordinated cascade of transcriptional events that results in the development of the mature adipocyte and in lipid accumulation. One of the early events of differentiation is the up-regulation of CCAAT/enhancer-binding protein beta (C/EBPbeta) expression. C/EBPbeta then acts to up-regulate the expression of adipogenic factors such as C/EBPalpha, which control the late stage of adipogenesis. Retinoic acid (RA) is a potent inhibitor of adipogenesis, and its action appears to block C/EBPbeta transcriptional potential early during differentiation. Using preadipocytes and mesenchymal stem cell models, we show that RA specifically blocks the occupancy of C/EBPbeta of the Cebpa promoter, thereby abrogating the differentiation process. RA does not act directly on C/EBPbeta but rather stimulates the expression of the transforming growth factor beta-effector protein Smad3, which can interact with C/EBPbeta via its Mad homology 1 domain and can interfere with C/EBPbeta DNA binding. The RA-induced increase in Smad3 expression results in increased cytoplasmic and nuclear Smad3, an important event as ectopic expression of Smad3 in preadipocytes in the absence of RA treatment only modestly inhibits adipogenesis and C/EBPbeta DNA binding, suggesting that Smad3 alone is not sufficient to completely recapitulate the effects of retinoic acid treatment during differentiation. However, in the absence of Smad3, RA is not able to inhibit adipocyte differentiation or to elicit a decrease in C/EBPbeta DNA occupancy suggesting that Smad3 is necessary to convey the inhibitory effects of retinoic acid during adipogenesis.
Collapse
Affiliation(s)
- François Marchildon
- Graduate Program in Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | | | | | | | | |
Collapse
|
20
|
Hallenborg P, Feddersen S, Madsen L, Kristiansen K. The tumor suppressors pRB and p53 as regulators of adipocyte differentiation and function. Expert Opin Ther Targets 2009; 13:235-46. [DOI: 10.1517/14712590802680141] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|