1
|
Teufelsbauer M, Stickler S, Hammond DC, Hamilton G. Vascular Endothelial Growth Factor Expression of Adipose-Derived Stromal Cells and Adipocytes Initiated from Fat Aspirations. Aesthetic Plast Surg 2024:10.1007/s00266-024-04587-w. [PMID: 39658669 DOI: 10.1007/s00266-024-04587-w] [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: 10/02/2024] [Accepted: 11/20/2024] [Indexed: 12/12/2024]
Abstract
BACKGROUND Fat grafting is frequently employed in aesthetic and reconstructive plastic surgery with a low complication rate. However, fat necrosis may occur in dependence of the mode of fat aspiration, processing of the tissue and graft size. Graft survival is critically dependent on the contained adipose-derived stromal cells (ADSCs), adipocyte precursors and their potential for vascular supply. This work investigated the potential role of the expression of vascular endothelial growth factor A (VEGF) and various cytokines by ADSCs and differentiated adipocytes as key factors of fat grafting. METHODS Adipokine expression of ADSCs and differentiated adipocytes were assessed using Proteome Profiler Arrays that detect 58 relevant proteins. RESULTS Collected fat grafts could be categorized according to their adipokine expression into VEGFhigh and VEGFlow ADSCs groups, the former exhibiting higher content of VEGF-related angiopoietin-like 2, nidogen-1/entactin, CCL2/MCP-1 and elevated expression of IGFBPs in association with a fourfold higher VEGF expression. Differentiation of ADSCs into adipocytes increased VEGF concentrations in VEGFlow ADSCs but not in ADSCs exhibiting initial high VEGF concentrations. The adipocytes revealed high expression of HGF, leptin, CCL2/MCP-1, nidogen-1/entactin, M-CSF but lower induction of angiopoietin-like 2. CONCLUSION Half of the ADSCs from fat grafts express high concentrations of VEGF and other adipokines that support angiogenesis and survival of this tissues following transfer. Differentiation of ADSClow cells to adipocytes may make up for the initially low VEGF expression, but this activation is 7-10 days delayed compared to the VEGFhigh ADSC cells and may fail to support angiogenesis from the beginning. NO LEVEL ASSIGNED This journal requires that authors assign a level of evidence to each submission to which Evidence-Based Medicine rankings are applicable. This excludes Review Articles, Book Reviews, and manuscripts that concern Basic Science, Animal Studies, Cadaver Studies, and Experimental Studies. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266 .
Collapse
Affiliation(s)
- Maryana Teufelsbauer
- Clinics of Plastic and Reconstructive Surgery, Medical University of Vienna, 1090, Vienna, Austria
| | - Sandra Stickler
- Institute of Pharmacology, Medical University of Vienna, Waehringerstraße 13a, 1090, Vienna, Austria
| | - Dennis C Hammond
- Center for Breast and Body Contouring, Grand Rapids, MI, 49546, USA
| | - Gerhard Hamilton
- Institute of Pharmacology, Medical University of Vienna, Waehringerstraße 13a, 1090, Vienna, Austria.
| |
Collapse
|
2
|
Hong S, Park S, Lee J, Park S, Park J, Lee Y. Anti-Obesity Effects of Pleurotus ferulae Water Extract on 3T3-L1 Adipocytes and High-Fat-Diet-Induced Obese Mice. Nutrients 2024; 16:4139. [PMID: 39683533 DOI: 10.3390/nu16234139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 11/27/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
This study offers promising insights into the anti-obesity potential of Pleurotus ferulae, an edible mushroom valued in Asian cuisine for its nutritional benefits. A hot water extract of P. ferulae (PWE) administered to high-fat diet-induced obese mice over an 8-week period significantly reduced their body weight gain and fat accumulation. PWE not only improved the body weight metrics but also positively influenced the serum lipid profile of obese mice by lowering their total cholesterol and low-density lipoprotein cholesterol levels. In vitro studies using 3T3-L1 adipocytes showed that PWE inhibited adipocyte differentiation and lipid accumulation by downregulating key adipogenic transcription factors, particularly PPARγ and C/EBPα, as well as related lipogenic genes involved in fat synthesis and storage, such as Fabp4, Fasn, and Scd1. Chemical analysis revealed that PWE is rich in polysaccharides, which have been associated with various health benefits, including anti-obesity, anti-diabetic, and anti-cancer properties. These findings suggest that the bioactive compounds in PWE may serve as functional food components that could potentially be applied for the prevention and management of obesity and other metabolic disorders.
Collapse
Affiliation(s)
- Seulmin Hong
- Food Functionality Research Division, Korea Food Research Institute (KFRI), Wanju-gun, Jeonbuk-do 55365, Republic of Korea
- Department of Food Science and Technology, Chung-Ang University, Anseong, Gyeonggi-do 17546, Republic of Korea
| | - Seonkyeong Park
- Food Functionality Research Division, Korea Food Research Institute (KFRI), Wanju-gun, Jeonbuk-do 55365, Republic of Korea
| | - Jangho Lee
- Food Functionality Research Division, Korea Food Research Institute (KFRI), Wanju-gun, Jeonbuk-do 55365, Republic of Korea
| | - Soohyun Park
- Food Functionality Research Division, Korea Food Research Institute (KFRI), Wanju-gun, Jeonbuk-do 55365, Republic of Korea
| | - Jaeho Park
- Food Functionality Research Division, Korea Food Research Institute (KFRI), Wanju-gun, Jeonbuk-do 55365, Republic of Korea
| | - Yugeon Lee
- Food Functionality Research Division, Korea Food Research Institute (KFRI), Wanju-gun, Jeonbuk-do 55365, Republic of Korea
| |
Collapse
|
3
|
Pollard AE. New concepts in the roles of AMPK in adipocyte stem cell biology. Essays Biochem 2024; 68:349-361. [PMID: 39175418 DOI: 10.1042/ebc20240008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 08/09/2024] [Accepted: 08/13/2024] [Indexed: 08/24/2024]
Abstract
Obesity is a major risk factor for many life-threatening diseases. Adipose tissue dysfunction is emerging as a driving factor in the transition from excess adiposity to comorbidities such as metabolic-associated fatty liver disease, cardiovascular disease, Type 2 diabetes and cancer. However, the transition from healthy adipose expansion to the development of these conditions is poorly understood. Adipose stem cells, residing in the vasculature and stromal regions of subcutaneous and visceral depots, are responsible for the expansion and maintenance of organ function, and are now recognised as key mediators of pathological transformation. Impaired tissue expansion drives inflammation, dysregulation of endocrine function and the deposition of lipids in the liver, muscle and around vital organs, where it is toxic. Contrary to previous hypotheses, it is the promotion of healthy adipose tissue expansion and function, not inhibition of adipogenesis, that presents the most attractive therapeutic strategy in the treatment of metabolic disease. AMP-activated protein kinase, a master regulator of energy homeostasis, has been regarded as one such target, due to its central role in adipose tissue lipid metabolism, and its apparent inhibition of adipogenesis. However, recent studies utilising AMP-activated protein kinase (AMPK)-specific compounds highlight a more subtle, time-dependent role for AMPK in the process of adipogenesis, and in a previously unexplored repression of leptin, independent of adipocyte maturity. In this article, I discuss historic evidence for AMPK-mediated adipogenesis inhibition and the multi-faceted roles for AMPK in adipose tissue.
Collapse
Affiliation(s)
- Alice E Pollard
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, U.K
| |
Collapse
|
4
|
Zhang W, Raza SHA, Li B, Yang W, Khan R, Aloufi BH, Zhang G, Zuo F, Zan L. LncBNIP3 Inhibits Bovine Intramuscular Preadipocyte Differentiation via the PI3K-Akt and PPAR Signaling Pathways. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:24260-24271. [PMID: 39453846 DOI: 10.1021/acs.jafc.4c05383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2024]
Abstract
Intramuscular fat (IMF) content is an economic trait in beef cattle that improves the meat quality. Studies have highlighted the correlation between long noncoding RNAs (lncRNAs) and IMF development. In this study, lncBNIP3 knockdown promoted bovine intramuscular preadipocyte differentiation. RNA-seq analysis of intramuscular preadipocytes with lncBNIP3 knockdown identified 230 differentially expressed genes. The PI3K-Akt and PPAR signaling pathways were enriched. lncBNIP3 interference promoted mRNA and protein expression of key genes in PI3K-Akt signaling pathway. LncBNIP3 interference reversed the effects of an AKT-inhibitor MK-2206 on Akt protein expression and lipid droplet accumulation, promoted mRNA and protein expression of essential genes in the PPAR signaling pathway, and ameliorated the inhibitory effects of a PPARg antagonist GW9662 on lipid accumulation. Therefore, lncBNIP3 inhibition of bovine intramuscular preadipocyte differentiation is likely mediated via the PI3K-Akt and PPAR signaling pathways. This study identified a valuable lncRNA with functional roles in IMF accumulation and revealed new strategies to improve beef quality.
Collapse
Affiliation(s)
- Wenzhen Zhang
- College of Animal Science and Technology, Southwest University, Rongchang, Chongqing 402460, China
| | - Sayed Haidar Abbas Raza
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, P.R. China
- Guangdong Provincial Key Laboratory of Food Quality and Safety, South China Agricultural University, Guangzhou 510642, P.R. China
| | - Bingzhi Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, P.R. China
- College of Animal Engineering, Yangling Vocational and Technical College, Yangling, Shaanxi 712100, P.R. China
| | - Wucai Yang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, P.R. China
| | - Rajwali Khan
- Department of Livestock Management, Breeding and Genetics, The University of Agriculturer, Peshawar 25130, Pakistan
| | - Bandar Hamad Aloufi
- Department of Biology, College of Science, University of Hail, P.O. Box 2440, Ha'il 2440, Saudi Arabia
| | - Gongwei Zhang
- College of Animal Science and Technology, Southwest University, Rongchang, Chongqing 402460, China
| | - Fuyuan Zuo
- College of Animal Science and Technology, Southwest University, Rongchang, Chongqing 402460, China
| | - Linsen Zan
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, P.R. China
| |
Collapse
|
5
|
Choi M, Jeong K, Pak Y. Caveolin-2 controls preadipocyte survival in the mitotic clonal expansion for adipogenesis. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119793. [PMID: 39038612 DOI: 10.1016/j.bbamcr.2024.119793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 06/11/2024] [Accepted: 06/27/2024] [Indexed: 07/24/2024]
Abstract
Here, we report that Caveolin-2 (Cav-2) is a cell cycle regulator in the mitotic clonal expansion (MCE) for adipogenesis. For the G2/M phase transition and re-entry into the G1 phase, dephosphorylated Cav-2 by protein tyrosine phosphatase 1B (PTP1B) controlled epigenetic activation of Ccnb1, Cdk1, and p21 in a lamin A/C-dependent manner, thereby ensuring the survival of preadipocytes. Cav-2, associated with lamin A/C, recruited the repressed promoters of Ccnb1 and Cdk1 for activation, and disengaged the active promoter of p21 from lamin A/C for inactivation through histone H3 modifications at the nuclear periphery. Cav-2 deficiency abrogated the histone H3 modifications and impeded the transactivation of Ccnb1, Cdk1, and p21, leading to a delay in mitotic entry, retardation of re-entry into G1 phase, and the apoptotic cell death of preadipocytes. Re-expression of Cav-2 restored the G2/M phase transition and G1 phase re-entry, preadipocyte survival, and adipogenesis in Cav-2-deficient preadipocytes. Our study uncovers a novel mechanism by which cell cycle transition and apoptotic cell death are controlled for adipocyte hyperplasia.
Collapse
Affiliation(s)
- Moonjeong Choi
- Division of Life Science, Graduate School of Applied Life Science, PMBBRC, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Kyuho Jeong
- Department of Biochemistry, College of Medicine, Dongguk University, Gyeongju 38066, Republic of Korea
| | - Yunbae Pak
- Division of Life Science, Graduate School of Applied Life Science, PMBBRC, Gyeongsang National University, Jinju 52828, Republic of Korea.
| |
Collapse
|
6
|
Kim HY, Jang HJ, Muthamil S, Shin UC, Lyu JH, Kim SW, Go Y, Park SH, Lee HG, Park JH. Novel insights into regulators and functional modulators of adipogenesis. Biomed Pharmacother 2024; 177:117073. [PMID: 38981239 DOI: 10.1016/j.biopha.2024.117073] [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: 04/15/2024] [Revised: 06/27/2024] [Accepted: 06/29/2024] [Indexed: 07/11/2024] Open
Abstract
Adipogenesis is a process that differentiates new adipocytes from precursor cells and is tightly regulated by several factors, including many transcription factors and various post-translational modifications. Recently, new roles of adipogenesis have been suggested in various diseases. However, the molecular mechanisms and functional modulation of these adipogenic genes remain poorly understood. This review summarizes the regulatory factors and modulators of adipogenesis and discusses future research directions to identify novel mechanisms regulating adipogenesis and the effects of adipogenic regulators in pathological conditions. The master adipogenic transcriptional factors PPARγ and C/EBPα were identified along with other crucial regulatory factors such as SREBP, Kroxs, STAT5, Wnt, FOXO1, SWI/SNF, KLFs, and PARPs. These transcriptional factors regulate adipogenesis through specific mechanisms, depending on the adipogenic stage. However, further studies related to the in vivo role of newly discovered adipogenic regulators and their function in various diseases are needed to develop new potent therapeutic strategies for metabolic diseases and cancer.
Collapse
Affiliation(s)
- Hyun-Yong Kim
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do 58245, Republic of Korea; New Drug Development Center, Osong Medical Innovation Foundation, 123, Osongsaengmyeong-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do 28160, Republic of Korea.
| | - Hyun-Jun Jang
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do 58245, Republic of Korea; Research Group of Personalized Diet, Korea Food Research Institute, Wanju-gun, Jeollabuk-do 55365, Republic of Korea.
| | - Subramanian Muthamil
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do 58245, Republic of Korea.
| | - Ung Cheol Shin
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do 58245, Republic of Korea.
| | - Ji-Hyo Lyu
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do 58245, Republic of Korea.
| | - Seon-Wook Kim
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do 58245, Republic of Korea.
| | - Younghoon Go
- Korean Medicine (KM)-application Center, Korea Institute of Oriental Medicine, Daegu 41062, Republic of Korea.
| | - Seong-Hoon Park
- Genetic and Epigenetic Toxicology Research Group, Korea Institute of Toxicology, Daejeon 34141, Republic of Korea.
| | - Hee Gu Lee
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea.
| | - Jun Hong Park
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do 58245, Republic of Korea; University of Science & Technology (UST), KIOM campus, Korean Convergence Medicine Major, Daejeon 34054, Republic of Korea.
| |
Collapse
|
7
|
Wu Y, Sun Y, Song Y, Wang J, Han Y, Yang N, Lin H, Yin Y, Han X. PPA1 promotes adipogenesis by regulating the stability of C/EBPs. Cell Death Differ 2024; 31:1044-1056. [PMID: 38762596 PMCID: PMC11303681 DOI: 10.1038/s41418-024-01309-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 04/24/2024] [Accepted: 05/01/2024] [Indexed: 05/20/2024] Open
Abstract
Adipogenesis significantly contributes to healthy adipose tissue expansion in obesity. Increasing adipocyte number or function to alleviate adipose tissue overload could serve as a therapeutic strategy for both lipodystrophy and obesity-related metabolic syndrome. Inorganic pyrophosphatase (PPA1) is an enzyme that catalyzes the hydrolysis of pyrophosphate (PPi) and is involved in many biochemical reactions, but its function in adipose tissue has not been studied previously. In this study, we demonstrated that adipose-specific PPA1 knockout (PPA1AKO) mice showed lipodystrophy and spontaneously developed hepatic steatosis and severe insulin resistance under normal chow diet feeding. PPA1 deficiency suppressed the differentiation of primary adipocyte precursors and 3T3-L1 cells. Notably, PPA1 overexpression can restore inhibited adipogenesis in preadipocytes isolated from db/db mice and type 2 diabetes patients. Mechanistic studies have revealed that PPA1 acts as a positive regulator of early adipocyte differentiation by promoting CCAAT/enhancer-binding proteinβ and δ (C/EBPβ and δ) protein stability. Moreover, the function of PPA1 in adipogenesis is independent of its PPi catalytic activity. Collectively, our in vivo and in vitro findings demonstrated that PPA1 is a novel critical upstream regulator of adipogenesis, controlling adipose tissue development and whole-body metabolic homeostasis.
Collapse
Affiliation(s)
- Yangyang Wu
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, China
- Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing Medical University, Nanjing, China
| | - Yue Sun
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yuqing Song
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jiateng Wang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ye Han
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Nan Yang
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Haiyan Lin
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Ye Yin
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Xiao Han
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
8
|
Peng H, Lin X, Wang Y, Chen J, Zhao Q, Chen S, Cheng Q, Chen C, Sang T, Zhou H, Xiao J, Wang W, Fang L, Wang X. Epigallocatechin gallate suppresses mitotic clonal expansion and adipogenic differentiation of preadipocytes through impeding JAK2/STAT3-mediated transcriptional cascades. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 129:155563. [PMID: 38552377 DOI: 10.1016/j.phymed.2024.155563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 02/03/2024] [Accepted: 03/21/2024] [Indexed: 05/30/2024]
Abstract
BACKGROUND Mitotic clonal expansion (MCE) is a prerequisite for preadipocyte differentiation and adipogenesis. Epigallocatechin gallate (EGCG) has been shown to inhibit preadipocyte differentiation. However, the exact molecular mechanisms are still elusive. PURPOSE This study investigated whether EGCG could inhibit adipogenesis and lipid accumulation by regulating the cell cycle in the MCE phase of adipogenesis and its underlying molecular mechanisms. METHOD 3T3-L1 preadipocytes were induced to differentiate by a differentiation cocktail (DMI) and were treated with EGCG (25-100 μM) for 9, 18, and 24 h to examine the effect on MCE, or eight days to examine the effect on terminal differentiation. C57BL/6 mice were fed a high-fat diet (HFD) for three months to induce obesity and were given EGCG (50 or 100 mg/kg) daily by gavage. RESULTS We showed that EGCG significantly inhibited terminal adipogenesis and lipid accumulation in 3T3-L1 cells and decreased expressions of PPARγ, C/EBPα, and FASN. Notably, at the MCE phase, EGCG regulated the cell cycle in sequential order, induced G0/G1 arrest at 18 h, and inhibited the G2/M phase at 24 h upon DMI treatment. Meanwhile, EGCG regulated the expressions of cell cycle regulators (cyclin D1, cyclin E1, CDK4, CDK6, cyclin B1, cyclin B2, p16, and p27), and decreased C/EBPβ, PPARγ, and C/EBPα expressions at MCE. Mechanistic studies using STAT3 agonist Colivelin and antagonist C188-9 revealed that EGCG-induced cell cycle arrest in the MCE phase and terminal adipocyte differentiation was mediated by the inhibition of JAK2/STAT3 signaling cascades and STAT3 (Tyr705) nuclear translocation. Furthermore, EGCG significantly protected mice from HFD-induced obesity, reduced body weight and lipid accumulations in adipose tissues, reduced hyperlipidemia and leptin levels, and improved glucose intolerance and insulin sensitivity. Moreover, RNA sequencing (RNA-seq) analysis showed that the cell cycle changes in epididymal white adipose tissue (eWAT) were significantly enriched upon EGCG treatment. We further verified that EGCG treatment significantly reduced expressions of adipogenic factors, cell cycle regulators, and p-STAT3 in eWAT. CONCLUSION EGCG inhibits MCE, resulting in the inhibition of early and terminal adipocyte differentiation and lipid accumulation, which were mediated by inhibiting p-STAT3 nucleus translocation and activation.
Collapse
Affiliation(s)
- He Peng
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 260 Baichuan Road, Hangzhou 311400, PR China
| | - Xiaojian Lin
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 260 Baichuan Road, Hangzhou 311400, PR China
| | - Ying Wang
- College of Food and Health, Zhejiang A&F University, Hangzhou 311300, PR China
| | - Jiajun Chen
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 260 Baichuan Road, Hangzhou 311400, PR China
| | - Qian Zhao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 260 Baichuan Road, Hangzhou 311400, PR China
| | - Shengjia Chen
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 260 Baichuan Road, Hangzhou 311400, PR China
| | - Qi Cheng
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 260 Baichuan Road, Hangzhou 311400, PR China
| | - Chaojie Chen
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 260 Baichuan Road, Hangzhou 311400, PR China
| | - Tingting Sang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 260 Baichuan Road, Hangzhou 311400, PR China
| | - Hongyu Zhou
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 260 Baichuan Road, Hangzhou 311400, PR China
| | - Jun Xiao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 260 Baichuan Road, Hangzhou 311400, PR China
| | - Wen Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 260 Baichuan Road, Hangzhou 311400, PR China
| | - Liu Fang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 260 Baichuan Road, Hangzhou 311400, PR China
| | - Xingya Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 260 Baichuan Road, Hangzhou 311400, PR China.
| |
Collapse
|
9
|
Yang Z, Lu Y, Li T, Zhou X, Yang J, Yang S, Bu S, Duan Y. Osmanthus fragrans Flavonoid Extract Inhibits Adipogenesis and Induces Beiging in 3T3-L1 Adipocytes. Foods 2024; 13:1894. [PMID: 38928836 PMCID: PMC11202805 DOI: 10.3390/foods13121894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/08/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024] Open
Abstract
Osmanthus fragrans has a long history of cultivation in Asia and is widely used in food production for its unique aroma, which has important cultural and economic values. It is rich in flavonoids with diverse pharmacological properties, such as antioxidant, anti-tumor, and anti-lipid activities. However, little is known regarding the effects of Osmanthus fragrans flavonoid extract (OFFE) on adipogenesis and pre-adipocyte transdifferentiation. Herein, this research aimed to investigate the effect of OFFE on the differentiation, adipogenesis, and beiging of 3T3-L1 adipocytes and to elucidate the underlying mechanism. Results showed that OFFE inhibited adipogenesis, reduced intracellular reactive oxygen species levels in mature adipocytes, and promoted mitochondrial biogenesis as well as beiging/browning in 3T3-L1 adipocytes. This effect was accompanied by increased mRNA and protein levels of the brown adipose-specific marker gene Pgc-1a, and the upregulation of the expression of UCP1, Cox7A1, and Cox8B. Moreover, the research observed a dose-dependent reduction in the mRNA expression of adipogenic genes (C/EBPα, GLUT-4, SREBP-1C, and FASN) with increasing concentrations of OFFE. Additionally, OFFE activated the AMPK signaling pathway to inhibit adipogenesis. These findings elucidate that OFFE has an inhibitory effect on adipogenesis and promotes browning in 3T3-L1 adipocytes, which lays the foundation for further investigation of the lipid-lowering mechanism of OFFE in vivo in the future.
Collapse
Affiliation(s)
- Zhiying Yang
- College of Life Science, Nanjing Forestry University, Nanjing 210037, China; (Z.Y.); (Y.L.); (J.Y.); (S.Y.); (Y.D.)
| | - Yuxin Lu
- College of Life Science, Nanjing Forestry University, Nanjing 210037, China; (Z.Y.); (Y.L.); (J.Y.); (S.Y.); (Y.D.)
| | - Tingting Li
- Department of Food Science and Technology, College of Light Industry and Food Engineer, Nanjing Forestry University, Nanjing 210037, China;
| | - Xunyong Zhou
- HC Enzyme (Shenzhen) Biotech. Co., Ltd., Shenzhen 518112, China;
| | - Jia Yang
- College of Life Science, Nanjing Forestry University, Nanjing 210037, China; (Z.Y.); (Y.L.); (J.Y.); (S.Y.); (Y.D.)
| | - Shuwen Yang
- College of Life Science, Nanjing Forestry University, Nanjing 210037, China; (Z.Y.); (Y.L.); (J.Y.); (S.Y.); (Y.D.)
| | - Su Bu
- College of Life Science, Nanjing Forestry University, Nanjing 210037, China; (Z.Y.); (Y.L.); (J.Y.); (S.Y.); (Y.D.)
| | - Yifan Duan
- College of Life Science, Nanjing Forestry University, Nanjing 210037, China; (Z.Y.); (Y.L.); (J.Y.); (S.Y.); (Y.D.)
- International Cultivar Registration Center for Osmanthus, Nanjing 210037, China
| |
Collapse
|
10
|
Lee Y, Iqbal N, Lee MH, Park DS, Kim YS. Anti-Obesity Effect of Lactobacillus acidophilus DS0079 (YBS1) by Inhibition of Adipocyte Differentiation through Regulation of p38 MAPK/PPARγ Signaling. J Microbiol Biotechnol 2024; 34:1073-1081. [PMID: 38719777 PMCID: PMC11180917 DOI: 10.4014/jmb.2402.02012] [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: 02/07/2024] [Revised: 03/13/2024] [Accepted: 03/18/2024] [Indexed: 05/29/2024]
Abstract
Obesity is spawned by an inequality between the portion of energy consumed and the quantity of energy expended. Disease entities such as cardiovascular disease, arteriosclerosis, hypertension, and cancer, which are correlated with obesity, influence society and the economy. Suppression of adipogenesis, the process of white adipocyte generation, remains a promising approach for treating obesity. Oil Red O staining was used to differentiate 3T3-L1 cells for screening 20 distinct Lactobacillus species. Among these, Lactobacillus acidophilus DS0079, referred to as YBS1, was selected for further study. YBS1 therapy decreased 3T3-L1 cell development. Triglyceride accumulation and mRNA expression of the primary adipogenic marker, peroxisome proliferator-activated receptor gamma (PPARγ), including its downstream target genes, adipocyte fatty acid binding protein 4 and adiponectin, were almost eliminated. YBS1 inhibited adipocyte differentiation at the early stage (days 0-2), but no significant difference was noted between the mid-stage (days 2-4) and late-stage (days 4-6) development. YBS1 stimulated the activation of p38 mitogen-activated protein kinase (p38 MAPK) during the early stages of adipogenesis; however, this effect was eliminated by the SB203580 inhibitor. The data showed that YBS1 administration inhibited the initial development of adipocytes via stimulation of the p38 MAPK signaling pathway, which in turn controlled PPARγ expression. In summary, YBS1 has potential efficacy as an anti-obesity supplement and requires further exploration.
Collapse
Affiliation(s)
- Youri Lee
- Department of Microbiology, College of Medicine, Soonchunhyang University, Cheonan 31151, Republic of Korea
- Institute of Tissue Regeneration, College of Medicine, Soonchunhyang University, Cheonan 31151, Republic of Korea
| | - Navid Iqbal
- Department of Microbiology, College of Medicine, Soonchunhyang University, Cheonan 31151, Republic of Korea
- Institute of Tissue Regeneration, College of Medicine, Soonchunhyang University, Cheonan 31151, Republic of Korea
| | - Mi-Hwa Lee
- Nakdonggang National Institute of Biological Resources, Sangju 37242, Republic of Korea
| | - Doo-Sang Park
- Biological Resource Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup 56212, Republic of Korea
| | - Yong-Sik Kim
- Department of Microbiology, College of Medicine, Soonchunhyang University, Cheonan 31151, Republic of Korea
- Institute of Tissue Regeneration, College of Medicine, Soonchunhyang University, Cheonan 31151, Republic of Korea
| |
Collapse
|
11
|
Abebe BK, Wang H, Li A, Zan L. A review of the role of transcription factors in regulating adipogenesis and lipogenesis in beef cattle. J Anim Breed Genet 2024; 141:235-256. [PMID: 38146089 DOI: 10.1111/jbg.12841] [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: 09/30/2023] [Revised: 11/25/2023] [Accepted: 11/30/2023] [Indexed: 12/27/2023]
Abstract
In the past few decades, genomic selection and other refined strategies have been used to increase the growth rate and lean meat production of beef cattle. Nevertheless, the fast growth rates of cattle breeds are often accompanied by a reduction in intramuscular fat (IMF) deposition, impairing meat quality. Transcription factors play vital roles in regulating adipogenesis and lipogenesis in beef cattle. Meanwhile, understanding the role of transcription factors in regulating adipogenesis and lipogenesis in beef cattle has gained significant attention to increase IMF deposition and meat quality. Therefore, the aim of this paper was to provide a comprehensive summary and valuable insight into the complex role of transcription factors in adipogenesis and lipogenesis in beef cattle. This review summarizes the contemporary studies in transcription factors in adipogenesis and lipogenesis, genome-wide analysis of transcription factors, epigenetic regulation of transcription factors, nutritional regulation of transcription factors, metabolic signalling pathways, functional genomics methods, transcriptomic profiling of adipose tissues, transcription factors and meat quality and comparative genomics with other livestock species. In conclusion, transcription factors play a crucial role in promoting adipocyte development and fatty acid biosynthesis in beef cattle. They control adipose tissue formation and metabolism, thereby improving meat quality and maintaining metabolic balance. Understanding the processes by which these transcription factors regulate adipose tissue deposition and lipid metabolism will simplify the development of marbling or IMF composition in beef cattle.
Collapse
Affiliation(s)
- Belete Kuraz Abebe
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, People's Republic of China
- Department of Animal Science, Werabe University, Werabe, Ethiopia
| | - Hongbao Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, People's Republic of China
| | - Anning Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, People's Republic of China
| | - Linsen Zan
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, People's Republic of China
| |
Collapse
|
12
|
Wang Q, Hu GL, Qiu MH, Cao J, Xiong WY. Coffee, tea, and cocoa in obesity prevention: Mechanisms of action and future prospects. Curr Res Food Sci 2024; 8:100741. [PMID: 38694556 PMCID: PMC11061710 DOI: 10.1016/j.crfs.2024.100741] [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: 01/13/2024] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 05/04/2024] Open
Abstract
Obesity, a major public health problem, causes numerous complications that threaten human health and increase the socioeconomic burden. The pathophysiology of obesity is primarily attributed to lipid metabolism disorders. Conventional anti-obesity medications have a high abuse potential and frequently deliver insufficient efficacy and have negative side-effects. Hence, functional foods are regarded as effective alternatives to address obesity. Coffee, tea, and cocoa, three widely consumed beverages, have long been considered to have the potential to prevent obesity, and several studies have focused on their intrinsic molecular mechanisms in past few years. Therefore, in this review, we discuss the mechanisms by which the bioactive ingredients in these three beverages counteract obesity from the aspects of adipogenesis, lipolysis, and energy expenditure (thermogenesis). The future prospects and challenges for coffee, tea, and cocoa as functional products for the treatment of obesity are also discussed, which can be pursued for future drug development and prevention strategies against obesity.
Collapse
Affiliation(s)
- Qian Wang
- Key Laboratory of Medicinal Chemistry for Natural Resource (Ministry of Education), Yunnan Provincial Center for Research & Development of Natural Products, School of Pharmacy, Yunnan University, Kunming, 650500, China
| | - Gui-Lin Hu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China
| | - Ming-Hua Qiu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China
| | - Jun Cao
- Key Laboratory for Transboundary Ecosecurity of Southwest China (Ministry of Education), Yunnan Key Laboratory of Plant Reproductive Adaptation and Evolutionary Ecology, Institute of Biodiversity, School of Ecology and Environmental Science, Yunnan University, Kunming, 650504, Yunnan, China
| | - Wen-Yong Xiong
- Key Laboratory of Medicinal Chemistry for Natural Resource (Ministry of Education), Yunnan Provincial Center for Research & Development of Natural Products, School of Pharmacy, Yunnan University, Kunming, 650500, China
| |
Collapse
|
13
|
Choi YR, Kim YS, Kim MJ. Cinnamyl Alcohol Attenuates Adipogenesis in 3T3-L1 Cells by Arresting the Cell Cycle. Int J Mol Sci 2024; 25:693. [PMID: 38255766 PMCID: PMC10815721 DOI: 10.3390/ijms25020693] [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: 12/18/2023] [Revised: 01/01/2024] [Accepted: 01/03/2024] [Indexed: 01/24/2024] Open
Abstract
Cinnamyl alcohol (CA) is an aromatic compound found in several plant-based resources and has been shown to exert anti-inflammatory and anti-microbial activities. However, the anti-adipogenic mechanism of CA has not been sufficiently studied. The present study aimed to investigate the effect and mechanism of CA on the regulation of adipogenesis. As evidenced by Oil Red O staining, Western blotting, and real-time PCR (RT-PCR) analyses, CA treatment (6.25-25 μM) for 8 d significantly inhibited lipid accumulation in a concentration-dependent manner and downregulated adipogenesis-related markers (peroxisome proliferator-activated receptor γ (PPARγ), CCAAT/enhancer-binding protein α (C/EBPα), fatty acid binding protein 4 (FABP4), adiponectin, fatty acid synthase (FAS)) in 3-isobutyl-1-methylxanthine, dexamethasone, and insulin(MDI)-treated 3T3-L1 adipocytes. In particular, among the various differentiation stages, the early stage of adipogenesis was critical for the inhibitory effect of CA. Cell cycle analysis using flow cytometry and Western blotting showed that CA effectively inhibited MDI-induced initiation of mitotic clonal expansion (MCE) by arresting the cell cycle in the G0/G1 phase and downregulating the expression of C/EBPβ, C/EBPδ, and cell cycle markers (cyclin D1, cyclin-dependent kinase 6 (CDK6), cyclin E1, CDK2, and cyclin B1). Moreover, AMP-activated protein kinase α (AMPKα), acetyl-CoA carboxylase (ACC), and extracellular signal-regulated kinase 1/2 (ERK1/2), markers of upstream signaling pathways, were phosphorylated during MCE by CA. In conclusion, CA can act as an anti-adipogenic agent by inhibiting the AMPKα and ERK1/2 signaling pathways and the cell cycle and may also act as a potential therapeutic agent for obesity.
Collapse
Affiliation(s)
- Yae Rim Choi
- Division of Food Functionality Research, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea;
- Department of Food Science and Biotechnology, Ewha Womans University, Seoul 03760, Republic of Korea;
| | - Young-Suk Kim
- Department of Food Science and Biotechnology, Ewha Womans University, Seoul 03760, Republic of Korea;
| | - Min Jung Kim
- Division of Food Functionality Research, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea;
| |
Collapse
|
14
|
Shrestha D, Kim E, Shrestha KK, Suh SS, Kim SH, Seo JB. Methanol extract of Elsholtzia fruticosa promotes 3T3-L1 preadipocyte differentiation. JOURNAL OF ANIMAL SCIENCE AND TECHNOLOGY 2024; 66:204-218. [PMID: 38618027 PMCID: PMC11007459 DOI: 10.5187/jast.2024.e6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 04/16/2024]
Abstract
Elsholtzia fruticosa (EF) is present in tropical regions throughout South Asian countries as well as the Himalayas. Although it has been used as a traditional medicine to treat digestive, respiratory, and inflammatory issues, its effect on preadipocyte differentiation is unknown. In this study, we examined the effects of a methanol extract prepared from EF on the differentiation of 3T3-L1 preadipocytes. Cell differentiation was assessed by microscopic observation and oil-red O staining. The expression of adipogenic and lipogenic genes, including PPARγ and C/EBPα, was measured by western blot analysis and quantitative real-time polymerase chain reaction (qRT-PCR), to provide insight into adipogenesis and lipogenesis mechanisms. The results indicated that EF promotes the differentiation of 3T3-L1 preadipocytes, with elevated lipid accumulation occurring in a concentration-dependent manner without apparent cytotoxicity. EF enhances the expression of adipogenic and lipogenic genes, including PPARγ, FABP4, adiponectin, and FAS, at the mRNA and protein levels. The effect of EF was more pronounced during the early and middle stages of 3T3-L1 cell differentiation. Treatment with EF decreased C/EBP homologous protein (CHOP) mRNA and protein levels, while increasing C/EBPα and PPARγ expression. Treatment with EF resulted in the upregulation of cyclin E and CDK2 gene expression within 24 h, followed by a decrease at 48 h, demonstrating the early-stage impact of EF. A concomitant increase in cyclin-D1 levels was observed compared with untreated cells, indicating that EF modulates lipogenic and adipogenic genes through intricate mechanisms involving CHOP and cell cycle pathways. In summary, EF induces the differentiation of 3T3-L1 preadipocytes by increasing the expression of adipogenic and lipogenic genes, possibly through CHOP and cell cycle-dependent mechanisms.
Collapse
Affiliation(s)
- Deumaya Shrestha
- Department of Biosciences, Mokpo National
University, Jeonnam 58554, Korea
| | - Eunbin Kim
- Department of Biomedicine, Health &
Life Convergence Sciences, BK21 Four, Biomedical and Healthcare Research
Institute, Mokpo National University, Jeonnam 58554,
Korea
| | | | - Sung-Suk Suh
- Department of Biosciences, Mokpo National
University, Jeonnam 58554, Korea
- Department of Biomedicine, Health &
Life Convergence Sciences, BK21 Four, Biomedical and Healthcare Research
Institute, Mokpo National University, Jeonnam 58554,
Korea
| | - Sung-Hak Kim
- Department of Animal Science, Chonnam
National University, Gwangju 61186, Korea
| | - Jong Bae Seo
- Department of Biosciences, Mokpo National
University, Jeonnam 58554, Korea
- Department of Biomedicine, Health &
Life Convergence Sciences, BK21 Four, Biomedical and Healthcare Research
Institute, Mokpo National University, Jeonnam 58554,
Korea
| |
Collapse
|
15
|
Li Y, Jiang B, Zeng L, Tang Y, Qi X, Wan Z, Feng W, Xie L, He R, Zhu H, Wu Y. Adipocyte-derived exosomes promote the progression of triple-negative breast cancer through circCRIM1-dependent OGA activation. ENVIRONMENTAL RESEARCH 2023; 239:117266. [PMID: 37775001 DOI: 10.1016/j.envres.2023.117266] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/13/2023] [Accepted: 09/27/2023] [Indexed: 10/01/2023]
Abstract
Triple-negative breast cancer (TNBC) has an escalating morbidity and a dismal prognosis. Obesity has been reported to be strongly linked to adverse TNBC outcomes. Exosomes (Exos) transport RNA and proteins between cells and serve as intermediaries for cell-to-cell communication. Accumulated evidence suggests that adipose-secreted circular RNAs (circRNAs) can modulate protein glycosylation in TNBC to facilitate tumor cell outgrowth. Herein, exo-circCRIM1 expression was found to be elevated in TNBC patients with a high body fat percentage. Functional experiments demonstrated that by inhibiting miR-503-5p, exo-circCRIM1 enhanced TNBC evolution and metastasis while activating glycosylation hydrolase OGA. Furthermore, OGA negatively regulates FBP1 by decreasing its protein stability. Moreover, the levels of OGA and FBP1 were positively related to the infiltration level of some immune cells in TNBC. These findings indicate that exo-cirCRIM1 secreted by adipocytes contributes to TNBC progression by inhibiting miR-503-5p and activating the OGA/FBP1 signaling pathway. The findings reveal a novel intercellular signaling pathway mediated by adipose-derived exosomes and suggest that treatment targeting the secreted exosome-circCRIM1 may reverse TNBC progression.
Collapse
Affiliation(s)
- Yuehua Li
- Institute of Pathogenic Biology, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China; Department of Medical Oncology, The First Affiliated Hospital of University of South China, Hengyang, Hunan, 421001, China
| | - Baohong Jiang
- Department of Pharmacy, The First Affiliated Hospital of University of South China, Hengyang, Hunan, 421001, China
| | - Lijun Zeng
- Department of Medical Oncology, The First Affiliated Hospital of University of South China, Hengyang, Hunan, 421001, China
| | - Yuanbin Tang
- Department of Medical Oncology, The First Affiliated Hospital of University of South China, Hengyang, Hunan, 421001, China
| | - Xiaowen Qi
- Department of Medical Oncology, The First Affiliated Hospital of University of South China, Hengyang, Hunan, 421001, China
| | - Zhixing Wan
- Department of Medical Oncology, The First Affiliated Hospital of University of South China, Hengyang, Hunan, 421001, China
| | - Wenjie Feng
- Department of Medical Oncology, The First Affiliated Hospital of University of South China, Hengyang, Hunan, 421001, China
| | - Liming Xie
- Department of Medical Oncology, The First Affiliated Hospital of University of South China, Hengyang, Hunan, 421001, China
| | - Rongfang He
- Department of Pathology, The First Affiliated Hospital of University of South China, Hengyang, Hunan, 421001, China
| | - Hongbo Zhu
- Department of Medical Oncology, The First Affiliated Hospital of University of South China, Hengyang, Hunan, 421001, China.
| | - Yimou Wu
- Institute of Pathogenic Biology, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
16
|
Kim N, Lee S, Jung EJ, Jung EY, Chang UJ, Jin CM, Suh HJ, Choi HS. Yeast-Hydrolysate-Derived 1-Methyl-1,2,3,4-tetrahydro-β-carboline-3-carboxylic Acid Inhibits Fat Accumulation during Adipocyte Differentiation. Foods 2023; 12:3466. [PMID: 37761175 PMCID: PMC10528377 DOI: 10.3390/foods12183466] [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: 08/30/2023] [Revised: 09/14/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
This study aimed to investigate the impact of yeast hydrolysate (YH) on lipogenesis, elucidate its mechanistic action, and identify the active compounds responsible for its anti-adipogenic effects. YH (2 mg/mL) significantly reduced Oil Red O-stained lipids. YH (2 mg/mL) also downregulated C/EBPβ and upregulated KLF2, both of which are early adipogenic factors. Moreover, YH (2 mg/mL) decreased C/EBPα, PPARγ, FABP4, FAS, ACC, and HMGCR mRNA expression. Additionally, YH significantly downregulated SEBP1c and SREBP2 and their target genes, which govern fatty acid and cholesterol metabolism; however, 2 mg/mL YH had a greater suppressive effect on SREBP1c than on SREBP2. YH (2 mg/mL) also significantly reduced the mRNA level of G6PD and malic enzyme, which are enzymes that synthesize NADPH for lipid synthesis, compared with the control. Furthermore, 1-methyl-1,2,3,4-tetrahydro-β-carboline-3-carboxylic acid (MTCA) was identified as the active compound with anti-adipogenic effects using solvent fractionation and chromatographic analysis of YH, and 1.1 μg/mL MTCA significantly downregulated SREBP1c/SREBP2 mRNAs by 47.8% and 69.2%, respectively, along with the target genes FAS, ACC, and HMGCR by 79.0%, 77.0%, and 40.9%, respectively. Collectively, YH effectively suppressed adipogenic lipid storage by downregulating SREBP- and NADPH-synthesizing genes. These findings suggest that YH containing MTCA has the potential to act as an anti-obesity agent.
Collapse
Affiliation(s)
- Nari Kim
- Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul 02841, Republic of Korea; (N.K.); (S.L.)
| | - Sekyung Lee
- Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul 02841, Republic of Korea; (N.K.); (S.L.)
- Transdisciplinary Major in Learning Health Systems, Department of Healthcare Sciences, Graduate School, Korea University, Seoul 02841, Republic of Korea
| | - Eun-Jin Jung
- Department of Food and Biotechnology, Korea University, Sejong 30019, Republic of Korea;
| | - Eun Young Jung
- Department of Home Economic Education, Jeonju University, Jeonju 55069, Republic of Korea;
| | - Un-Jae Chang
- Department of Food and Nutrition, Dongduk Women’s University, Seoul 02748, Republic of Korea;
| | - Cheng-Min Jin
- Analysis and Research Department, NeuroVIS, Inc., Hwaseong-si 18469, Republic of Korea;
| | - Hyung Joo Suh
- Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul 02841, Republic of Korea; (N.K.); (S.L.)
- Transdisciplinary Major in Learning Health Systems, Department of Healthcare Sciences, Graduate School, Korea University, Seoul 02841, Republic of Korea
| | - Hyeon-Son Choi
- Department of Food Nutrition, Sangmyung University, Hongjimun 2-Gil 20, Jongno-Gu, Seoul 03016, Republic of Korea
| |
Collapse
|
17
|
Anantha P, Liu Z, Raj P, Barman I. Optical diffraction tomography and Raman spectroscopy reveal distinct cellular phenotypes during white and brown adipocyte differentiation. Biosens Bioelectron 2023; 235:115388. [PMID: 37207582 PMCID: PMC10626559 DOI: 10.1016/j.bios.2023.115388] [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: 04/15/2023] [Revised: 05/06/2023] [Accepted: 05/10/2023] [Indexed: 05/21/2023]
Abstract
White adipose tissue (WAT) and brown adipose tissue (BAT) are the primary types of fats in humans, and they play prominent roles in energy storage and thermogenesis, respectively. While the mechanisms of terminal adipogenesis are well understood, much remains unknown about the early stages of adipogenic differentiation. Label-free approaches, such as optical diffraction tomography (ODT) and Raman spectroscopy, offer the ability to retrieve morphological and molecular information at the single cell level without the negative effects of photobleaching and system perturbation due to introduction of fluorophores. In this study, we employed 3D ODT and Raman spectroscopy to gain deeper insights into the early stages of differentiation of human white preadipocytes (HWPs) and human brown preadipocytes (HBPs). We utilized ODT to retrieve morphological information, including cell dry mass and lipid mass, and Raman spectroscopy to obtain molecular information about lipids. Our findings reveal that HWPs and HBPs undergo dynamic and differential changes during the differentiation process. Notably, we found that HBPs accumulated lipids more rapidly and had a higher lipid mass than HWPs. Additionally, both cell types experienced an increase and subsequent decrease in cell dry mass during the first seven days, followed by an increase after day 7, which we attribute to the transformation of adipogenic precursors in the early stages. Finally, HBPs had higher lipid unsaturation levels than HWPs for the same differentiation timepoints. The insights gained from our study provide crucial contributions towards the advancement of new therapies for obesity and related diseases.
Collapse
Affiliation(s)
- Pooja Anantha
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Zhenhui Liu
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Piyush Raj
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Ishan Barman
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA; Department of Oncology, Johns Hopkins University, Baltimore, MD, 21287, USA; The Russell H. Morgan Department of Radiology and Radiological Science, Division of Cancer Imaging Research, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
18
|
Molonia MS, Salamone FL, Muscarà C, Costa G, Vento G, Saija A, Speciale A, Cimino F. Regulation of mitotic clonal expansion and thermogenic pathway are involved in the antiadipogenic effects of cyanidin-3-O-glucoside. Front Pharmacol 2023; 14:1225586. [PMID: 37614314 PMCID: PMC10442822 DOI: 10.3389/fphar.2023.1225586] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 07/27/2023] [Indexed: 08/25/2023] Open
Abstract
Introduction: Obesity is a metabolic disease with an increase both in cell size (hypertrophy) and in cell number (hyperplasia) following differentiation of new adipocytes. Adipogenesis is a well-orchestrated program in which mitotic clonal expansion (MCE) occurs in the early step followed by the late terminal differentiation one. Methods: Aim of the study was to evaluate the in vitro effects of cyanidin-3-O-glucoside (C3G), an anthocyanin present in many fruits and vegetables, in the early or late phase of 3T3-L1 preadipocytes differentiation. Results: C3G exposure in the early phase of adipogenesis process induced a more marked reduction of CCAAT/enhancer-binding protein-β (C/EBPβ), peroxisome proliferator-activated receptor γ (PPAR-ɣ) and fatty acid synthase (Fasn) expression than late phase exposure and these effects were associated to a reduced MCE with cell cycle arrest at G0/G1 phase via p21 expression. Furthermore, C3G exposure during the early phase activated AMP-activated protein kinase (AMPK) pathway better than in the late phase promoting the enhancement of beige-like adipocytes. In fact, C3G induced thermogenic biomarkers uncoupling protein-1 (Ucp1) and peroxisome proliferator-activated receptor-gamma coactivator-1 alpha (Pgc1) and these effects were more evident during early phase exposure. Conclusion: Our data demonstrate that C3G reduces the terminal adipogenic process affecting the early phase of differentiation and inducing a thermogenic program.
Collapse
Affiliation(s)
- Maria Sofia Molonia
- Department of Chemical Biological Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
- “Prof Antonio Imbesi” Foundation, University of Messina, Messina, Italy
| | - Federica Lina Salamone
- Department of Chemical Biological Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Claudia Muscarà
- Department of Chemical Biological Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Gregorio Costa
- Department of Human and Pediatric Pathology “Gaetano Barresi”, University of Messina, Messina, Italy
| | - Grazia Vento
- Department of Experimental Medicine (DIMES), University of Genova, Genoa, Italy
| | - Antonella Saija
- Department of Chemical Biological Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Antonio Speciale
- Department of Chemical Biological Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Francesco Cimino
- Department of Chemical Biological Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| |
Collapse
|
19
|
Giordano D, Scafuri B, De Masi L, Capasso L, Maresca V, Altucci L, Nebbioso A, Facchiano A, Bontempo P. Sirtuin Inhibitor Cambinol Induces Cell Differentiation and Differently Interferes with SIRT1 and 2 at the Substrate Binding Site. Biomedicines 2023; 11:1624. [PMID: 37371719 DOI: 10.3390/biomedicines11061624] [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: 03/23/2023] [Revised: 05/28/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
Epigenetic mechanisms finely regulate gene expression and represent potential therapeutic targets. Cambinol is a synthetic heterocyclic compound that inhibits class III histone deacetylases known as sirtuins (SIRTs). The acetylating action that results could be crucial in modulating cellular functions via epigenetic regulations. The main aim of this research was to investigate the effects of cambinol, and its underlying mechanisms, on cell differentiation by combining wet experiments with bioinformatics analyses and molecular docking simulations. Our in vitro study evidenced the ability of cambinol to induce the differentiation in MCF-7, NB4, and 3T3-L1 cell lines. Interestingly, focusing on the latter that accumulated cytoplasmic lipid droplets, the first promising results related to the action mechanisms of cambinol have shown the induction of cell cycle-related proteins (such as p16 and p27) and modulation of the expression of Rb protein and nuclear receptors related to cell differentiation. Moreover, we explored the inhibitory mechanism of cambinol on human SIRT1 and 2 performing in silico molecular simulations by protein-ligand docking. Cambinol, unlike from other sirtuin inhibitors, is able to better interact with the substrate binding site of SIRT1 than with the inhibition site. Additionally, for SIRT2, cambinol partially interacts with the substrate binding site, although the inhibition site is preferred. Overall, our findings suggest that cambinol might contribute to the development of an alternative to the existing epigenetic therapies that modulate SIRTs.
Collapse
Affiliation(s)
- Deborah Giordano
- National Research Council (CNR), Institute of Food Science (ISA), Via Roma 64, 83100 Avellino, Italy
| | - Bernardina Scafuri
- Department of Chemistry and Biology "A. Zambelli", University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy
| | - Luigi De Masi
- National Research Council (CNR), Institute of Biosciences and Bioresources (IBBR), Via Università 133, 80055 Portici, Italy
| | - Lucia Capasso
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Via L. De Crecchio 7, 80138 Naples, Italy
| | - Viviana Maresca
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Via L. De Crecchio 7, 80138 Naples, Italy
| | - Lucia Altucci
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Via L. De Crecchio 7, 80138 Naples, Italy
| | - Angela Nebbioso
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Via L. De Crecchio 7, 80138 Naples, Italy
| | - Angelo Facchiano
- National Research Council (CNR), Institute of Food Science (ISA), Via Roma 64, 83100 Avellino, Italy
| | - Paola Bontempo
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Via L. De Crecchio 7, 80138 Naples, Italy
| |
Collapse
|
20
|
Macchi C, Moregola A, Norata GD, Ruscica M. Protocol to evaluate the impact of murine MCT1-deficient CD8 + T cells on adipogenesis. STAR Protoc 2023; 4:102301. [PMID: 37210722 DOI: 10.1016/j.xpro.2023.102301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/03/2023] [Accepted: 04/21/2023] [Indexed: 05/23/2023] Open
Abstract
The infiltration of activated T cells, such as CD8+ effector, in metabolic tissues plays a crucial role for the initiation and propagation of obesity-induced inflammation. Given the pivotal role of lactate transporter monocarboxylate transporter 1 (MCT1) in immune cell activation, we present a protocol for the isolation and activation of CD8+ T lymphocytes selectively lacking MCT1. We describe steps for the induction of adipocyte differentiation, CD8+ T isolation and activation, and adipocyte-CD8+ T cell co-culture. We then detail qPCR analysis on differentiated adipocytes. For complete details on the use and execution of this protocol, please refer to Macchi et al.1.
Collapse
Affiliation(s)
- Chiara Macchi
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti," Università degli Studi di Milano, 20133 Milan, Italy.
| | - Annalisa Moregola
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti," Università degli Studi di Milano, 20133 Milan, Italy.
| | - Giuseppe Danilo Norata
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti," Università degli Studi di Milano, 20133 Milan, Italy; SISA Center for the Study of Atherosclerosis, Bassini Hospital, Via M. Gorki 50, Cinisello Balsamo, 20092 Milan, Italy
| | - Massimiliano Ruscica
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti," Università degli Studi di Milano, 20133 Milan, Italy; Department of Cardio-Thoracic-Vascular Diseases - Foundation IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy.
| |
Collapse
|
21
|
Ye J, Gao C, Liang Y, Hou Z, Shi Y, Wang Y. Characteristic and fate determination of adipose precursors during adipose tissue remodeling. CELL REGENERATION (LONDON, ENGLAND) 2023; 12:13. [PMID: 37138165 PMCID: PMC10156890 DOI: 10.1186/s13619-023-00157-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 12/30/2022] [Indexed: 05/05/2023]
Abstract
Adipose tissues are essential for actively regulating systemic energy balance, glucose homeostasis, immune responses, reproduction, and longevity. Adipocytes maintain dynamic metabolic needs and possess heterogeneity in energy storage and supply. Overexpansion of adipose tissue, especially the visceral type, is a high risk for diabetes and other metabolic diseases. Changes in adipocytes, hypertrophy or hyperplasia, contribute to the remodeling of obese adipose tissues, accompanied by abundant immune cell accumulation, decreased angiogenesis, and aberrant extracellular matrix deposition. The process and mechanism of adipogenesis are well known, however, adipose precursors and their fate decision are only being defined with recent information available to decipher how adipose tissues generate, maintain, and remodel. Here, we discuss the key findings that identify adipose precursors phenotypically, with special emphasis on the intrinsic and extrinsic signals in instructing and regulating the fate of adipose precursors under pathophysiological conditions. We hope that the information in this review lead to novel therapeutic strategies to combat obesity and related metabolic diseases.
Collapse
Affiliation(s)
- Jiayin Ye
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Cheng Gao
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Yong Liang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Zongliu Hou
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, 650000, Yunnan, China
| | - Yufang Shi
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China.
- The Third Affiliated Hospital of Soochow University and State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University, 199 Renai Road, Suzhou, 215123, Jiangsu, China.
| | - Ying Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China.
| |
Collapse
|
22
|
Ahmad B, Friar EP, Vohra MS, Khan N, Serpell CJ, Garrett MD, Loo JSE, Fong IL, Wong EH. Hydroxylated polymethoxyflavones reduce the activity of pancreatic lipase, inhibit adipogenesis and enhance lipolysis in 3T3-L1 mouse embryonic fibroblast cells. Chem Biol Interact 2023; 379:110503. [PMID: 37084996 DOI: 10.1016/j.cbi.2023.110503] [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: 12/08/2022] [Revised: 04/13/2023] [Accepted: 04/18/2023] [Indexed: 04/23/2023]
Abstract
Hydroxylated polymethoxyflavones (HPMFs) have been shown to possess various anti-disease effects, including against obesity. This study investigates the anti-obesity effects of HPMFs in further detail, aiming to gain understanding of their mechanism of action in this context. The current study demonstrates that two HPMFs; 3'-hydroxy-5,7,4',5'-tetramethoxyflavone (3'OH-TetMF) and 4'-hydroxy-5,7,3',5'-tetramethoxyflavone (4'OH-TetMF) possess anti-obesity effects. They both significantly reduced pancreatic lipase activity in a competitive manner as demonstrated by molecular docking and kinetic studies. In cell studies, it was revealed that both of the HPMFs suppress differentiation of 3T3-L1 mouse embryonic fibroblast cells during the early stages of adipogenesis. They also reduced expression of key adipogenic and lipogenic marker genes, namely peroxisome proliferator-activated receptor-gamma (PPAR-γ), CCAAT/enhancer-binding protein α and β (C/EBP α and β), adipocyte binding protein 2 (aP2), fatty acid synthase (FASN), and sterol regulatory element-binding protein 1 (SREBF 1). They also enhanced the expression of cell cycle genes, i.e., cyclin D1 (CCND1) and C-Myc, and reduced cyclin A2 expression. When further investigated, it was also observed that these HPMFs accelerate lipid breakdown (lipolysis) and enhance lipolytic gene expression. Moreover, they also reduced the secretion of proteins (adipokines), including pro-inflammatory cytokines, from mature adipocytes. Taken together, this study concludes that these HPMFs have anti-obesity effects, which are worthy of further investigation.
Collapse
Affiliation(s)
- Bilal Ahmad
- School of Biosciences, Faculty of Health and Medical Sciences Taylor's University Lakeside Campus, No1 Jalan Taylor's, 47500, Subang Jaya, Malaysia
| | - Emily P Friar
- School of Chemistry and Forensic Science, Ingram Building, University of Kent, Canterbury, Kent, CT2 7NH, United Kingdom
| | - Muhammad Sufyan Vohra
- School of Medicine, Faculty of Health and Medical Sciences Taylor's University Lakeside Campus, No1 Jalan Taylor's, 47500, Subang Jaya, Malaysia
| | - Nasar Khan
- R3 Medical Research, 10045 East Dynamite Boulevard Suite 260, Scottsdale, AZ, 85262, United States
| | - Christopher J Serpell
- School of Chemistry and Forensic Science, Ingram Building, University of Kent, Canterbury, Kent, CT2 7NH, United Kingdom.
| | - Michelle D Garrett
- School of Biosciences, Stacey Building, University of Kent, Canterbury, Kent, CT2 7NJ, United Kingdom
| | - Jason Siau Ee Loo
- School of Pharmacy, Faculty of Health and Medical Sciences Taylor's University Lakeside Campus, No1 Jalan Taylor's, 47500, Subang Jaya, Malaysia
| | - Isabel Lim Fong
- Department of Paraclinical Sciences, Faculty of Medicine and Health Sciences, Universiti Malaysia Sarawak (UNIMAS), 94300, Kota Samarahan, Sarawak, Malaysia
| | - Eng Hwa Wong
- School of Medicine, Faculty of Health and Medical Sciences Taylor's University Lakeside Campus, No1 Jalan Taylor's, 47500, Subang Jaya, Malaysia.
| |
Collapse
|
23
|
Moreno-Castellanos N, Cuartas-Gómez E, Vargas-Ceballos O. Collagen microgel to simulate the adipocyte microenvironment for in vitro research on obesity. Integr Biol (Camb) 2023; 15:zyad011. [PMID: 37591513 DOI: 10.1093/intbio/zyad011] [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: 02/07/2023] [Revised: 04/27/2023] [Accepted: 07/19/2023] [Indexed: 08/19/2023]
Abstract
Obesity is linked to adipose tissue dysfunction, a dynamic endocrine organ. Two-dimensional cultures present technical hurdles hampering their ability to follow individual or cell groups for metabolic disease research. Three-dimensional type I collagen microgels with embedded adipocytes have not been thoroughly investigated to evaluate adipogenic maintenance as instrument for studying metabolic disorders. We aimed to develop a novel tunable Col-I microgel simulating the adipocyte microenvironment to maintain differentiated cells with only insulin as in vitro model for obesity research. Adipocytes were cultured and encapsulated in collagen microgels at different concentrations (2, 3 and 4 mg/mL). Collagen microgels at 3 and 4 mg/mL were more stable after 8 days of culture. However, cell viability and metabolic activity were maintained at 2 and 3 mg/mL, respectively. Cell morphology, lipid mobilization and adipogenic gene expression demonstrated the maintenance of adipocyte phenotype in an in vitro microenvironment. We demonstrated the adequate stability and biocompatibility of the collagen microgel at 3 mg/mL. Cell and molecular analysis confirmed that adipocyte phenotype is maintained over time in the absence of adipogenic factors. These findings will help better understand and open new avenues for research on adipocyte metabolism and obesity. Insight box In the context of adipose tissue dysfunction research, new struggles have arisen owing to the difficulty of cellular maintenance in 2D cultures. Herein, we sought a novel approach using a 3D type I collagen-based biomaterial to adipocyte culture with only insulin. This component was tailored as a microgel in different concentrations to support the growth and survival of adipocytes. We demonstrate that adipocyte phenotype is maintained and key adipogenesis regulators and markers are over time. The cumulative results unveil the practical advantage of this microgel platform as an in vitro model to study adipocyte dysfunction and obesity.
Collapse
Affiliation(s)
- Natalia Moreno-Castellanos
- Centro de Cromatografía y Espectrometría de Masas-CROM-MASS research group, Departamento de Ciencias básicas, Escuela de Medicina, Facultad de Salud, Universidad Industrial de Santander, Bucaramanga, Colombia
| | - Elías Cuartas-Gómez
- Centro de Cromatografía y Espectrometría de Masas-CROM-MASS research group, Departamento de Ciencias básicas, Escuela de Medicina, Facultad de Salud, Universidad Industrial de Santander, Bucaramanga, Colombia
| | - Oscar Vargas-Ceballos
- GIMAT research group, Escuela de Ingeniería Metalúrgica y Ciencia de Materiales, Universidad Industrial de Santander, Bucaramanga, Colombia
| |
Collapse
|
24
|
Clemente-Olivo MP, Hernández-Quiles M, Sparrius R, van der Stoel MM, Janssen V, Habibe JJ, van den Burg J, Jongejan A, Alcaraz-Sobrevals P, van Es R, Vos H, Kalkhoven E, de Vries CJM. Early adipogenesis is repressed through the newly identified FHL2-NFAT5 signaling complex. Cell Signal 2023; 104:110587. [PMID: 36610523 DOI: 10.1016/j.cellsig.2023.110587] [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: 10/31/2022] [Revised: 12/25/2022] [Accepted: 01/03/2023] [Indexed: 01/06/2023]
Abstract
The LIM-domain-only protein FHL2 is a modulator of signal transduction and has been shown to direct the differentiation of mesenchymal stem cells towards osteoblast and myocyte phenotypes. We hypothesized that FHL2 may simultaneously interfere with the induction of the adipocyte lineage. Therefore, we investigated the role of FHL2 in adipocyte differentiation. For these studies pre-adipocytes isolated from mouse adipose tissue and the 3T3-L1 (pre)adipocyte cell line were applied. We performed FHL2 gain of function and knockdown experiments followed by extensive RNAseq analyses and phenotypic characterization of the cells by oil-red O (ORO) lipid staining. Through affinity-purification mass spectrometry (AP-MS) novel FHL2 interacting proteins were identified. Here we report that FHL2 is expressed in pre-adipocytes and for accurate adipocyte differentiation, this protein needs to be downregulated during the early stages of adipogenesis. More specifically, constitutive overexpression of FHL2 drastically inhibits adipocyte differentiation in 3T3-L1 cells, which was demonstrated by suppressed activation of the adipogenic gene expression program as shown by RNAseq analyses, and diminished lipid accumulation. Analysis of the protein-protein interactions mediating this repressive activity of FHL2 on adipogenesis revealed the interaction of FHL2 with the Nuclear factor of activated T-cells 5 (NFAT5). NFAT5 is an established inhibitor of adipocyte differentiation and its knockdown rescued the inhibitory effect of FHL2 overexpression on 3T3-L1 differentiation, indicating that these proteins act cooperatively. We present a new regulatory function of FHL2 in early adipocyte differentiation and revealed that FHL2-mediated inhibition of pre-adipocyte differentiation is dependent on its interaction with NFAT5. FHL2 expression increases with aging, which may affect mesenchymal stem cell differentiation, more specifically inhibit adipocyte differentiation.
Collapse
Affiliation(s)
- Maria P Clemente-Olivo
- Amsterdam UMC location University of Amsterdam, Department of Medical Biochemistry, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, and Amsterdam Gastroenterology, Endocrinology and Metabolism, University of Amsterdam, Amsterdam, the Netherlands
| | - Miguel Hernández-Quiles
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Rinske Sparrius
- Amsterdam UMC location University of Amsterdam, Department of Medical Biochemistry, Amsterdam, the Netherlands
| | - Miesje M van der Stoel
- Amsterdam UMC location University of Amsterdam, Department of Medical Biochemistry, Amsterdam, the Netherlands
| | - Vera Janssen
- Amsterdam UMC location University of Amsterdam, Department of Medical Biochemistry, Amsterdam, the Netherlands
| | - Jayron J Habibe
- Amsterdam UMC location University of Amsterdam, Department of Medical Biochemistry, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, and Amsterdam Gastroenterology, Endocrinology and Metabolism, University of Amsterdam, Amsterdam, the Netherlands
| | - Janny van den Burg
- Amsterdam UMC location University of Amsterdam, Department of Medical Biochemistry, Amsterdam, the Netherlands
| | - Aldo Jongejan
- Amsterdam UMC location University of Amsterdam, Department of Bioinformatics, Amsterdam, the Netherlands
| | - Paula Alcaraz-Sobrevals
- Oncode Institute and Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Robert van Es
- Oncode Institute and Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Harmjan Vos
- Oncode Institute and Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Eric Kalkhoven
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Carlie J M de Vries
- Amsterdam UMC location University of Amsterdam, Department of Medical Biochemistry, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, and Amsterdam Gastroenterology, Endocrinology and Metabolism, University of Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
25
|
Elkhawaga SY, Ismail A, Elsakka EGE, Doghish AS, Elkady MA, El-Mahdy HA. miRNAs as cornerstones in adipogenesis and obesity. Life Sci 2023; 315:121382. [PMID: 36639051 DOI: 10.1016/j.lfs.2023.121382] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/06/2023] [Accepted: 01/07/2023] [Indexed: 01/12/2023]
Abstract
In recent decades, obesity has extensively emerged to the level of pandemics. It's significantly associated with serious co-morbidities that could decrease life quality and even life expectancy. Obesity has several determinants, such as age, sex, endocrine, and genetic factors. The miRNAs have emerged as genetic factors affecting obesity. The miRNAs are small noncoding nucleic acids that can modify gene expression and hence, control biological processes. The miRNAs can greatly affect many biological processes in obesity, such as adipogenesis, lipid metabolism, and homeostasis. As a result, the entry of miRNAs in obesity therapeutic approaches has been strongly advised as miRNAs mimics, inhibitors, and stimulators. Hence, this review aims to point out a summarized and updated overview of miRNAs and their roles in obesity and its included processes, such as adipogenesis and lipid metabolism. Besides, we also review recent applications of miRNAs as a treatment approach for obesity.
Collapse
Affiliation(s)
- Samy Y Elkhawaga
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Ahmed Ismail
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Elsayed G E Elsakka
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt.
| | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt.
| | - Mohamed A Elkady
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Hesham A El-Mahdy
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11231, Cairo, Egypt
| |
Collapse
|
26
|
Yu D, Xin L, Qing X, Hao Z, Yong W, Jiangjiang Z, Yaqiu L. Key circRNAs from goat: discovery, integrated regulatory network and their putative roles in the differentiation of intramuscular adipocytes. BMC Genomics 2023; 24:51. [PMID: 36707755 PMCID: PMC9883971 DOI: 10.1186/s12864-023-09141-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 01/17/2023] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND The procession of preadipocytes differentiation into mature adipocytes involves multiple cellular and signal transduction pathways. Recently. a seirces of noncoding RNAs (ncRNAs), including circular RNAs (circRNAs) were proved to play important roles in regulating differentiation of adipocytes. RESULT In this study, we aimed to identificate the potential circRNAs in the early and late stages of goat intramuscular adipocytes differentiation. Using bioinformatics methods to predict their biological functions and map the circRNA-miRNA interaction network. Over 104 million clean reads in goat intramuscular preadipocytes and adipocytes were mapped, of which16 circRNAs were differentially expressed (DE-circRNAs). Furthermore, we used real-time fluorescent quantitative PCR (qRT-PCR) technology to randomly detect the expression levels of 8 circRNAs among the DE-circRNAs, and our result verifies the accuracy of the RNA-seq data. From the Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis of the DE-circRNAs, two circRNAs, circ_0005870 and circ_0000946, were found in Focal adhesion and PI3K-Akt signaling pathway. Then we draw the circRNA-miRNA interaction network and obtained the miRNAs that possibly interact with circ_0005870 and circ_0000946. Using TargetScan, miRTarBase and miR-TCDS online databases, we further obtained the mRNAs that may interact with the miRNAs, and generated the final circRNA-miRNA-mRNA interaction network. Combined with the following GO (Gene Ontology) and KEGG enrichment analysis, we obtained 5 key mRNAs related to adipocyte differentiation in our interaction network, which are FOXO3(forkhead box O3), PPP2CA (protein phosphatase 2 catalytic subunit alpha), EEIF4E (eukaryotic translation initiation factor 4), CDK6 (cyclin dependent kinase 6) and ACVR1 (activin A receptor type 1). CONCLUSIONS By using Illumina HiSeq and online databases, we generated the final circRNA-miRNA-mRNA interaction network that have valuable functions in adipocyte differentiation. Our work serves as a valuable genomic resource for in-depth exploration of the molecular mechanism of ncRNAs interaction network regulating adipocyte differentiation.
Collapse
Affiliation(s)
- Du Yu
- grid.412723.10000 0004 0604 889XKey Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu, China ,grid.412723.10000 0004 0604 889XKey Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation of Sichuan Province, Southwest Minzu University, Chengdu, China ,grid.412723.10000 0004 0604 889XCollege of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu, China
| | - Li Xin
- grid.412723.10000 0004 0604 889XKey Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu, China ,grid.412723.10000 0004 0604 889XKey Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation of Sichuan Province, Southwest Minzu University, Chengdu, China ,grid.412723.10000 0004 0604 889XCollege of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu, China
| | - Xu Qing
- grid.412723.10000 0004 0604 889XKey Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu, China ,grid.412723.10000 0004 0604 889XKey Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation of Sichuan Province, Southwest Minzu University, Chengdu, China ,grid.412723.10000 0004 0604 889XCollege of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu, China
| | - Zhang Hao
- grid.412723.10000 0004 0604 889XKey Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu, China ,grid.412723.10000 0004 0604 889XKey Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation of Sichuan Province, Southwest Minzu University, Chengdu, China ,grid.412723.10000 0004 0604 889XCollege of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu, China
| | - Wang Yong
- grid.412723.10000 0004 0604 889XKey Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu, China ,grid.412723.10000 0004 0604 889XKey Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation of Sichuan Province, Southwest Minzu University, Chengdu, China ,grid.412723.10000 0004 0604 889XCollege of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu, China
| | - Zhu Jiangjiang
- grid.412723.10000 0004 0604 889XKey Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu, China ,grid.412723.10000 0004 0604 889XKey Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation of Sichuan Province, Southwest Minzu University, Chengdu, China
| | - Lin Yaqiu
- grid.412723.10000 0004 0604 889XKey Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Ministry of Education, Southwest Minzu University, Chengdu, China ,grid.412723.10000 0004 0604 889XKey Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Exploitation of Sichuan Province, Southwest Minzu University, Chengdu, China ,grid.412723.10000 0004 0604 889XCollege of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu, China
| |
Collapse
|
27
|
Cruciani S, Delitala AP, Cossu ML, Ventura C, Maioli M. Management of Obesity and Obesity-Related Disorders: From Stem Cells and Epigenetics to Its Treatment. Int J Mol Sci 2023; 24:2310. [PMID: 36768633 PMCID: PMC9916844 DOI: 10.3390/ijms24032310] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/17/2023] [Accepted: 01/20/2023] [Indexed: 01/26/2023] Open
Abstract
Obesity is a complex worldwide disease, characterized by an abnormal or excessive fat accumulation. The onset of this pathology is generally linked to a complex network of interactions among genetic and environmental factors, aging, lifestyle, and diets. During adipogenesis, several regulatory mechanisms and transcription factors are involved. As fat cells grow, adipose tissue becomes increasingly large and dysfunctional, losing its endocrine function, secreting pro-inflammatory cytokines, and recruiting infiltrating macrophages. This long-term low-grade systemic inflammation results in insulin resistance in peripheral tissues. In this review we describe the main mechanisms involved in adipogenesis, from a physiological condition to obesity. Current therapeutic strategies for the management of obesity and the related metabolic syndrome are also reported.
Collapse
Affiliation(s)
- Sara Cruciani
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
- Consorzio Interuniversitario “Istituto Nazionale Biostrutture e Biosistemi” (INBB), Viale delle Medaglie d’Oro 305, 00136 Roma, Italy
| | | | - Maria Laura Cossu
- General Surgery Unit 2 “Clinica Chirurgica” Medical, Surgical and Experimental Sciences Department, University of Sassari, 07100 Sassari, Italy
| | - Carlo Ventura
- National Laboratory of Molecular Biology and Stem Cell Engineering, Eldor Lab, Istituto Nazionale di Biostrutture e Biosistemi (INBB), Via di Corticella 183, 40128 Bologna, Italy
| | - Margherita Maioli
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
- Consorzio Interuniversitario “Istituto Nazionale Biostrutture e Biosistemi” (INBB), Viale delle Medaglie d’Oro 305, 00136 Roma, Italy
- Center for Developmental Biology and Reprogramming (CEDEBIOR), Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy
| |
Collapse
|
28
|
Mlyczyńska E, Bongrani A, Rame C, Węgiel M, Maślanka A, Major P, Zarzycki P, Ducluzeau PH, De Luca A, Bourbao-Tournois C, Froment P, Rak A, Dupont J. Concentration of Polycyclic Aromatic Hydrocarbons (PAHs) in Human Serum and Adipose Tissues and Stimulatory Effect of Naphthalene in Adipogenesis in 3T3-L1 Cells. Int J Mol Sci 2023; 24:ijms24021455. [PMID: 36674971 PMCID: PMC9861916 DOI: 10.3390/ijms24021455] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/03/2023] [Accepted: 01/10/2023] [Indexed: 01/13/2023] Open
Abstract
Polycyclic aromatic hydrocarbons (PAHs) are one of the most prevalent classes of environmental pollutants. Some evidence shows that PAHs could be involved in human obesity. However, little is known about the distribution patterns of PAHs in human adipose tissue (AT) and the role of PAHs on adipogenesis/lipogenesis. The aims of this pilot study were to determine concentrations of 16 PAHs defined as high-priority pollutants in the plasma and adipose tissue of French and Polish bariatric patients, as well as their correlation with body mass index (BMI), plasma and AT adipokines expression levels. We finally investigated the role of naphthalene on cell proliferation, viability, and differentiation in 3T3-L1 preadipocytes. The concentration of most PAHs was similar in the three types of AT and it was significantly higher in AT as compared to plasma, suggesting bioaccumulation. Polish patients had higher PAH levels in AT than French ones. Only the concentration of naphthalene in AT was positively correlated with the BMI and serum or adipose chemerin, adiponectin and resistin expression, in French but not in Polish patients, who had significantly higher BMIs. Moreover, naphthalene exposure increased the cell proliferation of 3T3-L1 preadipocytes and lipogenesis, and increased the expression of genes involved in adipogenesis after cell differentiation. Taken together, PAHs and more particularly naphthalene could be an obesogenic molecule and increase the risk of obesity.
Collapse
Affiliation(s)
- Ewa Mlyczyńska
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Gronostajowa 9 Street, 30-387 Krakow, Poland
| | - Alice Bongrani
- INRAE UMR 85 Physiologie de la Reproduction et des Comportements, 37380 Nouzilly, France
- CNRS UMR 7247 Physiologie de la Reproduction et des Comportements, 37380 Nouzilly, France
- Department of Animal Physiology, Université de Tours, 37041 Tours, France
| | - Christelle Rame
- INRAE UMR 85 Physiologie de la Reproduction et des Comportements, 37380 Nouzilly, France
- CNRS UMR 7247 Physiologie de la Reproduction et des Comportements, 37380 Nouzilly, France
- Department of Animal Physiology, Université de Tours, 37041 Tours, France
| | - Małgorzata Węgiel
- Faculty of Chemical Engineering and Technology, Cracow University of Technology, Warszawska 24 Street, 31-155 Cracow, Poland
| | - Anna Maślanka
- Faculty of Chemical Engineering and Technology, Cracow University of Technology, Warszawska 24 Street, 31-155 Cracow, Poland
| | - Piotr Major
- 2nd Department of General Surgery, Jagiellonian University Medical College, Macieja Jakubowskiego 2 Street, 30-688 Krakow, Poland
| | - Piotr Zarzycki
- 2nd Department of General Surgery, Jagiellonian University Medical College, Macieja Jakubowskiego 2 Street, 30-688 Krakow, Poland
| | - Pierre-Henri Ducluzeau
- INRAE UMR 85 Physiologie de la Reproduction et des Comportements, 37380 Nouzilly, France
- CNRS UMR 7247 Physiologie de la Reproduction et des Comportements, 37380 Nouzilly, France
- Department of Animal Physiology, Université de Tours, 37041 Tours, France
- CHRU of Tours, Department of Endocrinology-Diabetology and Nutrition, 37032 Tours, France
| | - Arnaud De Luca
- CHRU of Tours, Department of Endocrinology-Diabetology and Nutrition, 37032 Tours, France
- Nutrition, Growth and Cancer (N2C) UMR 1069, University of Tours, INSERM, 37032 Tours, France
| | | | - Pascal Froment
- INRAE UMR 85 Physiologie de la Reproduction et des Comportements, 37380 Nouzilly, France
- CNRS UMR 7247 Physiologie de la Reproduction et des Comportements, 37380 Nouzilly, France
- Department of Animal Physiology, Université de Tours, 37041 Tours, France
| | - Agnieszka Rak
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Gronostajowa 9 Street, 30-387 Krakow, Poland
| | - Joëlle Dupont
- INRAE UMR 85 Physiologie de la Reproduction et des Comportements, 37380 Nouzilly, France
- CNRS UMR 7247 Physiologie de la Reproduction et des Comportements, 37380 Nouzilly, France
- Department of Animal Physiology, Université de Tours, 37041 Tours, France
- Correspondence: ; Tel.: +33-2-47-42-77-89; Fax: +33-2-47-42-77-43
| |
Collapse
|
29
|
Baumgartner C, Krššák M, Vila G, Krebs M, Wolf P. Ectopic lipid metabolism in anterior pituitary dysfunction. Front Endocrinol (Lausanne) 2023; 14:1075776. [PMID: 36860364 PMCID: PMC9968795 DOI: 10.3389/fendo.2023.1075776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 02/02/2023] [Indexed: 02/17/2023] Open
Abstract
Over the past decades, adapted lifestyle and dietary habits in industrialized countries have led to a progress of obesity and associated metabolic disorders. Concomitant insulin resistance and derangements in lipid metabolism foster the deposition of excess lipids in organs and tissues with limited capacity of physiologic lipid storage. In organs pivotal for systemic metabolic homeostasis, this ectopic lipid content disturbs metabolic action, thereby promotes the progression of metabolic disease, and inherits a risk for cardiometabolic complications. Pituitary hormone syndromes are commonly associated with metabolic diseases. However, the impact on subcutaneous, visceral, and ectopic fat stores between disorders and their underlying hormonal axes is rather different, and the underlying pathophysiological pathways remain largely unknown. Pituitary disorders might influence ectopic lipid deposition indirectly by modulating lipid metabolism and insulin sensitivity, but also directly by organ specific hormonal effects on energy metabolism. In this review, we aim to I) provide information about the impact of pituitary disorders on ectopic fat stores, II) and to present up-to-date knowledge on potential pathophysiological mechanisms of hormone action in ectopic lipid metabolism.
Collapse
|
30
|
Ernst AM, Bauer H, Bauer HC, Steiner M, Malfertheiner A, Lipp AT. Lipedema Research-Quo Vadis? J Pers Med 2022; 13:98. [PMID: 36675759 PMCID: PMC9860653 DOI: 10.3390/jpm13010098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 12/25/2022] [Accepted: 12/27/2022] [Indexed: 01/03/2023] Open
Abstract
When studying the current literature, one might get the impression that lipedema is a "modern" disease, with increasing incidence and augmenting prevalence throughout Western countries during the last decade. However, a quick look into older textbooks shows that disproportionate accumulation of fat in female bodies has long been known without being recognized as an independent disease. Nevertheless, it was not until 1940 that Allen and Hines described a "syndrome characterized by fat legs and orthostatic edema" in a seminal publication. The mere awareness that people who have lipedema are not just overweight but suffer from a yet poorly defined pathological condition, may be considered a decisive leap forward in the understanding of lipedema. A number of comprehensive publications have since dealt with the clinical presentation of lipedema and have provided the first clues towards the potential pathological mechanisms underlying its initiation and progression. Nevertheless, despite all effort that has been undertaken to unravel lipedema pathology, many questions have remained unanswered. What can be deduced with certainty from all experimental and medical evidence available so far is that lipedema is neither a cosmetic problem nor is it a problem of lifestyle but should be accepted as a serious disease with yet undetermined genetic background, which makes women's lives unbearable from both a physical and psychological point of view. To date, results from clinical inspections have led to the categorization of various types and stages of lipedema, describing how the extremities are affected and evaluating its progression, as demonstrated by skin alterations, adipose tissue volume increase and physical and everyday-behavioral impediments. There is accumulating evidence showing that advanced stages of lipedema are usually accompanied by excessive weight or obesity. Thus, it is not unreasonable to assume that the progression of lipedema is largely driven by weight gain and the pathological alterations associated with it. Similarly, secondary lymphedema is frequently found in lipedema patients at advanced stages. Needless to say, both conditions considerably blur the clinical presentation of lipedema, making diagnosis difficult and scientific research challenging. The present literature review will focus on lipedema research, based on evidence fromex vivo and in vitro data, which has accumulated throughout the last few decades. We will also open the discussion as to whether the currently used categorization of lipedema stages is still sufficient and up-to-date for the accurate description of this enigmatic disease, whose name, strangely enough, does not match its pathologic correlate.
Collapse
Affiliation(s)
- Anna M. Ernst
- Department of Environment & Biodiversity, Paris Lodron University of Salzburg, 5020 Salzburg, Austria
| | - Hannelore Bauer
- Department of Environment & Biodiversity, Paris Lodron University of Salzburg, 5020 Salzburg, Austria
| | - Hans-Christian Bauer
- Department of Environment & Biodiversity, Paris Lodron University of Salzburg, 5020 Salzburg, Austria
- Institute for Tendon and Bone Regeneration, Paracelsus Medical University (PMU), 5020 Salzburg, Austria
| | - Marianne Steiner
- Department of Environment & Biodiversity, Paris Lodron University of Salzburg, 5020 Salzburg, Austria
| | - Anna Malfertheiner
- Department of Plastic Surgery and Hand Surgery, Klinikum rechts der Isar, Technical University of Munich (TUM), 81675 Munich, Germany
| | | |
Collapse
|
31
|
Alternative Methods as Tools for Obesity Research: In Vitro and In Silico Approaches. LIFE (BASEL, SWITZERLAND) 2022; 13:life13010108. [PMID: 36676057 PMCID: PMC9860640 DOI: 10.3390/life13010108] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 12/22/2022] [Accepted: 12/23/2022] [Indexed: 01/04/2023]
Abstract
The study of adipogenesis is essential for understanding and treating obesity, a multifactorial problem related to body fat accumulation that leads to several life-threatening diseases, becoming one of the most critical public health problems worldwide. In this review, we propose to provide the highlights of the adipogenesis study based on in vitro differentiation of human mesenchymal stem cells (hMSCs). We list in silico methods, such as molecular docking for identification of molecular targets, and in vitro approaches, from 2D, more straightforward and applied for screening large libraries of substances, to more representative physiological models, such as 3D and bioprinting models. We also describe the development of physiological models based on microfluidic systems applied to investigate adipogenesis in vitro. We intend to identify the main alternative models for adipogenesis evaluation, contributing to the direction of preclinical research in obesity. Future directions indicate the association of in silico and in vitro techniques to bring a clear picture of alternative methods based on adipogenesis as a tool for obesity research.
Collapse
|
32
|
Bernal K, Touma C, Erradhouani C, Boronat-Belda T, Gaillard L, Al Kassir S, Le Mentec H, Martin-Chouly C, Podechard N, Lagadic-Gossmann D, Langouet S, Brion F, Knoll-Gellida A, Babin PJ, Sovadinova I, Babica P, Andreau K, Barouki R, Vondracek J, Alonso-Magdalena P, Blanc E, Kim MJ, Coumoul X. Combinatorial pathway disruption is a powerful approach to delineate metabolic impacts of endocrine disruptors. FEBS Lett 2022; 596:3107-3123. [PMID: 35957500 DOI: 10.1002/1873-3468.14465] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 07/22/2022] [Accepted: 07/25/2022] [Indexed: 01/14/2023]
Abstract
The prevalence of metabolic diseases, such as obesity, diabetes, metabolic syndrome and chronic liver diseases among others, has been rising for several years. Epidemiology and mechanistic (in vivo, in vitro and in silico) toxicology have recently provided compelling evidence implicating the chemical environment in the pathogenesis of these diseases. In this review, we will describe the biological processes that contribute to the development of metabolic diseases targeted by metabolic disruptors, and will propose an integrated pathophysiological vision of their effects on several organs. With regard to these pathomechanisms, we will discuss the needs, and the stakes of evolving the testing and assessment of endocrine disruptors to improve the prevention and management of metabolic diseases that have become a global epidemic since the end of last century.
Collapse
Affiliation(s)
- Kévin Bernal
- INSERM UMR-S 1124, Paris, France.,Université Paris Cité, France
| | - Charbel Touma
- Inserm, EHESP, Irset (Institut de recherche en santé environnement et travail) - UMR_S 1085, Université Rennes, France
| | - Chedi Erradhouani
- Université Paris Cité, France.,Ecotoxicologie des substances et des milieux, Parc ALATA, INERIS, Verneuil-en-Halatte, France
| | - Talía Boronat-Belda
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universitas Miguel Hernández, Elche, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Lucas Gaillard
- INSERM UMR-S 1124, Paris, France.,Université Paris Cité, France
| | - Sara Al Kassir
- Department of Life and Health Sciences, INSERM U1211, MRGM, University of Bordeaux, Pessac, France
| | - Hélène Le Mentec
- Inserm, EHESP, Irset (Institut de recherche en santé environnement et travail) - UMR_S 1085, Université Rennes, France
| | - Corinne Martin-Chouly
- Inserm, EHESP, Irset (Institut de recherche en santé environnement et travail) - UMR_S 1085, Université Rennes, France
| | - Normand Podechard
- Inserm, EHESP, Irset (Institut de recherche en santé environnement et travail) - UMR_S 1085, Université Rennes, France
| | - Dominique Lagadic-Gossmann
- Inserm, EHESP, Irset (Institut de recherche en santé environnement et travail) - UMR_S 1085, Université Rennes, France
| | - Sophie Langouet
- Inserm, EHESP, Irset (Institut de recherche en santé environnement et travail) - UMR_S 1085, Université Rennes, France
| | - François Brion
- Ecotoxicologie des substances et des milieux, Parc ALATA, INERIS, Verneuil-en-Halatte, France
| | - Anja Knoll-Gellida
- Department of Life and Health Sciences, INSERM U1211, MRGM, University of Bordeaux, Pessac, France
| | - Patrick J Babin
- Department of Life and Health Sciences, INSERM U1211, MRGM, University of Bordeaux, Pessac, France
| | - Iva Sovadinova
- RECETOX, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Pavel Babica
- RECETOX, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Karine Andreau
- INSERM UMR-S 1124, Paris, France.,Université Paris Cité, France
| | - Robert Barouki
- INSERM UMR-S 1124, Paris, France.,Université Paris Cité, France
| | - Jan Vondracek
- Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic
| | - Paloma Alonso-Magdalena
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universitas Miguel Hernández, Elche, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Etienne Blanc
- INSERM UMR-S 1124, Paris, France.,Université Paris Cité, France
| | - Min Ji Kim
- INSERM UMR-S 1124, Paris, France.,Université Sorbonne Paris Nord, Bobigny, France
| | - Xavier Coumoul
- INSERM UMR-S 1124, Paris, France.,Université Paris Cité, France
| |
Collapse
|
33
|
KRAS Affects the Lipid Composition by Regulating Mitochondrial Functions and MAPK Activation in Bovine Mammary Epithelial Cells. Animals (Basel) 2022; 12:ani12223070. [PMID: 36428301 PMCID: PMC9686882 DOI: 10.3390/ani12223070] [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: 07/19/2022] [Revised: 10/30/2022] [Accepted: 10/31/2022] [Indexed: 11/09/2022] Open
Abstract
Kirsten rat sarcoma viral oncogene homolog (KRAS), or guanosine triphosphatase KRAS, is a proto-oncogene that encodes the small guanosine triphosphatase transductor protein. Previous studies have found that KRAS can promote cytokine secretion, cell chemotaxis, and survival. However, its effects on milk fat synthesis in bovine mammary epithelial cells are unclear. In this study, the effects of KRAS inhibition on cell metabolism, autophagy, oxidative stress, endoplasmic reticulum stress, mitochondrial function, and lipid composition as well as the potential mechanisms were detected in an immortalized dairy cow mammary epithelial cell line (MAC-T). The results showed that inhibition of KRAS changed the lipid composition (especially the triglyceride level), mitochondrial functions, autophagy, and endoplasmic reticulum stress in cells. Moreover, KRAS inhibition regulated the levels of the mammalian target of rapamycin and mitogen-activated protein kinase (extracellular regulated protein kinases, c-Jun N-terminal kinases, p38) activation. These results indicated that regulation of KRAS would affect the synthesis and composition of milk fat. These results are also helpful for exploring the synthesis and secretion of milk fat at the molecular level and provide a theoretical basis for improving the percentage of fat in milk and the yield of milk from cows.
Collapse
|
34
|
Thomaz FM, de Jesus Simão J, da Silva VS, Machado MMF, Oyama LM, Ribeiro EB, Alonso Vale MIC, Telles MM. Ginkgo biloba Extract Stimulates Adipogenesis in 3T3-L1 Preadipocytes. Pharmaceuticals (Basel) 2022; 15:ph15101294. [PMID: 36297406 PMCID: PMC9610090 DOI: 10.3390/ph15101294] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/08/2022] [Accepted: 10/12/2022] [Indexed: 11/16/2022] Open
Abstract
Smaller adipocytes are related to the reversal of metabolic disorders, suggesting that molecules that can act in the adipogenesis pathway are of great interest. The objective of this study was to investigate the effect of Ginkgo biloba extract (GbE) in modulating the differentiation in preadipocytes. 3T3-L1 preadipocytes were differentiated for 7 days into adipocytes without (control group) and with GbE at 1.0 mg/mL. Lipid content and gene expression were analyzed on day 7 (D7) by Oil Red O staining and PCR Array Gene Expression. Western blotting analysis of the key adipogenesis markers was evaluated during the differentiation process at days 3 (D3), 5 (D5), and 7 (D7). GbE increased lipid content and raised the gene expression of the main adipogenesis markers. Key proteins of the differentiation process were modulated by GbE, since C/EBPβ levels were decreased, while C/EBPα levels were increased at D7. Regarding the mature adipocytes’ markers, GbE enhanced the levels of both FABP4 at D5, and perilipin at D3 and D5. In summary, the present findings showed that GbE modulated the adipogenesis pathway suggesting that the treatment could accelerate the preadipocyte maturation, stimulating the expression of mature adipocyte proteins earlier than expected.
Collapse
Affiliation(s)
- Fernanda Malanconi Thomaz
- Post-Graduate Program in Chemical Biology, Institute of Environmental Sciences, Chemical and Pharmaceutical, Universidade Federal de São Paulo—UNIFESP, Diadema 09972-270, Brazil
| | - Jussara de Jesus Simão
- Post-Graduate Program in Chemical Biology, Institute of Environmental Sciences, Chemical and Pharmaceutical, Universidade Federal de São Paulo—UNIFESP, Diadema 09972-270, Brazil
| | - Viviane Simões da Silva
- Post-Graduate Program in Chemical Biology, Institute of Environmental Sciences, Chemical and Pharmaceutical, Universidade Federal de São Paulo—UNIFESP, Diadema 09972-270, Brazil
| | - Meira Maria Forcelini Machado
- Post-Graduate Program in Chemical Biology, Institute of Environmental Sciences, Chemical and Pharmaceutical, Universidade Federal de São Paulo—UNIFESP, Diadema 09972-270, Brazil
| | - Lila Missae Oyama
- Discipline of Nutrition Physiology, Department of Physiology, Universidade Federal de São Paulo—UNIFESP, São Paulo 04023-062, Brazil
| | - Eliane Beraldi Ribeiro
- Discipline of Nutrition Physiology, Department of Physiology, Universidade Federal de São Paulo—UNIFESP, São Paulo 04023-062, Brazil
| | - Maria Isabel Cardoso Alonso Vale
- Post-Graduate Program in Chemical Biology, Institute of Environmental Sciences, Chemical and Pharmaceutical, Universidade Federal de São Paulo—UNIFESP, Diadema 09972-270, Brazil
- Correspondence:
| | - Monica Marques Telles
- Post-Graduate Program in Chemical Biology, Institute of Environmental Sciences, Chemical and Pharmaceutical, Universidade Federal de São Paulo—UNIFESP, Diadema 09972-270, Brazil
- Discipline of Nutrition Physiology, Department of Physiology, Universidade Federal de São Paulo—UNIFESP, São Paulo 04023-062, Brazil
| |
Collapse
|
35
|
Lockridge A, Hanover JA. A nexus of lipid and O-Glcnac metabolism in physiology and disease. Front Endocrinol (Lausanne) 2022; 13:943576. [PMID: 36111295 PMCID: PMC9468787 DOI: 10.3389/fendo.2022.943576] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 07/27/2022] [Indexed: 11/13/2022] Open
Abstract
Although traditionally considered a glucose metabolism-associated modification, the O-linked β-N-Acetylglucosamine (O-GlcNAc) regulatory system interacts extensively with lipids and is required to maintain lipid homeostasis. The enzymes of O-GlcNAc cycling have molecular properties consistent with those expected of broad-spectrum environmental sensors. By direct protein-protein interactions and catalytic modification, O-GlcNAc cycling enzymes may provide both acute and long-term adaptation to stress and other environmental stimuli such as nutrient availability. Depending on the cell type, hyperlipidemia potentiates or depresses O-GlcNAc levels, sometimes biphasically, through a diversity of unique mechanisms that target UDP-GlcNAc synthesis and the availability, activity and substrate selectivity of the glycosylation enzymes, O-GlcNAc Transferase (OGT) and O-GlcNAcase (OGA). At the same time, OGT activity in multiple tissues has been implicated in the homeostatic regulation of systemic lipid uptake, storage and release. Hyperlipidemic patterns of O-GlcNAcylation in these cells are consistent with both transient physiological adaptation and feedback uninhibited obesogenic and metabolic dysregulation. In this review, we summarize the numerous interconnections between lipid and O-GlcNAc metabolism. These links provide insights into how the O-GlcNAc regulatory system may contribute to lipid-associated diseases including obesity and metabolic syndrome.
Collapse
Affiliation(s)
- Amber Lockridge
- Laboratory of Cell and Molecular Biology, National Institute for Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - John A. Hanover
- Laboratory of Cell and Molecular Biology, National Institute for Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
36
|
Expression and Role of β3-Adrenergic Receptor during the Differentiation of 3T3-L1 Preadipocytes into Adipocytes. BIOLOGY 2022; 11:biology11050772. [PMID: 35625499 PMCID: PMC9138837 DOI: 10.3390/biology11050772] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 05/16/2022] [Accepted: 05/16/2022] [Indexed: 02/04/2023]
Abstract
Simple Summary The β3-adrenergic receptor (β3-AR) has long been viewed as a potential therapeutic target for dealing with obesity. Although the lipolytic and thermogenic role of β3-AR in brown/beige adipocytes is well defined, the β3-AR’s adipogenic role in white adipocytes remains unclear at present. In this study, we investigated the expression and function of β3-AR in 3T3-L1 murine white preadipocytes. Knockdown of β3-AR led to less lipid accumulation and triglyceride (TG) content as well as less expression and phosphorylation levels of CCAAT/enhancer-binding protein-α (C/EBP-α) and peroxisome proliferator-activated receptor-γ (PPAR-γ) during 3T3-L1 preadipocyte differentiation. These findings reveal that the β3-AR inhibitor or antagonist could be a promising candidate for potential preventive and therapeutics against obesity. Abstract β3-adrenergic receptor (β3-AR) is expressed predominantly in mature white and brown/beige adipocytes. Although the lipolytic and thermogenic role of β3-AR in brown/beige adipocytes is well defined, the adipogenic role of β3-AR in white adipocytes remains unclear at present. In this study, we investigated the expression and function of β3-AR in differentiating 3T3-L1 cells, murine white preadipocytes. Of note, the expression of β3-AR at the protein and mRNA levels was highly induced in a time-dependent manner during 3T3-L1 preadipocyte differentiation. Interestingly, the results of the pharmacological inhibition study demonstrated the roles of p38 MAPK and PKC in the induction of β3-AR expression in differentiating 3T3-L1 cells. Knockdown of β3-AR led to less lipid accumulation and triglyceride (TG) content during 3T3-L1 preadipocyte differentiation with no cytotoxicity. Furthermore, knockdown of β3-AR resulted in a decrease in not only expression levels of CCAAT/enhancer-binding protein-α (C/EBP-α), peroxisome proliferator-activated receptor-γ (PPAR-γ), fatty acid synthase (FASN), perilipin A, and leptin but also phosphorylation levels of signal transducer and activator of transcription-5 (STAT-5) during 3T3-L1 preadipocyte differentiation. In summary, these results demonstrate firstly that β3-AR expression is highly up-regulated in p38 MAPK and PKC-dependent manners, and the up-regulated β3-AR plays a crucial role in lipid accumulation in differentiating 3T3-L1 cells, which is mediated through control of expression and phosphorylation levels of C/EBP-α, PPAR-γ, STAT-5, FASN, and perilipin A.
Collapse
|