1
|
Baskaran R, Chen YJ, Chang CF, Kuo HN, Liang CH, Abomughaid MM, Kumar KJS, Lin WT. Potato protein hydrolysate (PPH902) exerts anti-lipogenesis and lipolysis-promoting effect by inhibiting adipogenesis in 3T3-L1 adipocytes. 3 Biotech 2025; 15:83. [PMID: 40078570 PMCID: PMC11893942 DOI: 10.1007/s13205-025-04238-0] [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: 11/12/2024] [Accepted: 02/11/2025] [Indexed: 03/14/2025] Open
Abstract
Obesity is linked to cardiovascular disease, cerebrovascular disease, diabetes, and dyslipidemia, lowering quality of life, work productivity, and healthcare expenditures. The aim of this present study is to investigate the mechanism of potato protein (PP) post-treatment in regulating lipogenesis and lipolysis in 3T3-L1 adipocytes. 9% PP hydrolysed for 2 h (PPH902) shows high yield and better activity; thus, PPH902 was used in all other experiments. 3T3-L1 preadipocyte cells were used, the cell culture medium were changed every 2 days, IDH was added on Day 0. PPH902 was added on the 8th day, it was left for 72 h and then cells are collected. The relative triglyceride residual (RTR) content was quantified, and the expression of key lipid metabolism-related proteins was analyzed using Western blotting. PPH902 at concentrations of 400 ppm, 800 ppm, and 1600 ppm markedly decreases the RTR content. PPH902, at higher doses, modulates the expression of lipid production-associated transcription factors PPARγ, SREBP-1c, and FAS by activating AMPK, which inhibits lipogenesis and activates phosphorylated HSL to enhance lipolysis, so augmenting lipid metabolism. These findings suggest that PPH902 is an effective anti-lipogenic and lipolysis-promoting agent with potential applications in anti-obesity interventions.
Collapse
Affiliation(s)
- Rathinasamy Baskaran
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, 413305 Taiwan
| | - Yi-Ju Chen
- Department of Surgery, Taichung Veterans General Hospital, Taichung, 40704 Taiwan
| | - Ching-Fang Chang
- Department of Food Science, College of Agriculture and Health, Tunghai University, Taichung, 40704 Taiwan
| | - Hsin-Ning Kuo
- Department of Hospitality Management, College of Agriculture and Health, Tunghai University, Taichung, 407224 Taiwan
| | - Chih-Hung Liang
- Department of Food Science, College of Agriculture and Health, Tunghai University, Taichung, 40704 Taiwan
| | - Mosleh Mohammad Abomughaid
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, 61922 Bisha, Saudi Arabia
| | - K. J. Senthil Kumar
- Bachelor Program of Biotechnology, National Chung Hsing University, Taichung, Taiwan
- Center for General Education, National Chung Hsing University, Taichung, Taiwan
| | - Wan-Teng Lin
- Department of Hospitality Management, College of Agriculture and Health, Tunghai University, Taichung, 407224 Taiwan
| |
Collapse
|
2
|
Bae JY, Hong SS, Kim MJ. Antioxidant activity and anti-adipogenic effect of ethyl acetate fraction of cumin seeds on 3T3-L1 adipocytes differentiation. Food Sci Biotechnol 2025; 34:1699-1711. [PMID: 40151608 PMCID: PMC11936850 DOI: 10.1007/s10068-024-01746-2] [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: 09/26/2024] [Revised: 10/22/2024] [Accepted: 10/31/2024] [Indexed: 03/29/2025] Open
Abstract
Cumin seeds were fractionated with various solvents to evaluate their antioxidant activity and anti-adipogenic effects in 3T3-L1 adipocytes. In vitro antioxidant assays such as DPPH, ABTS cation scavenger, FRAP, ORAC, and TPC, and 3T3-L1 adipocytes differentiation were performed. The results of these assays show that ethyl acetate fraction of cumin seeds (CSEA) had significantly superior antioxidant activity compared to other cumin seeds fractions. Thymol, benzyl alcohol, and 2-methyl-3-phenylpropanal were analyzed as major volatile components in CSEA. Treatment of 50 and 75 μg/mL CSEA inhibited fat accumulation in 3T3-L1 adipocytes significantly (p < 0.05). When 3T3-L1 adipocytes were treated with CSEA at 75 μg/mL, the expressions of genes and proteins related to fat differentiation and accumulation were significantly reduced (p < 0.05). Therefore, CSEA could be used as antioxidants and as a functional ingredient in anti-adipogenic formulations.
Collapse
Affiliation(s)
- Ji-Yun Bae
- Department of Food and Nutrition, College of Health Science, Kangwon National University, Samcheok, Gangwon 25949 Republic of Korea
| | - Sung-Sil Hong
- Department of Nursing, College of Health Science, Kangwon National University, Samcheok, Gangwon 25949 Republic of Korea
| | - Mi-Ja Kim
- Department of Food and Nutrition, College of Health Science, Kangwon National University, Samcheok, Gangwon 25949 Republic of Korea
| |
Collapse
|
3
|
Lu S, Xiong W, Yi J, Liu S, Zhang F. S-phase kinase-associated protein 1 inhibits orbital fibroblasts adipogenesis to improve thyroid-associated ophthalmopathy (TAO). BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025:119937. [PMID: 40139511 DOI: 10.1016/j.bbamcr.2025.119937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 03/29/2025]
Abstract
Thyroid-associated ophthalmopathy (TAO), a localized manifestation of Graves' disease, involves complex autoimmune interactions leading to orbital tissue inflammation and remodeling. The pathophysiology of TAO is marked by significant orbital connective tissue and fat pad expansion, mononuclear cell infiltration, and fibrosis, ultimately affecting eye motility and quality of life. This study explores the role of S-phase kinase-associated protein 1 (SKP1) in the adipogenic differentiation of orbital fibroblasts (OFs), a key process in TAO. Using bioinformatics analysis of gene expression profiles from TAO patients (GSE105149 and GSE58331), SKP1 was identified as a critical regulator of adipogenesis. Experimental validation confirmed that SKP1 expression is significantly downregulated in TAO-derived OFs under adipogenic differentiation for 10 days, correlating with elevated lipid accumulation and increased expression levels of adipogenic markers. Furthermore, downregulation of SKP1 promotes adipogenic differentiation, while upregulation inhibits this process in OFs in vitro and in TAO mice models in vivo. Mechanistically, SKP1 was shown to modulate the PI3K/AKT signaling, with downregulation activating and upregulation inhibiting the pathway, thereby influencing adipogenesis. In summary, SKP1 exerts a crucial regulatory effect on TAO pathogenesis and might act as an underlying therapeutic target for mitigating OFs adipogenesis in TAO.
Collapse
Affiliation(s)
- Shiyao Lu
- Department of Ophthalmology, The Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, People's Republic of China
| | - Wei Xiong
- Department of Ophthalmology, The Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, People's Republic of China
| | - Jinping Yi
- Department of Ophthalmology, The Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, People's Republic of China
| | - Shenghua Liu
- Department of Ophthalmology, The Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, People's Republic of China
| | - Feng Zhang
- Department of Ophthalmology, The Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, People's Republic of China..
| |
Collapse
|
4
|
Zhong Y, Yang S, Li S, Yuan S, Chen X, Long H, Wu H, Guo Y, Wang T. IL-27 alleviates high-fat diet-induced obesity and metabolic disorders by inhibiting adipogenesis via activating HDAC6. Commun Biol 2025; 8:460. [PMID: 40108289 PMCID: PMC11923273 DOI: 10.1038/s42003-025-07918-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 03/11/2025] [Indexed: 03/22/2025] Open
Abstract
Obesity arises from an imbalance between adipogenesis and adipocyte thermogenesis. Interleukin-27 (IL-27), a heterodimer cytokine, is known to promote thermogenesis in brown adipose tissue. However, its role in adipogenesis remains unclear. This study aims to investigate the effects of IL-27 on adipogenesis both in vitro and in vivo, and to elucidate the underlying mechanisms. In vitro, an adipogenic differentiation model of adipose-derived mesenchymal stem cells (ADSCs) demonstrate that IL-27 is non-cytotoxic to ADSCs and inhibits ADSCs adipogenic differentiation. In vivo, using a high-fat diet (HFD)-induced obese mouse model and a targeted adipose tissue-specific IL-27 overexpression adeno-associated viral (AAV) vector, we confirm that IL-27 suppresses adipogenesis, prevents weight gain, and improves glucose and lipid metabolic homeostasis in obese mice. Additionally, the inhibition of adipogenesis by IL-27 is mediated through HDAC6 activation of the TGFβ/Smad3 signaling pathway. Our study suggests that IL-27 is a potential therapeutic target for obesity and metabolic disorders.
Collapse
Affiliation(s)
- Yinsheng Zhong
- Department of Emergency, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518003, PR China
| | - Shujun Yang
- Department of Emergency, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518003, PR China
| | - Shuangmei Li
- Department of Emergency, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518003, PR China
| | - Sijun Yuan
- Department of Emergency, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518003, PR China
| | - Xuxiang Chen
- Department of Emergency, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518003, PR China
| | - Huibao Long
- Department of Emergency, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518003, PR China
| | - Haidong Wu
- Department of Emergency, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518003, PR China
| | - Yajie Guo
- Department of Emergency, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518003, PR China.
| | - Tong Wang
- Department of Emergency, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, 518003, PR China.
| |
Collapse
|
5
|
Jeong DW, Yun JE, Lee KH, Moon GH, Hong KY, Park JW, Fukuda J, Lee YS, Chun YS. Comprehensive understanding of context-specific functions of PHF2 in lipid metabolic tissues. Sci Rep 2025; 15:9074. [PMID: 40097484 PMCID: PMC11914216 DOI: 10.1038/s41598-025-93438-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 03/06/2025] [Indexed: 03/19/2025] Open
Abstract
Adipose tissue and the liver are known to regulate lipid metabolism through the storage, synthesis, and breakdown of lipids. However, the shared molecular factors affecting lipid metabolism in both tissues remain unclear. Plant Homeodomain Finger 2 (PHF2), one of the histone lysine demethylase7 family, serves as an epigenetic regulator in adipose tissue and E3 ubiquitin ligase in liver cancer. This study uses bioinformatics to analyze the role of PHF2 in lipid metabolism, focusing on its functions in adipose tissue and the liver. We utilized cDNA-chip microarrays, public clinical data, and in vitro three-dimensional cell culture models, validating our bioinformatics findings. Consequently, our analyses showed that PHF2 is positively involved in histone demethylase activity and adipogenesis in patients with obesity and moderate liver disease. However, PHF2 suppressed de novo lipogenesis and tumor progression in patients with liver cancer, enhancing immune cell infiltration in liver cancer. Furthermore, it was experimentally confirmed that PHF2 increases lipid accumulation in adipocytes but acts as a tumor suppressor in liver cancer cells. Overall, this study provides a comprehensive overview of PHF2, highlighting its biological importance.
Collapse
Affiliation(s)
- Do-Won Jeong
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Department of Physiology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Jeong-Eun Yun
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Department of Physiology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Kyoung-Hwa Lee
- Songdo Bio-Engineering, Incheon Jaeneung University, Incheon, 21987, Republic of Korea
| | - Geon Ho Moon
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Department of Physiology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Ki Yong Hong
- Department of Plastic and Reconstructive Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Jong-Wan Park
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Junji Fukuda
- Faculty of Engineering, Yokohama National University, Yokohama, 240-8501, Japan
| | - Yong-Seok Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Department of Physiology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Yang-Sook Chun
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
- Department of Physiology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
- Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
| |
Collapse
|
6
|
Jeong AY, Ma EB, Hong SJ, Kim E, Ko S, Huh JY, Kim YM. Kombucha inhibits adipogenesis and promotes lipolytic activity in 3T3-L1 adipocytes. Food Sci Biotechnol 2025; 34:1037-1043. [PMID: 39974855 PMCID: PMC11832843 DOI: 10.1007/s10068-024-01740-8] [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: 08/09/2024] [Revised: 09/24/2024] [Accepted: 10/11/2024] [Indexed: 02/21/2025] Open
Abstract
This research was conducted to investigate the anti-obesity effects of black tea or green tea kombucha (BK, GK) and compared their compositional differences. As a result of kombucha treatment during the adipocyte differentiation process, peroxisome proliferator-activated receptor γ was significantly decreased, and CCAAT/enhancer binding protein α and adipocyte protein 2 showed a tendency to decrease with BK treatment. Oil red O staining results also demonstrated a reduction of lipid accumulation by BK treatment compared to the control. In mature adipocytes, BK significantly upregulated the gene expression of hormone-sensitive lipase and tended to increase the expression of adipose triglyceride lipase and adiponectin. Additionally, as a biomarker of lipolysis, glycerol content also marginally increased with either BK or GK treatment. The differences were observed in tea polyphenol compound and organic acid contents between BK and GK. In conclusion, these results suggest that black tea kombucha may have anti-obesity activity. Supplementary Information The online version contains supplementary material available at 10.1007/s10068-024-01740-8.
Collapse
Affiliation(s)
- Ah-Young Jeong
- Jeollanamdo Agricultural Research and Extension Services, Jeollanamdo, 58213 Republic of Korea
- Department of Integrative Food, Bioscience and Biotechnology, Chonnam National University, Gwangju, 61186 Republic of Korea
| | - Eun-Bi Ma
- College of Pharmacy, Chonnam National University, Gwangju, Republic of Korea
| | - Seong-Jin Hong
- Department of Integrative Food, Bioscience and Biotechnology, Chonnam National University, Gwangju, 61186 Republic of Korea
- Research Institute of Agricultural Science and Technology, Chonnam National University, Gwangju, 61186 Republic of Korea
| | - Eunhye Kim
- Jeollanamdo Agricultural Research and Extension Services, Jeollanamdo, 58213 Republic of Korea
| | - Sugju Ko
- Jeollanamdo Agricultural Research and Extension Services, Jeollanamdo, 58213 Republic of Korea
| | - Joo Young Huh
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
- Department of Global Innovative Drugs, The Graduate School of Chung-Ang University, Seoul, Republic of Korea
| | - Young-Min Kim
- Department of Integrative Food, Bioscience and Biotechnology, Chonnam National University, Gwangju, 61186 Republic of Korea
| |
Collapse
|
7
|
Albrecht FB, Schick AK, Klatt A, Schmidt FF, Nellinger S, Kluger PJ. Exploring Morphological and Molecular Properties of Different Adipose Cell Models: Monolayer, Spheroids, Gellan Gum-Based Hydrogels, and Explants. Macromol Biosci 2025; 25:e2400320. [PMID: 39450850 PMCID: PMC11904394 DOI: 10.1002/mabi.202400320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/04/2024] [Indexed: 10/26/2024]
Abstract
White adipose tissue (WAT) plays a crucial role in energy homeostasis and secretes numerous adipokines with far-reaching effects. WAT is linked to diseases such as diabetes, cardiovascular disease, and cancer. There is a high demand for suitable in vitro models to study diseases and tissue metabolism. Most of these models are covered by 2D-monolayer cultures. This study aims to evaluate the performance of different WAT models to better derive potential applications. The stability of adipocyte characteristics in spheroids and two 3D gellan gum hydrogels with ex situ lobules and 2D-monolayer culture is analyzed. First, the differentiation to achieve adipocyte-like characteristics is determined. Second, to evaluate the maintenance of differentiated ASC-based models, an adipocyte-based model, and explants over 3 weeks, viability, intracellular lipid content, perilipin A expression, adipokine, and gene expression are analyzed. Several advantages are supported using each of the models. Including, but not limited to, the strong differentiation in 2D-monolayers, the self-assembling within spheroids, the long-term stability of the stem cell-containing hydrogels, and the mature phenotype within adipocyte-containing hydrogels and the lobules. This study highlights the advantages of 3D models due to their more in vivo-like behavior and provides an overview of the different adipose cell models.
Collapse
Affiliation(s)
- Franziska B Albrecht
- Reutlingen Research Institute, Reutlingen University, Alteburgstraße 150, 72762, Reutlingen, Germany
- Faculty of Natural Science, University of Hohenheim, Schloss Hohenheim 1, 70599, Stuttgart, Germany
| | - Ann-Kathrin Schick
- Faculty of Science, Energy and Building Services, Esslingen University, Kanalstraße 33, 73728, Esslingen, Germany
| | - Annemarie Klatt
- Reutlingen Research Institute, Reutlingen University, Alteburgstraße 150, 72762, Reutlingen, Germany
| | - Freia F Schmidt
- Reutlingen Research Institute, Reutlingen University, Alteburgstraße 150, 72762, Reutlingen, Germany
| | - Svenja Nellinger
- Reutlingen Research Institute, Reutlingen University, Alteburgstraße 150, 72762, Reutlingen, Germany
| | - Petra J Kluger
- School of Life Sciences, Reutlingen University, Alteburgstraße 150, 72762, Reutlingen, Germany
| |
Collapse
|
8
|
Kim HJ, Kang SU, Kim HJ, Lee YS, Kim CH. GDF15 inhibits early-stage adipocyte differentiation by enhancing HOP2 expression and suppressing C/EBPα expression. Mol Cell Endocrinol 2025; 598:112461. [PMID: 39814165 DOI: 10.1016/j.mce.2025.112461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 01/02/2025] [Accepted: 01/11/2025] [Indexed: 01/18/2025]
Abstract
Excessive adipocyte differentiation and accumulation contribute to the development of metabolic disorders. Growth differentiation factor 15 (GDF15) plays an essential role in energy homeostasis and is considered an anti-obesity factor; however, elevated serum levels of endogenous GDF15 have been reported in certain individuals with obesity. In this study, to gain a better understanding of this complex relationship between GDF15 levels and obesity, we investigated GDF15 expression and function during adipogenesis. Compared with mice fed a normal diet, those fed a short-term high-fat diet exhibited a reduction in epididymal white adipose tissue and serum GDF15 expression. These results were confirmed in human adipose-derived stem cells that showed reduced GDF15 expression during adipogenesis differentiation. During adipogenesis, GDF15 was primarily degraded via the autophagy lysosomal pathway, and GDF15 overexpression in pre-adipocytes inhibited adipogenesis by suppressing CCAAT enhancer binding protein alpha (C/EBPα). Furthermore, whereas we detected a reduction in homologous-pairing protein 2 (HOP2) expression during adipogenesis, expression increased in response to an overexpression of GDF15. Furthermore, following knockdown of HOP2 during GDF15 overexpression, there was no suppression of C/EBPα expression. These findings indicate that GDF15 undergoes lysosomal degradation via an autophagic pathway and suppresses adipocyte differentiation via the HOP2-mediated inhibition of C/EBPα expression. Collectively, our findings indicate that GDF15 could serve as a potential therapeutic target for the treatment of metabolic disorders.
Collapse
Affiliation(s)
- Haeng Jun Kim
- Department of Otolaryngology, School of Medicine, Ajou University, Suwon, Republic of Korea; Department of Molecular Science and Technology, Ajou University, Suwon, Republic of Korea
| | - Sung-Un Kang
- Department of Otolaryngology, School of Medicine, Ajou University, Suwon, Republic of Korea
| | - Hyo Jeong Kim
- Department of Otolaryngology, School of Medicine, Ajou University, Suwon, Republic of Korea
| | - Yun Sang Lee
- Department of Otolaryngology, School of Medicine, Ajou University, Suwon, Republic of Korea
| | - Chul-Ho Kim
- Department of Otolaryngology, School of Medicine, Ajou University, Suwon, Republic of Korea.
| |
Collapse
|
9
|
Sol CM, Delgado G, Kannan K, Jaddoe VWV, Trasande L, Santos S. Fetal exposure to phthalates and body mass index from infancy to adolescence. The Generation R study. ENVIRONMENTAL RESEARCH 2025; 274:121253. [PMID: 40023387 DOI: 10.1016/j.envres.2025.121253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 02/07/2025] [Accepted: 02/26/2025] [Indexed: 03/04/2025]
Abstract
Prenatal exposure to phthalates might influence the development of childhood obesity. Most previous studies used body mass index (BMI) at a specific age instead of BMI development, which might be a better indicator of later health. We aimed to assess the association of prenatal phthalate exposure with longitudinal BMI development from infancy to adolescence. Among 1,379 mother-child pairs from a population-based cohort study, phthalate concentrations were measured in maternal spot urine samples, collected during first, second and third trimester. We estimated age- and sex-adjusted BMI standard deviation scores (SDS) at 6 months and 1, 2, 3, 4, 6, 10 and 13 years. We examined the associations of maternal phthalate urine concentrations during pregnancy with repeated measures of BMI using linear mixed effects models. An interquartile range higher natural log-transformed maternal first trimester high-molecular weight phthalate and di-2-ethylhexylphthalate (DEHP) urine concentrations were associated with a -0.10 (95% confidence interval (CI) -0.15 to -0.04), and -0.09 (95% CI -0.15 to -0.04) lower age- and sex-adjusted BMI at 6 months. An interquartile range higher natural log-transformed maternal first trimester phthalic acid and low-molecular weight phthalate urine concentrations were associated with a 0.11 (95% CI 0.03 to 0.18) and 0.13 (95% CI 0.04 to 0.21) higher age- and sex-adjusted BMI at 13 years old. No significant associations were observed for maternal second and third trimester phthalate urine concentrations with BMI. Thus, higher maternal phthalate metabolites urine concentrations appear to be related to lower BMI at early ages but with higher BMI at later ages.
Collapse
Affiliation(s)
- Chalana M Sol
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Geneviève Delgado
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Kurunthachalam Kannan
- Department of Pediatrics, New York University School of Medicine, New York City, NY, 10016, USA; Wadsworth Center, New York State Department of Health, and Department of Environmental Health Sciences, School of Public Health, State University of New York at Albany, Albany, NY12201, USA; Department of Environmental Medicine, New York University School of Medicine, New York City, NY, 10016, USA
| | - Vincent W V Jaddoe
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands.
| | - Leonardo Trasande
- Department of Pediatrics, New York University School of Medicine, New York City, NY, 10016, USA; Department of Environmental Medicine, New York University School of Medicine, New York City, NY, 10016, USA; Department of Population Health, New York University School of Medicine, New York City, NY, USA; New York Wagner School of Public Service, New York City, NY, 10016, USA; New York University Global Institute of Public Health, New York City, NY, 10016, USA
| | - Susana Santos
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; EPIUnit ITR, Instituto de Saúde Pública da Universidade do Porto, Universidade do Porto, Rua das Taipas, n(o)135, 4050-600, Porto, Portugal
| |
Collapse
|
10
|
Naqvi RA, Valverde A, Shukla D, Naqvi A. Long noncoding RNA PARAL1 regulates myeloid dendritic cell differentiation and TLR signaling. Genes Immun 2025:10.1038/s41435-025-00323-9. [PMID: 40000873 DOI: 10.1038/s41435-025-00323-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 02/03/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025]
Abstract
Dendritic cells (DCs) are professional antigen presentation cells (APCs) that bridge innate and adaptive immune functions to contain pathogenic threats. Long noncoding RNAs (lncRNAs) are implicated in regulating biological processes, including inflammation and immunity. However, the knowledge of myeloid DC-expressed lncRNA repertoire and their regulatory functions is limited. Here we profiled lncRNA expression kinetics during monocyte-to-DC (moDC) differentiation and characterized their functional roles. Our RNA-seq data identified a repertoire of differentially expressed lncRNAs associated with moDC differentiation and a large subset of these lncRNAs are distinct from M1 or M2 macrophages. We selected two DC-enriched lncRNAs and observed that PARAL1 silencing, or overexpression modulates DC surface markers expression. Importantly, PARAL1 RNAi significantly reduced, while its overexpression upregulated the levels of multiple TLRs. Upon treatment with TLR agonists PARAL1 knockdown cells exhibit reduced NF-κB, IRF3 and IRF7 phosphorylation substantiating its role in potentiating TLR signaling. Mechanistically, PARAL1 silencing showed significant downregulation of multiple NF-κB-induced genes and time-dependent inhibition of proinflammatory cytokine secretion upon challenge with TLR agonists. Finally, PARAL1 RNAi in DCs significantly impaired antigen processing and presentation to T cells. Overall, this study characterized novel functions of PARAL1 in regulating DC differentiation, TLR-dependent innate immunity and activation of adaptive immune response.
Collapse
Affiliation(s)
- Raza Ali Naqvi
- Department of Periodontics, College of Dentistry, University of Illinois Chicago, Chicago, IL, 60612, USA
| | - Araceli Valverde
- Department of Periodontics, College of Dentistry, University of Illinois Chicago, Chicago, IL, 60612, USA
| | - Deepak Shukla
- Department of Microbiology and Immunology, College of Medicine, University of Illinois Chicago, Chicago, IL, 60612, USA
- Department of Ophthalmology and Visual Sciences, University of Illinois Medical Center, Chicago, IL, 60612, USA
| | - Afsar Naqvi
- Department of Periodontics, College of Dentistry, University of Illinois Chicago, Chicago, IL, 60612, USA.
- Department of Microbiology and Immunology, College of Medicine, University of Illinois Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
11
|
Xu L, Yang C, Pang K, Zhang Y, He Y, Liu S, Tian H, Shao Z, Wang S, Liu X, Li T, Cao Y, Yan L, Liu J, Wang Y, Li Y, Zhao W, Wang Y, Yan Y, Wang S. Adipocyte Septin-7 attenuates obesogenic adipogenesis and promotes lipolysis to prevent obesity. Mol Metab 2025; 95:102114. [PMID: 40015624 PMCID: PMC11930438 DOI: 10.1016/j.molmet.2025.102114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 02/09/2025] [Accepted: 02/16/2025] [Indexed: 03/01/2025] Open
Abstract
OBJECTIVES The white adipose tissue (WAT) expansion plays a significant role in the development of obesity. Cytoskeletal remodeling directly impacts adipogenic program, however, the precise mechanism remains poorly understood. Here, we identified a crucial role of Septin-7 (SEPT7), a cytoskeleton component, in the regulation of diet-induced processes of adipogenesis, lipogenesis, and lipolysis in WAT. METHODS A high-fat diet (HFD)-induced obesity model was constructed using mice with inducible adipocyte-specific SEPT7 deficiency. The impact of SEPT7 on adipocyte morphology, cell number and metabolism capacity were evaluated with immunofluorescence, isoproterenol induced lipolysis assay, glucose tolerance test and insulin tolerance test. Adipocyte mTmG reporter line was established to trace in vivo adipogenesis. The preadipocyte 3T3-L1 cell was induced for exploring role of SEPT7 in adipocyte differentiation. qRT-PCR and Western-blot were used to investigate the expression of PPARγ, C/EBPα, and HSL in 3T3-L1 cell with siRNA-mediated SEPT7 knockdown. RESULTS SEPT7 expression was greatly induced in obesogenic human and murine adipocytes. Mice lacking SEPT7 in mature white adipocytes demonstrated defective differentiation of preadipocyte into mature adipocytes when fed HFD resulting in larger adipocytes, increased WAT inflammation and reduced lipolysis, which leading to increased WAT mass, liver fat accumulation and impaired glucose tolerance. Mechanistically, we identified SEPT7 restrains store-operated Ca2+ entry (SOCE) and regulates adipocyte adipogenesis and lipolysis by targeting PPARγ, C/EBPα and HSL. CONCLUSIONS We demonstrated that SEPT7 negatively regulates adipogenesis while promotes lipolysis and its repression drives WAT expansion and impaired metabolic health.
Collapse
Affiliation(s)
- Liran Xu
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Chao Yang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710000, China.
| | - Kaidan Pang
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Ying Zhang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710000, China
| | - Yu He
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710000, China
| | - Siyu Liu
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710000, China
| | - Huijing Tian
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710000, China
| | - Zehua Shao
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710000, China
| | - Siyu Wang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710000, China
| | - Xingqian Liu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710000, China
| | - Ting Li
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yapeng Cao
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Luqin Yan
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710000, China
| | - Jinjin Liu
- Patient Service Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710000, China
| | - Yanan Wang
- Med-X institute, Center for Immunological and Metabolic Diseases, and Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710000, China
| | - Yongxin Li
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710000, China
| | - Wei Zhao
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710000, China
| | - Youhua Wang
- Institute of Sports and Exercise Biology, School of Physical Education, Shaanxi Normal University, Xi'an, 710000, China.
| | - Yang Yan
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710000, China.
| | - Shengpeng Wang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710000, China.
| |
Collapse
|
12
|
Bernhardt SM, House CD. Bisphenol A and DDT disrupt adipocyte function in the mammary gland: implications for breast cancer risk and progression. Front Oncol 2025; 15:1490898. [PMID: 40034592 PMCID: PMC11873108 DOI: 10.3389/fonc.2025.1490898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 01/31/2025] [Indexed: 03/05/2025] Open
Abstract
As breast cancer incidence continues to rise worldwide, there is a pressing need to understand the environmental factors that contribute to its development. Obesogens, including Bisphenol A (BPA) and Dichlorodiphenyltrichloroethane (DDT), are highly prevalent in the environment, and have been associated with obesity and metabolic dysregulation. BPA and DDT, known to disrupt hormone signaling in breast epithelial cells, also promote adipogenesis, lipogenesis, and adipokine secretion in adipose tissue, directly contributing to the pathogenesis of obesity. While the adipose-rich mammary gland may be particularly vulnerable to environmental obesogens, there is a scarcity of research investigating obesogen-mediated changes in adipocytes that drive oncogenic transformation of breast epithelial cells. Here, we review the preclinical and clinical evidence linking BPA and DDT to impaired mammary gland development and breast cancer risk. We discuss how the obesogen-driven mechanisms that contribute to obesity, including changes in adipogenesis, lipogenesis, and adipokine secretion, could provide a pro-inflammatory, nutrient-rich environment that promotes activation of oncogenic pathways in breast epithelial cells. Understanding the role of obesogens in breast cancer risk and progression is essential for informing public health guidelines aimed at minimizing obesogen exposure, to ultimately reduce breast cancer incidence and improve outcomes for women.
Collapse
Affiliation(s)
- Sarah M. Bernhardt
- Department of Biology, San Diego State University, San Diego, CA, United States
| | - Carrie D. House
- Department of Biology, San Diego State University, San Diego, CA, United States
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
13
|
Silva AVL, Lima RP, Oliveira FTB, Quinderé ALG, Benevides NMB, Santos FA. Sulfated galactan from Acanthophora muscoides inhibits adipogenesis via regulating adipogenic transcription factors and AMPK in 3T3-L1 cells. BRAZ J BIOL 2025; 85:e289036. [PMID: 39969001 DOI: 10.1590/1519-6984.289036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 12/07/2024] [Indexed: 02/20/2025] Open
Abstract
Obesity is a global public health issue, closely linked to cardiovascular disease and type 2 diabetes. Pharmacological interventions for weight loss are one option for treating obesity; however, these drugs often come with side effects or limited efficacy, highlighting the need for new therapies. Marine algae offer a promising source of biologically active compounds for human health, including antidiabetic, anti-inflammatory, and anti-obesity properties. Sulfated galactan isolated from the red marine algae Acanthophora muscoides (SGAM) has demonstrated diverse biological activities including anti-inflammatory activity in vivo and in vitro studies. However, its potential impact on adipogenesis remains unexplored. This study evaluated the effect of SGAM on adipogenesis in 3T3-L1 cells using Oil Red O staining and analyzed the protein expression of key transcription factors associated with adipogenesis. SGAM (25-100 μg/mL) was found to reduce intracellular lipid accumulation in adipocytes without compromising cell viability. Furthermore, our findings suggest that SGAM significantly inhibits adipocyte differentiation by downregulating the expression of key adipogenesis-related transcription factors, including C/EBPβ, C/EBPδ, C/EBPα, and PPARγ. Additionally, SGAM reduced the protein expression of SREBP-1 and promoted the activation of AMPK. In conclusion, SGAM inhibits adipogenesis by negatively modulating the expression of the main adipogenic transcription factors and activating AMPK.
Collapse
Affiliation(s)
- A V L Silva
- Universidade Federal do Ceará - UFC, Faculdade de Medicina, Departamento de Fisiologia e Farmacologia, Laboratório de Produtos Naturais, Fortaleza, CE, Brasil
| | - R P Lima
- Universidade Federal do Ceará - UFC, Faculdade de Medicina, Departamento de Fisiologia e Farmacologia, Laboratório de Produtos Naturais, Fortaleza, CE, Brasil
| | - F T B Oliveira
- Universidade Federal do Ceará - UFC, Faculdade de Medicina, Departamento de Fisiologia e Farmacologia, Laboratório de Produtos Naturais, Fortaleza, CE, Brasil
| | - A L G Quinderé
- Universidade Federal do Ceará - UFC, Faculdade de Medicina, Departamento de Fisiologia e Farmacologia, Laboratório de Produtos Naturais, Fortaleza, CE, Brasil
| | - N M B Benevides
- Universidade Federal do Ceará - UFC, Departamento de Bioquímica e Biologia Molecular, Laboratório de Carboidratos e Lectinas, Fortaleza, CE, Brasil
| | - F A Santos
- Universidade Federal do Ceará - UFC, Faculdade de Medicina, Departamento de Fisiologia e Farmacologia, Laboratório de Produtos Naturais, Fortaleza, CE, Brasil
| |
Collapse
|
14
|
Inthanon K, Wong-A-Nan N, Dheeranupattana S, Guerra AG, Davies NM, Kesornpun C, Sangher S, Kittakoop P. Regulation of adipocyte differentiation and lipid metabolism by novel synthetic chromenes exploring anti-obesity and broader therapeutic potential. Sci Rep 2025; 15:4051. [PMID: 39900791 PMCID: PMC11790873 DOI: 10.1038/s41598-025-87945-1] [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: 02/29/2024] [Accepted: 01/23/2025] [Indexed: 02/05/2025] Open
Abstract
Obesity poses a significant global health challenge, necessitating the search for novel therapeutic agents to address this epidemic. Chromenes, known for their diverse bioactivities, hold promise as potential anti-obesity compounds, yet research in this area remains limited. This pioneering study represents the first exploration of synthetic chromenes as potential anti-obesity agents, unveiling the underlying molecular pathways governing adipogenesis and lipolysis. Twenty-nine chromenes were synthesized using green chemistry approaches, resulting in five novel compounds: 1, 2, 3, 4, and 5. Among them, 14 chromenes demonstrated a lack of toxicity to pre-adipocytes (PAs) and mature adipocytes (MAs) of 3T3-L1 cells. The anti-adipogenesis and lipolysis enhancement potential of these non-toxic 14 chromenes were comprehensively evaluated using Oil Red O staining technique, LDH activity measurement, and glycerol release assays. Notably, 4, 5, 21 and 25 exhibited remarkable efficacy in reducing intracellular lipid accumulation without inducing cellular stress or cell death. Molecular analysis revealed significant alterations in the expression of key transcription factors involved in adipogenesis and lipid metabolism, including PPARγ, C/EBPα, ADD-1, Pref-1, IRS-1, GLUT-4, adiponectin, FAS, aP2, ATGL, and HSL. This suggests their potential role in anti-adipogenesis. Additionally, the treatments with 4 and 25 showed potential for enhancing lipolysis, providing further evidence of their anti-obesity properties. This study presents several promising prospects for the development of synthetic chromenes as potential anti-obesity agents, opening new avenues for drug discovery and benefitting individuals worldwide in addressing obesity-related challenges to human health. In addition, predictive in silico modeling was performed on the identified candidate chromenes. This modeling provides prospective anti-HIV activity, pharmacokinetic, metabolism, and permeability data, setting the groundwork for further investigation into these potential new chemical entities.
Collapse
Affiliation(s)
- Kewalin Inthanon
- Department of Biotechnology, Faculty Science and Technology, Thammasat University Lampang Campus, Lampang, 52190, Thailand.
| | - Natthawut Wong-A-Nan
- Department of Clinical Pathology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | | | - Andres Garcia Guerra
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Neal M Davies
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Chatchai Kesornpun
- Future Biomanufacturing Research Hub (FBRH), Manchester Institute of Biotechnology, Department of Chemistry, School of Natural Science, The University of Manchester, Manchester, M1 7DN, UK
- Chulabhorn Research Institute, Kamphaeng Phet 6 Road, Laksi, Bangkok, 10210, Thailand
| | - Sasithorn Sangher
- Chulabhorn Research Institute, Kamphaeng Phet 6 Road, Laksi, Bangkok, 10210, Thailand
| | - Prasat Kittakoop
- Chulabhorn Research Institute, Kamphaeng Phet 6 Road, Laksi, Bangkok, 10210, Thailand.
- Chulabhorn Graduate Institute, Program in Chemical Sciences, Kamphaeng Phet 6 Road, Laksi, Bangkok, 10210, Thailand.
- Center of Excellence on Environmental Health and Toxicology (EHT), OPS, Ministry of Higher Education, Science, Research and Innovation, Bangkok, 10400, Thailand.
| |
Collapse
|
15
|
Wu Y, Ma J, Chen J, Liu X, Wang Z, Luo L, Sun C. Ablation of CD44 Attenuates Adipogenesis in White Adipocytes via the Tryptophan 5-Hydroxylase 2/5-Hydroxytryptamine Axis to Protect Mice from High-Fat Diet-Induced Obesity. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:247-264. [PMID: 39476955 DOI: 10.1016/j.ajpath.2024.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 10/09/2024] [Accepted: 10/16/2024] [Indexed: 11/11/2024]
Abstract
CD44 is a transmembrane protein that plays an essential role in transducing extracellular stimuli into intracellular signaling cascades, especially in cancer cells. Recent studies have shown that CD44 contributes to metabolic regulation. However, the effect of CD44 on adipogenesis in white adipose tissue (WAT) remains unclear. Herein, the expression of CD44 was largely increased in the inguinal and epididymal WAT of obese mice. Ablation or neutralization of CD44 inhibited adipogenesis in cultured adipocytes. CD44-deficient mice were resistant to high-fat diet-induced obesity and metabolic dysfunction. RNA-sequencing, together with functional studies, revealed that reduced expression of tryptophan 5-hydroxylase 2 (Tph2) in WAT was responsible for the repressed adipogenesis in the absence of CD44. The application of 5-hydroxytryptamine, a product of TPH2, rescued the repressed adipogenesis induced by CD44 neutralization. Moreover, the inhibition of TPH2 by p-chlorophenylalanine recapitulated the beneficial phenotypes observed in CD44-deficient mice. Taken together, these data indicate that CD44 plays a pivotal role in adipogenesis in WAT. In this regard, CD44 and its downstream target TPH2 may hold great therapeutic potential for treating excessive adiposity-related metabolic disorders, such as obesity, insulin resistance, and type 2 diabetes.
Collapse
Affiliation(s)
- Yuting Wu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Jinyu Ma
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Jing Chen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Xiaoyu Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Zhe Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Lan Luo
- Department of Geriatrics, Affiliated Hospital of Nantong University, Nantong, China.
| | - Cheng Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China.
| |
Collapse
|
16
|
Krupka S, Aldehoff AS, Goerdeler C, Engelmann B, Rolle-Kampczyk U, Schubert K, Klöting N, von Bergen M, Blüher M. Metabolic and molecular Characterization, following dietary exposure to DINCH, Reveals new Implications for its role as a Metabolism-Disrupting chemical. ENVIRONMENT INTERNATIONAL 2025; 196:109306. [PMID: 39884247 DOI: 10.1016/j.envint.2025.109306] [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: 08/29/2024] [Revised: 12/20/2024] [Accepted: 01/24/2025] [Indexed: 02/01/2025]
Abstract
Plastic materials are ubiquitous, leading to constant human exposure to plastic additives such as plasticizers. There is growing evidence that plasticizers may contribute to obesity due to their disruptive effects on metabolism. Alternatives like diisononylcyclohexane-1,2-dicarboxylate (DINCH) are replacing traditional phthalates such as di-(2-ethylhexyl) phthalate (DEHP), which are now banned due to their proven harmful health effects. While DINCH is considered a safer alternative to DEHP and no adipogenic effects have been demonstrated in in vivo studies, recent research suggests that the primary metabolite, monoisononylcyclohexane-1,2-dicarboxylic acid ester (MINCH), promotes adipocyte differentiation and dysfunction in vitro. However, metabolic and molecular effects are not fully understood in vivo. Here, we performed a comprehensive in vivo analysis using C57BL/6N mice to investigate the effects of DINCH on adipose tissue physiology and function. Mice were exposed to two doses of DINCH for 16 weeks, followed by a 10-week recovery period. Tissue analysis confirmed the presence of DINCH and MINCH in liver and adipose tissue after treatment and recovery. After the recovery period, elevated DINCH concentrations in adipose tissue depots indicated possible bioaccumulation. Although no changes were observed in body composition and energy expenditure, sex-specific metabolic effects were identified. Female mice exhibited impaired whole-body insulin sensitivity and higher triglyceride levels, while male mice showed an altered insulin/C-peptide ratio and elevated cholesterol, HDL, and LDL levels. Proteomic profiling of serum, adipose and liver tissues revealed changes in pathways related to central energy metabolism and immune response, highlighting the systemic impact of DINCH, potentially on inflammatory processes. Most effects of DINCH, such as changes in insulin response and serum lipid levels, were diminished after the recovery period. Despite many findings consistent with the existing literature suggesting DINCH as a safer DEHP substitute, the observed sex-specific effects on insulin sensitivity, lipid metabolism and inflammatory processes, as well as potential bioaccumulation and long-term metabolic effects of DINCH exposure warrant careful consideration in further risk assessment.
Collapse
Affiliation(s)
- Sontje Krupka
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Centre München at the University of Leipzig Germany; Department of Endocrinology Nephrology Rheumatology University Hospital Leipzig Medical Research Center Leipzig Germany
| | - Alix Sarah Aldehoff
- Department of Molecular Toxicology, Helmholtz-Centre for Environmental Research GmbH (UFZ) Leipzig Germany
| | - Cornelius Goerdeler
- Department of Molecular Toxicology, Helmholtz-Centre for Environmental Research GmbH (UFZ) Leipzig Germany
| | - Beatrice Engelmann
- Department of Molecular Toxicology, Helmholtz-Centre for Environmental Research GmbH (UFZ) Leipzig Germany
| | - Ulrike Rolle-Kampczyk
- Department of Molecular Toxicology, Helmholtz-Centre for Environmental Research GmbH (UFZ) Leipzig Germany
| | - Kristin Schubert
- Department of Molecular Toxicology, Helmholtz-Centre for Environmental Research GmbH (UFZ) Leipzig Germany.
| | - Nora Klöting
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Centre München at the University of Leipzig Germany.
| | - Martin von Bergen
- Department of Molecular Toxicology, Helmholtz-Centre for Environmental Research GmbH (UFZ) Leipzig Germany; Institute of Biochemistry Faculty of Biosciences, Pharmacy and Psychology Leipzig University Leipzig Germany; German Centre for Integrative Biodiversity Research (iDiv) Halle-Jena-Leipzig Leipzig Germany.
| | - Matthias Blüher
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Centre München at the University of Leipzig Germany; Department of Endocrinology Nephrology Rheumatology University Hospital Leipzig Medical Research Center Leipzig Germany
| |
Collapse
|
17
|
Lu C, Han M, Ma Q, Ying L, Zhang Y. Identification of biomarkers associated with coronary artery disease and non-alcoholic fatty liver disease by bioinformatics analysis and machine learning. Sci Rep 2025; 15:3557. [PMID: 39875572 PMCID: PMC11775188 DOI: 10.1038/s41598-025-87923-7] [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: 04/08/2024] [Accepted: 01/22/2025] [Indexed: 01/30/2025] Open
Abstract
The constantly emerging evidence indicates a close association between coronary artery disease (CAD) and non-alcoholic fatty liver disease (NAFLD). However, the exact mechanisms underlying their mutual relationship remain undefined. This study aims to explore the common signature genes, potential mechanisms, diagnostic markers, and therapeutic targets for CAD and NAFLD. We downloaded CAD and NAFLD datasets from the Gene Expression Omnibus (GEO) database and analyzed the differentially expressed genes (DEGs) by limma. Protein-protein interaction (PPI) network was constructed with common DEGs (co-DEGs), and hub genes were screened by Maximal Clique Centrality (MCC) algorithm. Candidate biomarkers were selected from intersection of three machine learning algorithms. Expression levels, nomogram, the areas under the receiver operating characteristic curve (AUC) of candidate biomarkers were performed. CIBERSORT algorithm was used to assess the immune cell infiltration, and Spearman's correlations tests were used for calculating the correlation of biomarker genes. A total of 554 overlapping DEGs associated with CAD and NAFLD were obtained by analysis of GSE113079 and GSE89632 datasets. Gene Ontology, Kyoto Encyclopedia of Genes and Genomes enrichment analysis showed that the co-DEGs were significantly enriched in immune effector process, inflammation response and lipid metabolism. The PPI network generated a 1245-edge network, and top 50 genes were selected using the MCC algorithm. The candidate biomarkers were screened from intersection of machine learning in GSE89632, including CEBPA, CXCL2, JUN and FOXO1. The ROC results showed that these four biomarker genes have good diagnostic value for patients with both CAD and NAFLD. Then we explored the immune landscape, immune infiltration and the correlation between biomarker gene expression in CAD and NAFLD samples. In this study, we predict that CEBPA, CXCL2, JUN and FOXO1 can be used to diagnose CAD and NAFLD. Our study provided new insights for potential biomarkers, molecular mechanism and therapeutic targets for both diseases.
Collapse
Affiliation(s)
- Chuan Lu
- Department of Cardiology, the Second Hospital of Dalian Medical University, Dalian, 116021, China
| | - Mei Han
- Department of Gastroenterology, the Second Hospital of Dalian Medical University, Dalian, 116021, China
| | - Qiqi Ma
- Department of Gastroenterology, the Second Hospital of Dalian Medical University, Dalian, 116021, China
| | - Li Ying
- Department of Gastroenterology, the Second Hospital of Dalian Medical University, Dalian, 116021, China.
| | - Yue Zhang
- Department of Gastroenterology, the Second Hospital of Dalian Medical University, Dalian, 116021, China.
| |
Collapse
|
18
|
Cho YJ, Lee JB, Lee Y, Lee MS, Choi J. Inhibition of Differentiation of 3T3-L1 Cells by Increasing Glioma-Associated Oncogene Expression in Chrysanthemum indicum L. Using Lactococcus lactis KCTC 3115. Prev Nutr Food Sci 2024; 29:533-545. [PMID: 39759806 PMCID: PMC11699586 DOI: 10.3746/pnf.2024.29.4.533] [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: 08/21/2024] [Revised: 09/21/2024] [Accepted: 09/25/2024] [Indexed: 01/07/2025] Open
Abstract
The inhibitory effect of Chrysanthemum indicum L. on adipocyte differentiation can be enhanced by lactic acid bacteria (LAB) fermentation. In this study, we assessed the cellulose resolution, C. indicum L. quantity, and fermentation time and process to verify the LAB selection and fermentation efficiency. In addition, the antioxidant activity, adipocyte signaling and differentiation, and hedgehog (Hh) signaling were investigated, and the changes in compounds before and after fermentation were determined by ultra-high performance liquid chromatography (UHPLC). All strains exhibited satisfactory cellulose resolution. With 20% C. indicum L., fermentation was only effective up to 24 h. The results of the antioxidant assays showed that the 2,2-diphenyl-1-picrylhydrazyl and 2,2'-azino-bis(3-ethylbenzothiazoline-6-sulfonate) radical scavenging capacities were higher in all fermentations than in unfermented C. indicum L. extract (CI). 3T3-L1 cell differentiation signaling evaluation revealed that CI inhibited adipocyte differentiation by reducing peroxisome proliferator-activated receptor-γ, CCAAT/enhancer binding protein-α, and phosphorylated AMP-activated protein kinase activity in all fermentations. In the Hh signaling analysis, CI fermented with Lactococcus lactis KCTC 3115 significantly increased glioma-associated oncogene 1 (GLI1) activity by inhibiting patched 1 activity and activating smoothened (P<0.001). UHPLC quantitative analysis revealed elevated levels of luteolin and quercetin. Fermentation with C. indicum L. and L. lactis KCTC 3115 activated GLI1, a transcription factor in the Hh signaling pathway, which enhanced the inhibition of adipocyte differentiation, indicating its potential in anti-obesity treatment. However, the exact compounds affecting GLI1 activity require further elucidation in future studies.
Collapse
Affiliation(s)
| | | | - Yunjung Lee
- Department of Hotel & Foodservice Culinary Art, JEI University, Incheon 22573, Korea
| | - Min Soo Lee
- Department of Culinary Arts & Hotel Food Service, Yeonsung University, Gyeonggi 14011, Korea
| | - Jaeyoung Choi
- Department of Culinary Arts & Hotel Food Service, Yeonsung University, Gyeonggi 14011, Korea
| |
Collapse
|
19
|
Sagliocchi S, Schiano E, Acampora L, Iannuzzo F, Cicatiello AG, Miro C, Nappi A, Restolfer F, Stornaiuolo M, Zarrilli S, Guerra F, Tenore GC, Dentice M, Novellino E. AbaComplex Enhances Mitochondrial Biogenesis and Adipose Tissue Browning: Implications for Obesity and Glucose Regulation. Foods 2024; 14:48. [PMID: 39796338 PMCID: PMC11720057 DOI: 10.3390/foods14010048] [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: 10/04/2024] [Revised: 12/04/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
Adipose tissue, particularly white adipose tissue (WAT), plays a central role in energy storage and metabolic regulation. Excess WAT, especially visceral fat, is strongly linked to metabolic disorders such as obesity and type 2 diabetes. The browning of WAT, whereby white fat cells acquire characteristics of brown adipose tissue (BAT) with enhanced thermogenic capacity, represents a promising strategy to enhance metabolic health. In this study, we investigated the effects of chronic supplementation with an infusion based on lyophilized, thin nectarines rich in abscisic acid (ABA), named AbaComplex, on promoting browning of WAT and activating BAT in mice. Over 30 days, C57BL/6 mice were treated with the ABA-rich infusion, and various metabolic and molecular parameters were assessed. The results showed that the AbaComplex significantly increased the expression of browning markers, such as UCP1 and PGC1-α, in both visceral and subcutaneous WAT. Additionally, mitochondrial biogenesis and function were enhanced, evidenced by elevated mitochondrial DNA content and activity. The treatment also reduced the weight of WAT (both visceral and subcutaneous) and BAT and significantly improved glucose uptake in WAT via upregulation of GLUT4, suggesting enhanced insulin sensitivity. Overall, the pronounced browning effect in WAT underscores the potential of AbaComplex as a natural approach for combating obesity and improving metabolic health.
Collapse
Affiliation(s)
- Serena Sagliocchi
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Napoli, Italy; (S.S.); (L.A.); (A.G.C.); (C.M.); (A.N.); (F.R.); (S.Z.)
| | - Elisabetta Schiano
- Inventia Biotech-Healthcare Food Research Center s.r.l., Strada Statale Sannitica KM 20.700, 81020 Caserta, Italy; (E.S.); (F.G.); (E.N.)
| | - Lucia Acampora
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Napoli, Italy; (S.S.); (L.A.); (A.G.C.); (C.M.); (A.N.); (F.R.); (S.Z.)
| | - Fortuna Iannuzzo
- Department of Pharmacy, University of Chieti-Pescara G. D’Annunzio, 66100 Chieti, Italy;
| | - Annunziata Gaetana Cicatiello
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Napoli, Italy; (S.S.); (L.A.); (A.G.C.); (C.M.); (A.N.); (F.R.); (S.Z.)
| | - Caterina Miro
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Napoli, Italy; (S.S.); (L.A.); (A.G.C.); (C.M.); (A.N.); (F.R.); (S.Z.)
| | - Annarita Nappi
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Napoli, Italy; (S.S.); (L.A.); (A.G.C.); (C.M.); (A.N.); (F.R.); (S.Z.)
| | - Federica Restolfer
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Napoli, Italy; (S.S.); (L.A.); (A.G.C.); (C.M.); (A.N.); (F.R.); (S.Z.)
| | - Mariano Stornaiuolo
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano 59, 80131 Napoli, Italy; (M.S.); (G.C.T.)
| | - Stefano Zarrilli
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Napoli, Italy; (S.S.); (L.A.); (A.G.C.); (C.M.); (A.N.); (F.R.); (S.Z.)
| | - Fabrizia Guerra
- Inventia Biotech-Healthcare Food Research Center s.r.l., Strada Statale Sannitica KM 20.700, 81020 Caserta, Italy; (E.S.); (F.G.); (E.N.)
| | - Gian Carlo Tenore
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano 59, 80131 Napoli, Italy; (M.S.); (G.C.T.)
| | - Monica Dentice
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Napoli, Italy; (S.S.); (L.A.); (A.G.C.); (C.M.); (A.N.); (F.R.); (S.Z.)
| | - Ettore Novellino
- Inventia Biotech-Healthcare Food Research Center s.r.l., Strada Statale Sannitica KM 20.700, 81020 Caserta, Italy; (E.S.); (F.G.); (E.N.)
- Faculty of Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy
| |
Collapse
|
20
|
Lee K, Kim HJ, Kim JY, Shim JJ, Lee JH. Synergistic Effect of Lactobacillus Mixtures and Lagerstroemia speciosa Leaf Extract in Reducing Obesity in High-Fat Diet-Fed Mice. BIOLOGY 2024; 13:1047. [PMID: 39765714 PMCID: PMC11673097 DOI: 10.3390/biology13121047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 12/06/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025]
Abstract
In this study, we describe the anti-obesity effects of a novel combination of Lactobacillus mixture (Lactobacillus curvatus HY7601 and Lactobacillus plantarum KY1032) and leaf extract of Lagerstroemia speciosa (L. speciosa) in mice. The administration of the probiotic mixture of HY7601 and KY1032 in combination with the leaf extract of L. speciosa significantly attenuated fat tissue formation and body weight gain in mice fed a high-fat diet. The white adipose fat mass, comprising the inguinal and epididymal fat pads, was most effectively reduced when the probiotic mixture and L. speciosa leaf extract was orally administered to the mice in combination. This combination also reduced the mRNA expression of adipogenic genes (those encoding CCAAT/enhancer-binding protein alpha, peroxisome proliferator-activated receptor gamma, and fatty acid-binding protein 4) in inguinal and epididymal white adipose tissue depots and the liver. Finally, the combination of reduced blood glucose concentrations regulated the insulin resistance of high-fat diet-fed obese mice. These findings provide insight into the mechanisms underlying the effect of this combination and suggest that using Lactobacillus mixture (HY7601 and KY1032) is as safe as microbial monotherapy, but more effective at preventing obesity.
Collapse
Affiliation(s)
| | | | - Joo Yun Kim
- R&BD Center, hy Co., Ltd., 22 Giheungdanji-ro 24 Beon-gil, Giheung-gu, Yongin-si 17086, Republic of Korea; (K.L.); (H.-J.K.); (J.J.S.)
| | | | - Jae Hwan Lee
- R&BD Center, hy Co., Ltd., 22 Giheungdanji-ro 24 Beon-gil, Giheung-gu, Yongin-si 17086, Republic of Korea; (K.L.); (H.-J.K.); (J.J.S.)
| |
Collapse
|
21
|
Ganesan S, Awan-Toor S, Guidez F, Maslah N, Rahimy R, Aoun C, Gou P, Guiguen C, Soret J, Ravdan O, Bisio V, Dulphy N, Lobry C, Schlageter MH, Souyri M, Giraudier S, Kiladjian JJ, Chomienne C, Cassinat B. Comprehensive analysis of mesenchymal cells reveals a dysregulated TGF-β/WNT/HOXB7 axis in patients with myelofibrosis. JCI Insight 2024; 9:e173665. [PMID: 39470742 PMCID: PMC11623938 DOI: 10.1172/jci.insight.173665] [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: 07/12/2023] [Accepted: 10/22/2024] [Indexed: 11/01/2024] Open
Abstract
Despite the advances in the understanding and treatment of myeloproliferative neoplasm (MPN), the disease remains incurable with the risk of evolution to acute myeloid leukemia or myelofibrosis (MF). Unfortunately, the evolution of the disease to MF remains poorly understood, impeding preventive and therapeutic options. Recent studies in solid tumor microenvironment and organ fibrosis have shed instrumental insights on their respective pathogenesis and drug resistance, yet such precise data are lacking in MPN. In this study, through a patient sample-driven transcriptomic and epigenetic description of the MF microenvironment landscape and cell-based analyses, we identify homeobox B7 (HOXB7) overexpression and more precisely a potentially novel TGF-β/WNT/HOXB7 pathway as associated to a pro-fibrotic and pro-osteoblastic biased differentiation of mesenchymal stromal cells (MSCs). Using gene-based and chemical inhibition of this pathway, we reversed the abnormal phenotype of MSCs from patients with MF, providing the MPN field a potentially novel target to prevent and manage evolution to MF.
Collapse
Affiliation(s)
- Saravanan Ganesan
- INSERM UMRS 1131, Institut de Recherche Saint-Louis, Université Paris Cité, Paris, France
| | - Sarah Awan-Toor
- INSERM UMRS 1131, Institut de Recherche Saint-Louis, Université Paris Cité, Paris, France
| | - Fabien Guidez
- INSERM UMRS 1131, Institut de Recherche Saint-Louis, Université Paris Cité, Paris, France
- INSERM U1232/LNC, Team Epi2THM, Université Bourgogne Franche-Comté, Dijon, France
| | - Nabih Maslah
- INSERM UMRS 1131, Institut de Recherche Saint-Louis, Université Paris Cité, Paris, France
- Service de Biologie Cellulaire, Hôpital Saint-Louis, AP-HP, Paris, France
| | - Rifkath Rahimy
- Laboratoire de recherche en génétique et hématologie translationnelle, Institut Gonçalo Moniz, Salvador, Bahia, Brazil
| | - Céline Aoun
- INSERM UMRS 1131, Institut de Recherche Saint-Louis, Université Paris Cité, Paris, France
| | - Panhong Gou
- INSERM UMRS 1131, Institut de Recherche Saint-Louis, Université Paris Cité, Paris, France
| | - Chloé Guiguen
- INSERM UMRS 1131, Institut de Recherche Saint-Louis, Université Paris Cité, Paris, France
| | - Juliette Soret
- INSERM CIC 1427, Université Paris Cité, Centre d’Investigations Cliniques, Hôpital Saint-Louis, AP-HP, Paris, France
| | - Odonchimeg Ravdan
- Service de Biologie Cellulaire, Hôpital Saint-Louis, AP-HP, Paris, France
| | - Valeria Bisio
- INSERM UMRS 1160, Institut de Recherche Saint-Louis, Université Paris-Cité, Paris, France
| | - Nicolas Dulphy
- INSERM UMRS 1160, Institut de Recherche Saint-Louis, Université Paris-Cité, Paris, France
- Laboratoire d’Immunologie et d’Histocompatibilite, Hôpital Saint-Louis, AP-HP, Paris, France
| | - Camille Lobry
- INSERM U944, CNRS UMR7212, Institut de Recherche Saint-Louis, Université Paris-Cité, Paris, France
| | | | - Michèle Souyri
- INSERM UMRS 1131, Institut de Recherche Saint-Louis, Université Paris Cité, Paris, France
| | - Stéphane Giraudier
- INSERM UMRS 1131, Institut de Recherche Saint-Louis, Université Paris Cité, Paris, France
- Service de Biologie Cellulaire, Hôpital Saint-Louis, AP-HP, Paris, France
| | - Jean-Jacques Kiladjian
- INSERM UMRS 1131, Institut de Recherche Saint-Louis, Université Paris Cité, Paris, France
- INSERM CIC 1427, Université Paris Cité, Centre d’Investigations Cliniques, Hôpital Saint-Louis, AP-HP, Paris, France
| | - Christine Chomienne
- INSERM UMRS 1131, Institut de Recherche Saint-Louis, Université Paris Cité, Paris, France
| | - Bruno Cassinat
- INSERM UMRS 1131, Institut de Recherche Saint-Louis, Université Paris Cité, Paris, France
- Service de Biologie Cellulaire, Hôpital Saint-Louis, AP-HP, Paris, France
| |
Collapse
|
22
|
Sorokina M, Bobkov D, Khromova N, Vilchinskaya N, Shenkman B, Kostareva A, Dmitrieva R. Fibro-adipogenic progenitor cells in skeletal muscle unloading: metabolic and functional impairments. Skelet Muscle 2024; 14:31. [PMID: 39639402 PMCID: PMC11622572 DOI: 10.1186/s13395-024-00362-2] [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: 08/26/2024] [Accepted: 11/20/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND Skeletal muscle resident fibro-adipogenic progenitor cells (FAPs) control skeletal muscle regeneration providing a supportive role for muscle stem cells. Altered FAPs characteristics have been shown for a number of pathological conditions, but the influence of temporary functional unloading of healthy skeletal muscle on FAPs remains poorly studied. This work is aimed to investigate how skeletal muscle disuse affects the functionality and metabolism of FAPs. METHODS Hindlimb suspension (HS) rat model employed to investigate muscle response to decreased usage. FAPs were purified from m. soleus functioning muscle (Contr) and after functional unloading for 7 and 14 days (HS7 and HS14). FAPs were expanded in vitro, and tested for: immunophenotype; in vitro expansion rate, and migration activity; ability to differentiate into adipocytes in vitro; metabolic changes. Crosstalk between FAPs and muscle stem cells was estimated by influence of medium conditioned by FAP's on migration and myogenesis of C2C12 myoblasts. To reveal the molecular mechanisms behind unloading-induced alterations in FAP's functionality transcriptome analysis was performed. RESULTS FAPs isolated from Contr and HS muscles exhibited phenotype of MSC cells. FAPs in vitro expansion rate and migration were altered by functional unloading conditions. All samples of FAPs demonstrated the ability to adipogenic differentiation in vitro, however, HS FAPs formed fat droplets of smaller volume and transcriptome analysis showed fatty acids metabolism and PPAR signaling suppression. Skeletal muscle unloading resulted in metabolic reprogramming of FAPs: decreased spare respiratory capacity, decreased OCR/ECAR ratio detected in both HS7 and HS14 samples point to reduced oxygen consumption, decreased potential for substrate oxidation and a shift to glycolytic metabolism. Furthermore, C2C12 cultures treated with medium conditioned by FAPs showed diverse alterations: while the HS7 FAPs-derived paracrine factors supported the myoblasts fusion, the HS14-derived medium stimulated proliferation of C2C12 myoblasts; these observations were supported by increased expression of cytokines detected by transcriptome analysis. CONCLUSION the results obtained in this work show that the skeletal muscle functional unloading affects properties of FAPs in time-dependent manner: in atrophying skeletal muscle FAPs act as the sensors for the regulatory signals that may stimulate the metabolic and transcriptional reprogramming to preserve FAPs properties associated with maintenance of skeletal muscle homeostasis during unloading and in course of rehabilitation.
Collapse
Affiliation(s)
| | - Danila Bobkov
- Almazov National Medical Research Centre, Saint Petersburg, Russia
- Institute of Cytology, Russian Academy of Sciences, Saint Petersburg, Russia
| | - Natalia Khromova
- Almazov National Medical Research Centre, Saint Petersburg, Russia
| | | | - Boris Shenkman
- Myology Laboratory, Institute of Biomedical Problems RAS, Moscow, Russia
| | - Anna Kostareva
- Almazov National Medical Research Centre, Saint Petersburg, Russia
- Department of Women's and Children's Health and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Renata Dmitrieva
- Almazov National Medical Research Centre, Saint Petersburg, Russia.
| |
Collapse
|
23
|
Yang Y, Fan C, Zhang Y, Kang T, Jiang J. Untargeted Metabolomics Reveals the Role of Lipocalin-2 in the Pathological Changes of Lens and Retina in Diabetic Mice. Invest Ophthalmol Vis Sci 2024; 65:19. [PMID: 39656472 PMCID: PMC11636665 DOI: 10.1167/iovs.65.14.19] [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: 06/24/2024] [Accepted: 11/07/2024] [Indexed: 12/14/2024] Open
Abstract
Purpose To identify the role of lipocalin-2 (LCN2) in diabetic cataract (DC) and diabetic retinopathy (DR), diabetes models were established in wild-type (WT) and LCN2 gene knockout (LCN2-/-) mice by streptozotocin (STZ), this study aimed to investigate the metabolic alterations and underlying pathways in the lens and retina. Methods Untargeted metabolomic analysis was performed on the lenses and retinas of WT and LCN2-/- diabetic mice, and relevant pathways were predicted through bioinformatics analysis. Results LCN2 was notably elevated in the anterior capsules of DC and the vitreous humor of DR. Metabolic profiling of the lenses and retinas of diabetic mice indicated that the differential metabolites were mostly amino acids, fatty acids, carbohydrates, and their derivatives. In the lenses of STZ-induced WT mice, the differential abundance score (DA-score) revealed an increase in metabolites associated with the citrate (or TCA) cycle and glucagon signaling pathway, whereas a decrease was observed in metabolites related to cholesterol metabolism. After the knockout of LCN2, the DA-score indicated that the majority of metabolites involved in cholesterol metabolism, cysteine and methionine metabolism, and tryptophan metabolism were diminished. In the STZ-induced retina, there was an increase in metabolites associated with the mTOR signaling pathway, and this increase was inhibited by the knockout of LCN2. Conclusions Numerous metabolites exhibited substantial alterations in the lenses and retinas of diabetic mice. Untargeted metabolomics has provided insights into the function of LCN2 in DC and DR. These changes in metabolites, along with their related pathways, could be the mechanisms by which LCN2 modulated DC and DR.
Collapse
Affiliation(s)
- Yu Yang
- Eye Center of Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Cong Fan
- Eye Center of Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yue Zhang
- Eye Center of Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Tianyi Kang
- Eye Center of Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Jian Jiang
- Eye Center of Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
24
|
Yilmaz B, Erdogan CS, Sandal S, Kelestimur F, Carpenter DO. Obesogens and Energy Homeostasis: Definition, Mechanisms of Action, Exposure, and Adverse Effects on Human Health. Neuroendocrinology 2024; 115:72-100. [PMID: 39622213 DOI: 10.1159/000542901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 11/28/2024] [Indexed: 02/26/2025]
Abstract
BACKGROUND Obesity is a major risk factor for noncommunicable diseases and is associated with a reduced life expectancy of up to 20 years, as well as with other consequences such as unemployment and increased economic burden for society. It is a multifactorial disease, and physiopathology of obesity involves dysregulated calorie utilization and energy balance, disrupted homeostasis of appetite and satiety, lifestyle factors including sedentary lifestyle, lower socioeconomic status, genetic predisposition, epigenetics, and environmental factors. Some endocrine-disrupting chemicals (EDCs) have been proposed as "obesogens" that stimulate adipogenesis leading to obesity. In this review, definition of obesogens, their adverse effects, underlying mechanisms, and metabolic implications will be updated and discussed. SUMMARY Disruption of lipid homeostasis by EDCs involves multiple mechanisms including increase in the number and size of adipocytes, disruption of endocrine-regulated adiposity and metabolism, alteration of hypothalamic regulation of appetite, satiety, food preference and energy balance, and modification of insulin sensitivity in the liver, skeletal muscle, pancreas, gastrointestinal system, and the brain. At a cellular level, obesogens can exert their endocrine disruptive effects by interfering with peroxisome proliferator-activated receptors and steroid receptors. Human exposure to chemical obesogens mainly occurs by ingestion and, to some extent, by inhalation and dermal uptake, usually in an unconscious manner. Persistent pollutants are lipophilic features; thus, they bioaccumulate in adipose tissue. KEY MESSAGES Although there are an increasing number of reports studying the effects of obesogens, their mechanisms of action remain to be elucidated. In addition, epidemiological studies are needed in order to evaluate human exposure to obesogens.
Collapse
Affiliation(s)
- Bayram Yilmaz
- Department of Physiology, Faculty of Medicine, Yeditepe University, Istanbul, Turkey
- Izmir Biomedicine and Genome Center, Izmir, Turkey
- Department of Physiology, Faculty of Medicine, Dokuz Eylül University, Izmir, Turkey
| | | | - Suleyman Sandal
- Department of Physiology, Faculty of Medicine, Inonu University, Malatya, Turkey
| | - Fahrettin Kelestimur
- Department of Clinical Endocrinology, Faculty of Medicine, Yeditepe University, Istanbul, Turkey
| | - David O Carpenter
- Institute for Health and the Environment, 5 University Place, University at Albany, Rensselaer, New York, USA
| |
Collapse
|
25
|
Zohdi RM, Adli MA, Mukhtar SM, Awang Junaidi AH, Bakar MZA. Sygyzium claviflorum fruit extract preadipocyte differentiation inhibition in 3T3-L1 cells. J Taibah Univ Med Sci 2024; 19:1181-1192. [PMID: 39807375 PMCID: PMC11728925 DOI: 10.1016/j.jtumed.2024.12.008] [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: 08/18/2024] [Revised: 11/01/2024] [Accepted: 12/04/2024] [Indexed: 01/16/2025] Open
Abstract
Objective Concerns over the increasing number of obese individuals and the associated health risks have prompted therapeutic option explorations. Similarly, this study aimed to establish Sygyzium claviflorum fruit extract (SCFE) anti-adipogenic attributes in 3T3-L1 cells. Methods The polyphenolic compounds in SCFE were identified with Reverse phase-high performance liquid chromatography (RP-HPLC). Meanwhile, murine 3T3-L1 preadipocytes, measuring leptin levels, reactive oxygen species (ROS), and lipid and triglyceride (TG) contents were utilized during anti-adipogenic activity assessments. Concurrently, the effects of SCFE on adipogenic transcription factors were established with quantitative real-time-polymerase chain reaction (qRT-PCR). Results The RP-HPLC results indicated three polyphenolic compounds in SCFE, including one flavonoid (naringin) and two phenolic acids (syringic and p-coumaric). Although SCFE treatments (250-1000 μg/mL) did not result in cell toxicity, they significantly reduced dose-dependent lipid accumulation, ROS production, and TG and leptin levels relative to control-differentiated adipocytes. Moreover, SCFE suppressed sterol regulatory element binding protein-1 (SREBP-1), peroxisome proliferator-activated receptor-gamma (PPAR-γ), and CCAAT/enhancer-binding protein-alpha (C/EBP-α) gene expressions during preadipocyte differentiation into adipocytes. Conclusion The findings revealed the anti-adipogenic properties of SCFE, indicating its potential as a natural obesity management remedy. Nevertheless, more studies are necessary to elucidate the reactions resulting in SCFE anti-adipogenic effects and the active constituents responsible for the property.
Collapse
Affiliation(s)
- Rozaini M. Zohdi
- Atta-ur-Rahman Institute for Natural Product Discovery, Universiti Teknologi MARA, Selangor Branch, Puncak Alam, Selangor, Malaysia
- Department of Pharmacology and Life Sciences, Faculty of Pharmacy, Universiti Teknologi MARA, Selangor Branch, Puncak Alam, Selangor, Malaysia
| | - Muhammad A. Adli
- Atta-ur-Rahman Institute for Natural Product Discovery, Universiti Teknologi MARA, Selangor Branch, Puncak Alam, Selangor, Malaysia
- Department of Pharmacology and Life Sciences, Faculty of Pharmacy, Universiti Teknologi MARA, Selangor Branch, Puncak Alam, Selangor, Malaysia
| | - Shahida M. Mukhtar
- Atta-ur-Rahman Institute for Natural Product Discovery, Universiti Teknologi MARA, Selangor Branch, Puncak Alam, Selangor, Malaysia
| | - Awang H. Awang Junaidi
- Department of Veterinary Pre-Clinical Science, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Md Zuki A. Bakar
- Department of Veterinary Pre-Clinical Science, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| |
Collapse
|
26
|
Al Harake SN, Abedin Y, Hatoum F, Nassar NZ, Ali A, Nassar A, Kanaan A, Bazzi S, Azar S, Harb F, Ghadieh HE. Involvement of a battery of investigated genes in lipid droplet pathophysiology and associated comorbidities. Adipocyte 2024; 13:2403380. [PMID: 39329369 PMCID: PMC11445895 DOI: 10.1080/21623945.2024.2403380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 08/29/2024] [Accepted: 09/05/2024] [Indexed: 09/28/2024] Open
Abstract
Lipid droplets (LDs) are highly specialized energy storage organelles involved in the maintenance of lipid homoeostasis by regulating lipid flux within white adipose tissue (WAT). The physiological function of adipocytes and LDs can be compromised by mutations in several genes, leading to NEFA-induced lipotoxicity, which ultimately manifests as metabolic complications, predominantly in the form of dyslipidemia, ectopic fat accumulation, and insulin resistance. In this review, we delineate the effects of mutations and deficiencies in genes - CIDEC, PPARG, BSCL2, AGPAT2, PLIN1, LIPE, LMNA, CAV1, CEACAM1, and INSR - involved in lipid droplet metabolism and their associated pathophysiological impairments, highlighting their roles in the development of lipodystrophies and metabolic dysfunction.
Collapse
Affiliation(s)
- Sami N. Al Harake
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Yasamin Abedin
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Fatema Hatoum
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Nour Zahraa Nassar
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Ali Ali
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Aline Nassar
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Amjad Kanaan
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Samer Bazzi
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Sami Azar
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Frederic Harb
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Hilda E. Ghadieh
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| |
Collapse
|
27
|
Liermann-Wooldrik KT, Kosmacek EA, Oberley-Deegan RE. Adipose Tissues Have Been Overlooked as Players in Prostate Cancer Progression. Int J Mol Sci 2024; 25:12137. [PMID: 39596205 PMCID: PMC11594286 DOI: 10.3390/ijms252212137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/07/2024] [Accepted: 11/10/2024] [Indexed: 11/28/2024] Open
Abstract
Obesity is a common risk factor in multiple tumor types, including prostate cancer. Obesity has been associated with driving metastasis, therapeutic resistance, and increased mortality. The effect of adipose tissue on the tumor microenvironment is still poorly understood. This review aims to highlight the work conducted in the field of obesity and prostate cancer and bring attention to areas where more research is needed. In this review, we have described key differences between healthy adipose tissues and obese adipose tissues, as they relate to the tumor microenvironment, focusing on mechanisms related to metabolic changes, abnormal adipokine secretion, altered immune cell presence, and heightened oxidative stress as drivers of prostate cancer formation and progression. Interestingly, common treatment options for prostate cancer ignore the adipose tissue located near the site of the tumor. Because of this, we have outlined how excess adipose tissue potentially affects therapeutics' efficacy, such as androgen deprivation, chemotherapy, and radiation treatment, and identified possible drug targets to increase prostate cancer responsiveness to clinical treatments. Understanding how obesity affects the tumor microenvironment will pave the way for understanding why some prostate cancers become metastatic or treatment-resistant, and why patients experience recurrence.
Collapse
Affiliation(s)
| | | | - Rebecca E. Oberley-Deegan
- Department of Biochemistry and Molecular Biology, 985870 University of Nebraska Medical Center, Omaha, NE 68198, USA; (K.T.L.-W.)
| |
Collapse
|
28
|
Nobushi Y, Wada T, Miura M, Onoda R, Ishiwata R, Oikawa N, Shigematsu K, Nakakita T, Toriyama M, Shimba S, Kishikawa Y. Effects of Flavanone Derivatives on Adipocyte Differentiation and Lipid Accumulation in 3T3-L1 Cells. Life (Basel) 2024; 14:1446. [PMID: 39598244 PMCID: PMC11595554 DOI: 10.3390/life14111446] [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: 10/10/2024] [Revised: 11/05/2024] [Accepted: 11/06/2024] [Indexed: 11/29/2024] Open
Abstract
Flavanones, a class of flavonoids, are abundant in fruits, vegetables, and herbs. They are known to have several biological activities, such as anti-inflammatory and anti-cancer activities, but their effects on obesity remain unclear. Obesity is closely associated with adipocyte differentiation and lipid accumulation in adipose tissue. Therefore, in this study, we examined the effects of flavanone derivatives on adipocyte differentiation and lipid accumulation by using 3T3-L1 cells. Among the 15 flavanone derivatives studied, 4'-phenylflavanone (4PF), with a biphenyl structure, significantly inhibited adipocyte differentiation-related lipid accumulation in 3T3-L1 cells; this inhibition of lipid accumulation was dose-dependent. Gene expression analysis showed that 4PF suppressed the expression of adipogenic marker genes. Although the induction of peroxisome proliferator activator γ2 (Pparγ2), a master regulator of adipocyte differentiation, and its target genes during adipocyte differentiation was attenuated in 4PF-treated cells, 4PF did not directly regulate Pparγ2 gene expression and its activation. In contrast, 4PF suppressed mitotic clonal expansion (MCE), which is associated with changes in the expression of proliferation-related genes at the early stages of adipocyte differentiation. Taken together, these results suggest that 4PF inhibits lipid accumulation because it suppresses MCE during adipocyte differentiation. Thus, our findings may help in the development of anti-obesity drugs.
Collapse
Affiliation(s)
- Yasuhito Nobushi
- Laboratory of Clinical Pharmacy, School of Pharmacy, Nihon University, 7-7-1, Narashinodai, Funabashi 274-8555, Chiba, Japan; (R.O.); (R.I.); (Y.K.)
| | - Taira Wada
- Laboratory of Health Science, School of Pharmacy, Nihon University, 7-7-1, Narashinodai, Funabashi 274-8555, Chiba, Japan; (T.W.); (S.S.)
| | - Motofumi Miura
- Laboratory of Molecular Chemistry, School of Pharmacy, Nihon University, 7-7-1, Narashinodai, Funabashi 274-8555, Chiba, Japan; (M.M.); (K.S.); (M.T.)
| | - Rikuto Onoda
- Laboratory of Clinical Pharmacy, School of Pharmacy, Nihon University, 7-7-1, Narashinodai, Funabashi 274-8555, Chiba, Japan; (R.O.); (R.I.); (Y.K.)
| | - Ryuta Ishiwata
- Laboratory of Clinical Pharmacy, School of Pharmacy, Nihon University, 7-7-1, Narashinodai, Funabashi 274-8555, Chiba, Japan; (R.O.); (R.I.); (Y.K.)
| | - Naoki Oikawa
- Laboratory of Medicinal Chemistry, School of Pharmacy, Nihon University, 7-7-1, Narashinodai, Funabashi 274-8555, Chiba, Japan;
| | - Karin Shigematsu
- Laboratory of Molecular Chemistry, School of Pharmacy, Nihon University, 7-7-1, Narashinodai, Funabashi 274-8555, Chiba, Japan; (M.M.); (K.S.); (M.T.)
| | - Toshinori Nakakita
- Medicine Analysis Research Laboratory, Yokohama University of Pharmacy, 601, Matano-cho, Totsuka-ku, Yokohama 245-0066, Kanagawa, Japan;
| | - Masaharu Toriyama
- Laboratory of Molecular Chemistry, School of Pharmacy, Nihon University, 7-7-1, Narashinodai, Funabashi 274-8555, Chiba, Japan; (M.M.); (K.S.); (M.T.)
| | - Shigeki Shimba
- Laboratory of Health Science, School of Pharmacy, Nihon University, 7-7-1, Narashinodai, Funabashi 274-8555, Chiba, Japan; (T.W.); (S.S.)
| | - Yukinaga Kishikawa
- Laboratory of Clinical Pharmacy, School of Pharmacy, Nihon University, 7-7-1, Narashinodai, Funabashi 274-8555, Chiba, Japan; (R.O.); (R.I.); (Y.K.)
| |
Collapse
|
29
|
Kucera J, Chalupova Z, Wabitsch M, Bienertova-Vasku J. Endocrine disruption of adipose physiology: Screening in SGBS cells. J Appl Toxicol 2024; 44:1784-1792. [PMID: 39044430 DOI: 10.1002/jat.4679] [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: 06/12/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 07/25/2024]
Abstract
The increasing use of industrial chemicals has raised concerns regarding exposure to endocrine-disrupting chemicals (EDCs), which interfere with developmental, reproductive and metabolic processes. Of particular concern is their interaction with adipose tissue, a vital component of the endocrine system regulating metabolic and hormonal functions. The SGBS (Simpson Golabi Behmel Syndrome) cell line, a well-established human-relevant model for adipocyte research, closely mimics native adipocytes' properties. It responds to hormonal stimuli, undergoes adipogenesis and has been successfully used to study the impact of EDCs on adipose biology. In this study, we screened human exposure-relevant doses of various EDCs on the SGBS cell line to investigate their effects on viability, lipid accumulation and adipogenesis-related protein expression. Submicromolar doses were generally well tolerated; however, at higher doses, EDCs compromised cell viability, with cadmium chloride (CdCl2) showing the most pronounced effects. Intracellular lipid levels remained unaffected by EDCs, except for tributyltin (TBT), used as a positive control, which induced a significant increase. Analysis of adipogenesis-related protein expression revealed several effects, including downregulation of fatty acid-binding protein 4 (FABP4) by dibutyl phthalate, upregulation by CdCl2 and downregulation of perilipin 1 and FABP4 by perfluorooctanoic acid. Additionally, TBT induced dose-dependent upregulation of C/EBPα, perilipin 1 and FABP4 protein expression. These findings underscore the importance of employing appropriate models to study EDC-adipocyte interactions. Conclusions from this research could guide strategies to reduce the negative impacts of EDC exposure on adipose tissue.
Collapse
Affiliation(s)
- Jan Kucera
- RECETOX, Faculty of Science, Masaryk University, Brno, Czech Republic
- Department of Physical Activities and Health Sciences, Faculty of Sports Studies, Masaryk University, Brno, Czech Republic
| | - Zuzana Chalupova
- Department of Internal Medicine and Cardiology, University Hospital Brno, Brno, Czech Republic
| | - Martin Wabitsch
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, University of Ulm, Ulm, Germany
| | - Julie Bienertova-Vasku
- Department of Physical Activities and Health Sciences, Faculty of Sports Studies, Masaryk University, Brno, Czech Republic
| |
Collapse
|
30
|
Guo G, Wang W, Tu M, Zhao B, Han J, Li J, Pan Y, Zhou J, Ma W, Liu Y, Sun T, Han X, An Y. Deciphering adipose development: Function, differentiation and regulation. Dev Dyn 2024; 253:956-997. [PMID: 38516819 DOI: 10.1002/dvdy.708] [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: 11/07/2023] [Revised: 03/02/2024] [Accepted: 03/10/2024] [Indexed: 03/23/2024] Open
Abstract
The overdevelopment of adipose tissues, accompanied by excess lipid accumulation and energy storage, leads to adipose deposition and obesity. With the increasing incidence of obesity in recent years, obesity is becoming a major risk factor for human health, causing various relevant diseases (including hypertension, diabetes, osteoarthritis and cancers). Therefore, it is of significance to antagonize obesity to reduce the risk of obesity-related diseases. Excess lipid accumulation in adipose tissues is mediated by adipocyte hypertrophy (expansion of pre-existing adipocytes) or hyperplasia (increase of newly-formed adipocytes). It is necessary to prevent excessive accumulation of adipose tissues by controlling adipose development. Adipogenesis is exquisitely regulated by many factors in vivo and in vitro, including hormones, cytokines, gender and dietary components. The present review has concluded a comprehensive understanding of adipose development including its origin, classification, distribution, function, differentiation and molecular mechanisms underlying adipogenesis, which may provide potential therapeutic strategies for harnessing obesity without impairing adipose tissue function.
Collapse
Affiliation(s)
- Ge Guo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Wanli Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Mengjie Tu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Binbin Zhao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Jiayang Han
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Jiali Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Yanbing Pan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Jie Zhou
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Wen Ma
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Yi Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Tiantian Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Xu Han
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Yang An
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| |
Collapse
|
31
|
Kim S, Na GH, Yim DJ, Liu CF, Lin TH, Shih TW, Pan TM, Lee CL, Koo YK. Lactobacillus paracasei subsp. paracasei NTU 101 prevents obesity by regulating AMPK pathways and gut microbiota in obese rat. Biochem Biophys Res Commun 2024; 731:150279. [PMID: 39018972 DOI: 10.1016/j.bbrc.2024.150279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/10/2024] [Accepted: 06/17/2024] [Indexed: 07/19/2024]
Abstract
This study assessed the anti-obesity effects of Lactobacillus paracasei subsp. paracasei NTU 101 (NTU 101) both in vitro and in vivo. Initially, the cytotoxicity and lipid accumulation inhibitory effects of NTU 101 on 3T3-L1 cells were evaluated using the MTT assay and oil red O assay, respectively. Subsequently, the anti-obesity effects of NTU 101 were investigated in high-fat diet-induced obese rat. Moreover, western blotting was performed to measure the obesity-related protein expression of PPARα, PPARβ, PPARγ, C/EBPα, C/EBPβ, ATGL, p-p38 MAPK, p-ERK1/2, p-AMPK and CPT-1 in both 3T3-L1 adipocytes and adipose and liver tissues. Treatment with 16 × 108 CFU/mL NTU 101 reduced lipid accumulation in 3T3-L1 adipocytes by more than 50 %. Oral administration of NTU 101 significantly attenuated body weight gain, as well as adipose tissue weight. NTU 101 administration enhanced fatty acid oxidation increasing expression levels of PPARα, CPT-1, and p-AMPK proteins in liver tissue, while simultaneously inhibited adipogenesis by reducing PPARγ and C/EBPα proteins in adipose tissue. Furthermore, NTU 101 supplementation positively modulated the composition of gut microbiota, notably increasing the abundance of Akkermansiaceae. This present study suggests that NTU 101 exerts anti-obesity effects by regulating gut microbiota, fatty acid oxidation, lipolysis and adipogenesis.
Collapse
Affiliation(s)
- SukJin Kim
- Department of R&I Center, COSMAXBIO, Seongnam, 13486, South Korea
| | - Gwi Hwan Na
- Department of R&I Center, COSMAXBIO, Seongnam, 13486, South Korea
| | - Dong Joon Yim
- Department of R&I Center, COSMAXBIO, Seongnam, 13486, South Korea
| | - Chin-Feng Liu
- Continuing Education Program of Food Biotechnology Applications, National Taitung University, Taitung, Taiwan, ROC
| | - Tse-Han Lin
- Department of Life Science, National Taitung University, 369, Sec. 2, University Rd., Taitung, 95092, Taitung, Taiwan, ROC
| | | | - Tzu-Ming Pan
- SunWay Biotech Co. LTD., Taipei, Taiwan, ROC; Department of Biochemical Science and Technology, National Taiwan University, Taipei, Taiwan, ROC
| | - Chun-Lin Lee
- Continuing Education Program of Food Biotechnology Applications, National Taitung University, Taitung, Taiwan, ROC.
| | - Yean Kyoung Koo
- Department of R&I Center, COSMAXBIO, Seongnam, 13486, South Korea.
| |
Collapse
|
32
|
Moreira AMS, Nogueira JM, Carceroni J, Guadalupe JL, dos Santos AEA, Fagundes AMA, Copola AGL, Silva GAB, da Silva AB, Santos JPF, Albergaria JDS, Oliveira Andrade LD, Jorge EC. Acetate cellulose fibrous scaffold is suitable for cultivated fat production. Curr Res Food Sci 2024; 9:100903. [PMID: 39555016 PMCID: PMC11564054 DOI: 10.1016/j.crfs.2024.100903] [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: 08/28/2024] [Revised: 10/21/2024] [Accepted: 10/27/2024] [Indexed: 11/19/2024] Open
Abstract
Fat is an essential component of meat which contributes to its sensory characteristics. Therefore, producing cultivated fat is essential to replicate the texture, flavor, and juiciness of conventional meat. One of the challenges in obtaining cultivated fat is that once adipocytes reach differentiation in culture, they tend to float. In this study, we tested whether immortalized pre-adipocytes could be viable, grow, and differentiate when cultivated onto a fibrous scaffold produced by the electrospun of cellulose acetate. Our results demonstrated that the cells attach, proliferate, colonize, and differentiate into mature adipocytes in the three-dimensional fibrous structure during the culture period. Moreover, when layers of the scaffold containing differentiated cells were stacked, it acquired a characteristic similar to conventional animal fat. Therefore, this research suggests that fibrous scaffolds produced using cellulose acetate are a promising substrate for producing cultivated fat.
Collapse
Affiliation(s)
- Amanda Maria Siqueira Moreira
- Department of Morphology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Júlia Meireles Nogueira
- Department of Morphology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Jade Carceroni
- Department of Morphology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Jorge Luís Guadalupe
- Department of Morphology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Ana Elisa Antunes dos Santos
- Department of Morphology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Ana Maria Alvarenga Fagundes
- Department of Morphology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Aline Gonçalves Lio Copola
- Department of Morphology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | | - Aline Bruna da Silva
- Laboratory of Biomaterials, Department of Materials Engineering, Federal Center for Technological Education of Minas Gerais (CEFET-MG), Belo Horizonte, Brazil
| | - João Paulo Ferreira Santos
- Laboratory of Biomaterials, Department of Materials Engineering, Federal Center for Technological Education of Minas Gerais (CEFET-MG), Belo Horizonte, Brazil
| | - Juliano Douglas Silva Albergaria
- Laboratory of Biomaterials, Department of Materials Engineering, Federal Center for Technological Education of Minas Gerais (CEFET-MG), Belo Horizonte, Brazil
| | - Luciana de Oliveira Andrade
- Department of Morphology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Erika Cristina Jorge
- Department of Morphology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
33
|
Bugueno IM, Alastra G, Balic A, Stadlinger B, Mitsiadis TA. Limited Adipogenic Differentiation Potential of Human Dental Pulp Stem Cells Compared to Human Bone Marrow Stem Cells. Int J Mol Sci 2024; 25:11105. [PMID: 39456888 PMCID: PMC11508566 DOI: 10.3390/ijms252011105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 10/08/2024] [Accepted: 10/12/2024] [Indexed: 10/28/2024] Open
Abstract
Bone marrow and teeth contain mesenchymal stem cells (MSCs) that could be used for cell-based regenerative therapies. MSCs from these two tissues represent heterogeneous cell populations with varying degrees of lineage commitment. Although human bone marrow stem cells (hBMSCs) and human dental pulp stem cells (hDPSCs) have been extensively studied, it is not yet fully defined if their adipogenic potential differs. Therefore, in this study, we compared the in vitro adipogenic differentiation potential of hDPSCs and hBMSCs. Both cell populations were cultured in adipogenic differentiation media, followed by specific lipid droplet staining to visualise cytodifferentiation. The in vitro differentiation assays were complemented with the expression of specific genes for adipogenesis and osteogenesis-dentinogenesis, as well as for genes involved in the Wnt and Notch signalling pathways. Our findings showed that hBMSCs formed adipocytes containing numerous and large lipid vesicles. In contrast to hBMSCs, hDPSCs did not acquire the typical adipocyte morphology and formed fewer lipid droplets of small size. Regarding the gene expression, cultured hBMSCs upregulated the expression of adipogenic-specific genes (e.g., PPARγ2, LPL, ADIPONECTIN). Furthermore, in these cells most Wnt pathway genes were downregulated, while the expression of NOTCH pathway genes (e.g., NOTCH1, NOTCH3, JAGGED1, HES5, HEY2) was upregulated. hDPSCs retained their osteogenic/dentinogenic molecular profile (e.g., RUNX2, ALP, COLIA1) and upregulated the WNT-specific genes but not the NOTCH pathway genes. Taken together, our in vitro findings demonstrate that hDPSCs are not entirely committed to the adipogenic fate, in contrast to the hBMSCs, which are more effective to fully differentiate into adipocytes.
Collapse
Affiliation(s)
- Isaac Maximiliano Bugueno
- Orofacial Development and Regeneration, Institute of Oral Biology, Faculty of Medicine, Centre of Dental Medicine, University of Zurich, CH-8032 Zurich, Switzerland; (I.M.B.); (G.A.); (A.B.)
| | - Giuseppe Alastra
- Orofacial Development and Regeneration, Institute of Oral Biology, Faculty of Medicine, Centre of Dental Medicine, University of Zurich, CH-8032 Zurich, Switzerland; (I.M.B.); (G.A.); (A.B.)
- Department of Veterinary Medical Sciences, University of Bologna, 40126 Bologna, Italy
| | - Anamaria Balic
- Orofacial Development and Regeneration, Institute of Oral Biology, Faculty of Medicine, Centre of Dental Medicine, University of Zurich, CH-8032 Zurich, Switzerland; (I.M.B.); (G.A.); (A.B.)
| | - Bernd Stadlinger
- Clinic of Cranio-Maxillofacial and Oral Surgery, University of Zurich, CH-8032 Zurich, Switzerland;
| | - Thimios A. Mitsiadis
- Orofacial Development and Regeneration, Institute of Oral Biology, Faculty of Medicine, Centre of Dental Medicine, University of Zurich, CH-8032 Zurich, Switzerland; (I.M.B.); (G.A.); (A.B.)
| |
Collapse
|
34
|
Dania V, Stavropoulos NA, Gavriil P, Trikoupis I, Koulouvaris P, Savvidou OD, Mavrogenis AF, Papagelopoulos PJ. Treatment Modalities for Refractory-Recurrent Tenosynovial Giant Cell Tumor (TGCT): An Update. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1675. [PMID: 39459462 PMCID: PMC11509811 DOI: 10.3390/medicina60101675] [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: 09/13/2024] [Revised: 10/05/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024]
Abstract
Background and Objectives: Tenosynovial giant cell tumor (TGCT) is a rare, locally aggressive, benign neoplasm arising from the synovium of joints, tendon sheaths, and bursa. There are two main subtypes of TGCT: localized-type TGCT(L-TGCT) and diffuse-type TGCT (D-TGCT). While surgical excision is still considered the gold standard of treatment, the high recurrence rate, especially for D-TGCT, may suggest the need for other treatment modalities. Materials and Methods: This study reviews current literature on the current treatment modalities for refractory-relapsed TGCT disease. Results: The gold standard of treatment modality in TGCT remains surgical excision of the tumor nevertheless, the elevated recurrence rate and refractory disease, particularly in D-TGCT indicates and underscores the necessity for additional treatment alternatives. Conclusions: TGCT is a benign tumor with inflammatory features and a potential destructive and aggressive course that can lead to significant morbidity and functional impairment with a high impact on quality of life. Surgical resection remains the gold standard current treatment and the optimal surgical approach depends on the location and extent of the tumor. Systemic therapies have been recently used for relapsed mainly cases.
Collapse
Affiliation(s)
| | - Nikolaos A. Stavropoulos
- First Department of Orthopedic Surgery, School of Medicine, National and Kapodistrian University of Athens, “ATTIKON” University General Hospital, 12462 Athens, Greece
| | | | | | | | | | | | | |
Collapse
|
35
|
Ramlagan P, Rondeau P, Bourdon E, Bahorun T, Neergheen VS. Insulin Sensitivity of Adipocytes is Improved by Pomegranate Mesocarp Through Reduced Oxidative Stress and Inflammation. JOURNAL OF THE AMERICAN NUTRITION ASSOCIATION 2024; 43:592-603. [PMID: 38775816 DOI: 10.1080/27697061.2024.2353295] [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: 12/20/2023] [Revised: 02/26/2024] [Accepted: 05/04/2024] [Indexed: 09/25/2024]
Abstract
OBJECTIVE Inflammatory phenomena and increase in oxidative stress in cell physiopathology progression render therapeutic strategies based on nutritional antioxidants necessary. It was thus aimed at assessing the effectiveness of the pomegranate mesocarp extract (PME) on differentiation of preadipocytes to adipocytes in the presence/absence of hydrogen peroxide (H2O2), a model mimicking insulin resistance. METHOD The effect of PME on lipid accumulation, protein expression of antioxidant, inflammatory and adipogenic biomarkers, reactive oxygen species production, activity of antioxidant enzymes and secretion of IL-6 has been evaluated during the differentiation of preadipocytes to adipocytes, in the presence or absence of H2O2. RESULTS H2O2 reduced the expression of the regulator of insulin sensitivity PPARγ and suppressed adipocyte differentiation. PME counteracted the effect of H2O2. The latter induced a higher level of fat accumulation by promoting the expressions of the adipogenic markers PPARγ, C/EBPα, FABP4 and CD36 as compared to the control and the H2O2-treated differentiating cells. During the progression of adipogenesis, highest increase (p < 0.05) in IL-6 secretion, by 3.16 and 3.85 folds, was observed on day 2 of differentiation in control and H2O2-treated cells, respectively, compared to day 0. PME significantly decreased (p < 0.01) the secretion of the cytokine in addition to suppressing the expression of NFκB. PME also prevented the reduction of superoxide dismutase, catalase and glutathione peroxidase activities that occurred during adipogenesis, by at most 33%, 119% and 42%, respectively. CONCLUSION These findings indicate that PME efficiently improves insulin sensitivity and can significantly counteract oxidative stress and inflammation.
Collapse
Affiliation(s)
- Piteesha Ramlagan
- Biopharmaceutical Unit, Centre for Biomedical and Biomaterials Research, University of Mauritius, Réduit, Republic of Mauritius
- Department of Health Sciences, Faculty of Medicine and Health Sciences, University of Mauritius, Réduit, Republic of Mauritius
| | - Philippe Rondeau
- UMR 1188 Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, INSERM, Saint-Denis de La Réunion, France
| | - Emmanuel Bourdon
- UMR 1188 Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de La Réunion, INSERM, Saint-Denis de La Réunion, France
| | - Theeshan Bahorun
- Biopharmaceutical Unit, Centre for Biomedical and Biomaterials Research, University of Mauritius, Réduit, Republic of Mauritius
- Mauritius Research and Innovation Council, Ebène, Republic of Mauritius
| | - Vidushi S Neergheen
- Biopharmaceutical Unit, Centre for Biomedical and Biomaterials Research, University of Mauritius, Réduit, Republic of Mauritius
| |
Collapse
|
36
|
Takahashi A, Koike R, Watanabe S, Kuribayashi K, Wabitsch M, Miyamoto M, Komuro A, Seki M, Nashimoto M, Shimizu-Ibuka A, Yamashita K, Iwata T. Polypeptide N-acetylgalactosaminyltransferase-15 regulates adipogenesis in human SGBS cells. Sci Rep 2024; 14:20049. [PMID: 39209927 PMCID: PMC11362553 DOI: 10.1038/s41598-024-70930-5] [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] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024] Open
Abstract
Adipogenesis involves intricate molecular mechanisms regulated by various transcription factors and signaling pathways. In this study, we aimed to identify factors specifically induced during adipogenesis in the human preadipocyte cell line, SGBS, but not in the mouse preadipocyte cell line, 3T3-L1. Microarray analysis revealed distinct gene expression profiles, with 1460 genes induced in SGBS cells and 1297 genes induced in 3T3-L1 cells during adipogenesis, with only 297 genes commonly induced. Among the genes uniquely induced in SGBS cells, we focused on GALNT15, which encodes polypeptide N-acetylgalactosaminyltransferase-15. Its expression increased transiently during adipogenesis in SGBS cells but remained low in 3T3-L1 cells. Overexpression of GALNT15 increased mRNA levels of CCAAT-enhancer binding protein (C/EBPα) and leptin but had no significant impact on adipogenesis in SGBS cells. Conversely, knockdown of GALNT15 suppressed mRNA expression of adipocyte marker genes, reduced lipid accumulation, and decreased the percentage of cells with oil droplets. The induction of C/EBPα and peroxisome proliferator-activated receptor γ during adipogenesis was promoted or suppressed in SGBS cells subjected to overexpression or knockdown of GALNT15, respectively. These data suggest that polypeptide N-acetylgalactosaminyltransferase-15 is a novel regulatory molecule that enhances adipogenesis in SGBS cells.
Collapse
Affiliation(s)
- Asuka Takahashi
- Department of Functional Morphology, Graduate School of Pharmaceutical Sciences, Niigata University of Pharmacy and Medical and Life Sciences, Niigata, 956-8603, Japan
| | - Ryo Koike
- Department of Functional Morphology, Faculty of Pharmaceutical Sciences, Niigata University of Pharmacy and Medical and Life Sciences, 265-1 Higashijima, Akiha-ku, Niigata, 956-8603, Japan
| | - Shota Watanabe
- Department of Functional Morphology, Faculty of Pharmaceutical Sciences, Niigata University of Pharmacy and Medical and Life Sciences, 265-1 Higashijima, Akiha-ku, Niigata, 956-8603, Japan
| | - Kyoko Kuribayashi
- Department of Oral and Maxillofacial Surgery, Ehime University Graduate School of Medicine, Tōon, 791-0295, Japan
| | - Martin Wabitsch
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, 89075, Ulm, Germany
| | - Masahiko Miyamoto
- Department of Biochemistry, Faculty of Pharmaceutical Sciences, Niigata University of Pharmacy and Medical and Life Sciences, Niigata, 956-8603, Japan
| | - Akihiko Komuro
- Department of Biochemistry, Faculty of Pharmaceutical Sciences, Niigata University of Pharmacy and Medical and Life Sciences, Niigata, 956-8603, Japan
| | - Mineaki Seki
- Division of DNA Repair and Genome Integrity, Faculty of Medical Technology, Niigata University of Pharmacy and Medical and Life Sciences, Niigata, 956-8603, Japan
| | - Masayuki Nashimoto
- RNA Therapeutics Division, Faculty of Medical Technology, Niigata University of Pharmacy and Medical and Life Sciences, Niigata, 956-8603, Japan
| | | | - Kikuji Yamashita
- Division of Anatomy and Histology, Faculty of Medical Technology, Niigata University of Pharmacy and Medical and Life Sciences, Niigata, 956-8603, Japan
| | - Takeo Iwata
- Department of Functional Morphology, Graduate School of Pharmaceutical Sciences, Niigata University of Pharmacy and Medical and Life Sciences, Niigata, 956-8603, Japan.
- Department of Functional Morphology, Faculty of Pharmaceutical Sciences, Niigata University of Pharmacy and Medical and Life Sciences, 265-1 Higashijima, Akiha-ku, Niigata, 956-8603, Japan.
| |
Collapse
|
37
|
Okumuş EB, Böke ÖB, Turhan SŞ, Doğan A. From development to future prospects: The adipose tissue & adipose tissue organoids. Life Sci 2024; 351:122758. [PMID: 38823504 DOI: 10.1016/j.lfs.2024.122758] [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: 01/10/2024] [Revised: 05/22/2024] [Accepted: 05/27/2024] [Indexed: 06/03/2024]
Abstract
Living organisms store their energy in different forms of fats including lipid droplets, triacylglycerols, and steryl esters. In mammals and some non-mammal species, the energy is stored in adipose tissue which is the innervated specialized connective tissue that incorporates a variety of cell types such as macrophages, fibroblasts, pericytes, endothelial cells, adipocytes, blood cells, and several kinds of immune cells. Adipose tissue is so complex that the scope of its function is not only limited to energy storage, it also encompasses to thermogenesis, mechanical support, and immune defense. Since defects and complications in adipose tissue are heavily related to certain chronic diseases such as obesity, cardiovascular diseases, type 2 diabetes, insulin resistance, and cholesterol metabolism defects, it is important to further study adipose tissue to enlighten further mechanisms behind those diseases to develop possible therapeutic approaches. Adipose organoids are accepted as very promising tools for studying fat tissue development and its underlying molecular mechanisms, due to their high recapitulation of the adipose tissue in vitro. These organoids can be either derived using stromal vascular fractions or pluripotent stem cells. Due to their great vascularization capacity and previously reported incontrovertible regulatory role in insulin sensitivity and blood glucose levels, adipose organoids hold great potential to become an excellent candidate for the source of stem cell therapy. In this review, adipose tissue types and their corresponding developmental stages and functions, the importance of adipose organoids, and the potential they hold will be discussed in detail.
Collapse
Affiliation(s)
- Ezgi Bulut Okumuş
- Faculty of Engineering, Genetics and Bioengineering Department, Yeditepe University, İstanbul, Turkey
| | - Özüm Begüm Böke
- Faculty of Engineering, Genetics and Bioengineering Department, Yeditepe University, İstanbul, Turkey
| | - Selinay Şenkal Turhan
- Faculty of Engineering, Genetics and Bioengineering Department, Yeditepe University, İstanbul, Turkey
| | - Ayşegül Doğan
- Faculty of Engineering, Genetics and Bioengineering Department, Yeditepe University, İstanbul, Turkey.
| |
Collapse
|
38
|
Dowker-Key PD, Jadi PK, Gill NB, Hubbard KN, Elshaarrawi A, Alfatlawy ND, Bettaieb A. A Closer Look into White Adipose Tissue Biology and the Molecular Regulation of Stem Cell Commitment and Differentiation. Genes (Basel) 2024; 15:1017. [PMID: 39202377 PMCID: PMC11353785 DOI: 10.3390/genes15081017] [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: 06/24/2024] [Revised: 07/26/2024] [Accepted: 07/28/2024] [Indexed: 09/03/2024] Open
Abstract
White adipose tissue (WAT) makes up about 20-25% of total body mass in healthy individuals and is crucial for regulating various metabolic processes, including energy metabolism, endocrine function, immunity, and reproduction. In adipose tissue research, "adipogenesis" is commonly used to refer to the process of adipocyte formation, spanning from stem cell commitment to the development of mature, functional adipocytes. Although, this term should encompass a wide range of processes beyond commitment and differentiation, to also include other stages of adipose tissue development such as hypertrophy, hyperplasia, angiogenesis, macrophage infiltration, polarization, etc.… collectively, referred to herein as the adipogenic cycle. The term "differentiation", conversely, should only be used to refer to the process by which committed stem cells progress through distinct phases of subsequent differentiation. Recognizing this distinction is essential for accurately interpreting research findings on the mechanisms and stages of adipose tissue development and function. In this review, we focus on the molecular regulation of white adipose tissue development, from commitment to terminal differentiation, and examine key functional aspects of WAT that are crucial for normal physiology and systemic metabolic homeostasis.
Collapse
Affiliation(s)
- Presley D. Dowker-Key
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996-0840, USA
| | - Praveen Kumar Jadi
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996-0840, USA
| | - Nicholas B. Gill
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996-0840, USA
| | - Katelin N. Hubbard
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996-0840, USA
| | - Ahmed Elshaarrawi
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996-0840, USA
| | - Naba D. Alfatlawy
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996-0840, USA
| | - Ahmed Bettaieb
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996-0840, USA
- Graduate School of Genome Science and Technology, University of Tennessee, Knoxville, TN 37996-0840, USA
- Department of Biochemistry, Cellular and Molecular Biology, University of Tennessee, Knoxville, TN 37996-0840, USA
| |
Collapse
|
39
|
Choi JW, Park GH, Choi HJ, Lee JW, Kwon HY, Choi MY, Jeong JB. Anti‑obesity and immunostimulatory activity of Chrysosplenium flagelliferum in mouse preadipocytes 3T3‑L1 cells and mouse macrophage RAW264.7 cells. Exp Ther Med 2024; 28:315. [PMID: 38911047 PMCID: PMC11190883 DOI: 10.3892/etm.2024.12604] [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: 04/04/2024] [Accepted: 05/22/2024] [Indexed: 06/25/2024] Open
Abstract
Chrysosplenium flagelliferum (CF) is known for its anti-inflammatory, antioxidant and antibacterial activities. However, there is a lack of research on its other pharmacological properties. In the present study, the bifunctional roles of CF in 3T3-L1 and RAW264.7 cells were investigated, focusing on its anti-obesity and immunostimulatory effects. In 3T3-L1 cells, CF effectively mitigated the accumulation of lipid droplets and triacylglycerol. Additionally, CF downregulated the peroxisome proliferator-activated receptor (PPAR)-γ and CCAAT/enhancer-binding protein α protein levels; however, this effect was impeded by the knockdown of β-catenin using β-catenin-specific small interfering RNA. Consequently, CF-mediated inhibition of lipid accumulation was also decreased. CF increased the protein levels of adipose triglyceride lipase and phosphorylated hormone-sensitive lipase, while decreasing those of perilipin-1. Moreover, CF elevated the protein levels of phosphorylated AMP-activated protein kinase and PPARγ coactivator 1-α. In RAW264.7 cells, CF enhanced the production of pro-inflammatory mediators, such as nitric oxide (NO), inducible NO synthase, interleukin (IL)-1β, IL-6 and tumor necrosis factor-α, and increased their phagocytic capacities. Inhibition of Toll-like receptor (TLR)-4 significantly reduced the effects of CF on the production of pro-inflammatory mediators and phagocytosis, indicating its crucial role in facilitating these effects. CF-induced increase in the production of pro-inflammatory mediators was controlled by the activation of c-Jun N-terminal kinase (JNK) and nuclear factor (NF)-κB pathways, and TLR4 inhibition attenuated the phosphorylation of these kinases. The results of the pesent study suggested that CF inhibits lipid accumulation by suppressing adipogenesis and inducing lipolysis and thermogenesis in 3T3-L1 cells, while stimulating macrophage activation via the activation of JNK and NF-κB signaling pathways mediated by TLR4 in RAW264.7 cells. Therefore, CF simultaneously exerts both anti-obesity and immunostimulatory effects.
Collapse
Affiliation(s)
- Jeong Won Choi
- Department of Forest Science, Andong National University, Andong, Gyeongsangbuk 36729, Republic of Korea
| | - Gwang Hun Park
- Forest Medicinal Resources Research Center, National Institute of Forest Science, Yeongju, Gyeongsangbuk 36040, Republic of Korea
| | - Hyeok Jin Choi
- Department of Forest Science, Andong National University, Andong, Gyeongsangbuk 36729, Republic of Korea
| | - Jae Won Lee
- Department of Forest Science, Andong National University, Andong, Gyeongsangbuk 36729, Republic of Korea
| | - Hae-Yun Kwon
- Forest Medicinal Resources Research Center, National Institute of Forest Science, Yeongju, Gyeongsangbuk 36040, Republic of Korea
| | - Min Yeong Choi
- Forest Medicinal Resources Research Center, National Institute of Forest Science, Yeongju, Gyeongsangbuk 36040, Republic of Korea
| | - Jin Boo Jeong
- Department of Forest Science, Andong National University, Andong, Gyeongsangbuk 36729, Republic of Korea
| |
Collapse
|
40
|
Elsaid S, Wu X, Tee SS. Fructose vs. glucose: modulating stem cell growth and function through sugar supplementation. FEBS Open Bio 2024; 14:1277-1290. [PMID: 38923793 PMCID: PMC11301265 DOI: 10.1002/2211-5463.13846] [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: 01/29/2024] [Revised: 04/17/2024] [Accepted: 05/30/2024] [Indexed: 06/28/2024] Open
Abstract
In multicellular organisms, stem cells are impacted by microenvironmental resources such as nutrient availability and oxygen tension for their survival, growth, and differentiation. However, the accessibility of these resources in the pericellular environment greatly varies from organ to organ. This divergence in resource availability leads to variations in the potency and differentiation potential of stem cells. This study aimed to explore the distinct effects of glucose and fructose, as well as different oxygen tensions, on the growth dynamics, cytokine production, and differentiation of stem cells. We showed that replacing glucose with fructose subjected stem cells to stress, resulting in increased Hif1α expression and stability, which in turn led to a reduction in cell proliferation, and alterations in cytokine production. However, fructose failed to induce differentiation of human mesenchymal stem cells (hMSCs) as well as mouse fibroblasts into mature adipocytes compared to glucose, despite the upregulation of key markers of adipogenesis, including C/EBPβ, and PPARγ. Conversely, we showed that fructose induced undifferentiated mouse fibroblasts to release cytokines associated with senescence, including IL1α1, IL6, IL8, MCP1, and TNF1α, suggesting that these cells were undergoing lipolysis. Taken together, our results suggest that altering the culture conditions through changes in hexose levels and oxygen tension places considerable stress on stem cells. Additional research is required to further characterize the mechanisms governing stem cell response to their microenvironments.
Collapse
Affiliation(s)
- Salaheldeen Elsaid
- Department of Diagnostic Radiology and Nuclear MedicineUniversity of Maryland School of MedicineBaltimoreMDUSA
| | - Xiangdong Wu
- Department of Diagnostic Radiology and Nuclear MedicineUniversity of Maryland School of MedicineBaltimoreMDUSA
| | - Sui Seng Tee
- Department of Diagnostic Radiology and Nuclear MedicineUniversity of Maryland School of MedicineBaltimoreMDUSA
| |
Collapse
|
41
|
Beausoleil C, Thébault A, Andersson P, Cabaton NJ, Ermler S, Fromenty B, Garoche C, Griffin JL, Hoffmann S, Kamstra JH, Kubickova B, Lenters V, Kos VM, Poupin N, Remy S, Sapounidou M, Zalko D, Legler J, Jacobs MN, Rousselle C. Weight of evidence evaluation of the metabolism disrupting effects of triphenyl phosphate using an expert knowledge elicitation approach. Toxicol Appl Pharmacol 2024; 489:116995. [PMID: 38862081 DOI: 10.1016/j.taap.2024.116995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 06/05/2024] [Accepted: 06/07/2024] [Indexed: 06/13/2024]
Abstract
Identification of Endocrine-Disrupting Chemicals (EDCs) in a regulatory context requires a high level of evidence. However, lines of evidence (e.g. human, in vivo, in vitro or in silico) are heterogeneous and incomplete for quantifying evidence of the adverse effects and mechanisms involved. To date, for the regulatory appraisal of metabolism-disrupting chemicals (MDCs), no harmonised guidance to assess the weight of evidence has been developed at the EU or international level. To explore how to develop this, we applied a formal Expert Knowledge Elicitation (EKE) approach within the European GOLIATH project. EKE captures expert judgment in a quantitative manner and provides an estimate of uncertainty of the final opinion. As a proof of principle, we selected one suspected MDC -triphenyl phosphate (TPP) - based on its related adverse endpoints (obesity/adipogenicity) relevant to metabolic disruption and a putative Molecular Initiating Event (MIE): activation of peroxisome proliferator activated receptor gamma (PPARγ). We conducted a systematic literature review and assessed the quality of the lines of evidence with two independent groups of experts within GOLIATH, with the objective of categorising the metabolic disruption properties of TPP, by applying an EKE approach. Having followed the entire process separately, both groups arrived at the same conclusion, designating TPP as a "suspected MDC" with an overall quantitative agreement exceeding 85%, indicating robust reproducibility. The EKE method provides to be an important way to bring together scientists with diverse expertise and is recommended for future work in this area.
Collapse
Affiliation(s)
- Claire Beausoleil
- French Agency for Food, Environmental and Occupational Health and Safety (Anses), 94701 Maisons-Alfort, France.
| | - Anne Thébault
- French Agency for Food, Environmental and Occupational Health and Safety (Anses), 94701 Maisons-Alfort, France
| | | | - Nicolas J Cabaton
- INRAE. UMR1331 Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UT3, 31027 Toulouse, France
| | - Sibylle Ermler
- Department of Life Sciences, Centre of Genome Engineering and Maintenance, College of Health, Medicine and Life Sciences, Brunel University London, UB8 3PH Uxbridge, United Kingdom
| | - Bernard Fromenty
- INSERM, Univ Rennes, INRAE, Institut NUMECAN (Nutrition Metabolisms and Cancer) UMR_A 1341, UMR_S 1317, F-35000 Rennes, France
| | - Clémentine Garoche
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm U1194, Université Montpellier, Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Julian L Griffin
- The Rowett Institute, Foresterhill Health Campus, University of Aberdeen, Aberdeen, UK
| | | | - Jorke H Kamstra
- Institute for Risk Assessment Sciences, Department of Population Health Sciences, Utrecht University, Utrecht, the Netherlands
| | - Barbara Kubickova
- Radiation, Chemical and Environmental Hazards (RCE), Department of Toxicology, UK Health Security Agency (UKHSA), Harwell Science and Innovation Campus, Chilton OX11 0RQ, Oxon, United Kingdom
| | - Virissa Lenters
- Institute for Risk Assessment Sciences, Department of Population Health Sciences, Utrecht University, Utrecht, the Netherlands
| | - Vesna Munic Kos
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Nathalie Poupin
- INRAE. UMR1331 Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UT3, 31027 Toulouse, France
| | - Sylvie Remy
- Flemish Institute for Technological Research (VITO), 2400 Mol, Belgium
| | | | - Daniel Zalko
- INRAE. UMR1331 Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UT3, 31027 Toulouse, France
| | - Juliette Legler
- Institute for Risk Assessment Sciences, Department of Population Health Sciences, Utrecht University, Utrecht, the Netherlands
| | - Miriam N Jacobs
- Radiation, Chemical and Environmental Hazards (RCE), Department of Toxicology, UK Health Security Agency (UKHSA), Harwell Science and Innovation Campus, Chilton OX11 0RQ, Oxon, United Kingdom
| | - Christophe Rousselle
- French Agency for Food, Environmental and Occupational Health and Safety (Anses), 94701 Maisons-Alfort, France
| |
Collapse
|
42
|
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
|
43
|
Ramos CC, Pires J, Gonzalez E, Garcia-Vallicrosa C, Reis CA, Falcon-Perez JM, Freitas D. Extracellular vesicles in tumor-adipose tissue crosstalk: key drivers and therapeutic targets in cancer cachexia. EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2024; 5:371-396. [PMID: 39697630 PMCID: PMC11648493 DOI: 10.20517/evcna.2024.36] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 06/28/2024] [Accepted: 07/15/2024] [Indexed: 12/20/2024]
Abstract
Cancer cachexia is a complex metabolic syndrome characterized by unintentional loss of skeletal muscle and body fat. This syndrome is frequently associated with different types of cancer and negatively affects the prognosis and outcome of these patients. It involves a dynamic interplay between tumor cells and adipose tissue, where tumor-derived extracellular vesicles (EVs) play a crucial role in mediating intercellular communication. Tumor cells release EVs containing bioactive molecules such as hormones (adrenomedullin, PTHrP), pro-inflammatory cytokines (IL-6), and miRNAs (miR-1304-3p, miR-204-5p, miR-155, miR-425-3p, miR-146b-5p, miR-92a-3p), which can trigger lipolysis and induce the browning of white adipocytes contributing to a cancer cachexia phenotype. On the other hand, adipocyte-derived EVs can reprogram the metabolism of tumor cells by transporting fatty acids and enzymes involved in fatty acid oxidation, resulting in tumor growth and progression. These vesicles also carry leptin and key miRNAs (miR-155-5p, miR-10a-3p, miR-30a-3p, miR-32a/b, miR-21), thereby supporting tumor cell proliferation, metastasis formation, and therapy resistance. Understanding the intricate network underlying EV-mediated communication between tumor cells and adipocytes can provide critical insights into the mechanisms driving cancer cachexia. This review consolidates current knowledge on the crosstalk between tumor cells and adipose tissue mediated by EVs and offers valuable insights for future research. It also addresses controversial topics in the field and possible therapeutic approaches to manage cancer cachexia and ultimately improve patient outcomes and quality of life.
Collapse
Affiliation(s)
- Cátia C. Ramos
- i3S - Institute for Research and Innovation in Health, University of Porto, Porto 4200, Portugal
- IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, Porto 4200, Portugal
- Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto 4050, Portugal
| | - José Pires
- i3S - Institute for Research and Innovation in Health, University of Porto, Porto 4200, Portugal
- Faculty of Medicine, University of Porto (FMUP), Porto 4200, Portugal
| | | | | | - Celso A. Reis
- i3S - Institute for Research and Innovation in Health, University of Porto, Porto 4200, Portugal
- IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, Porto 4200, Portugal
- Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto 4050, Portugal
- Faculty of Medicine, University of Porto (FMUP), Porto 4200, Portugal
| | - Juan M. Falcon-Perez
- Exosomes Laboratory, CIC bioGUNE-BRTA, CIBERehd, Derio 48160, Spain
- IKERBASQUE Research Foundation, Bilbao 48009, Spain
| | - Daniela Freitas
- i3S - Institute for Research and Innovation in Health, University of Porto, Porto 4200, Portugal
- IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, Porto 4200, Portugal
| |
Collapse
|
44
|
Tan Q, Wellmerling JH, Song S, Dresler SR, Meridew JA, Choi KM, Li Y, Prakash Y, Tschumperlin DJ. Targeting CEBPA to restore cellular identity and tissue homeostasis in pulmonary fibrosis. JCI Insight 2024; 9:e175290. [PMID: 39012710 PMCID: PMC11343593 DOI: 10.1172/jci.insight.175290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 07/03/2024] [Indexed: 07/18/2024] Open
Abstract
Fibrosis in the lung is thought to be driven by epithelial cell dysfunction and aberrant cell-cell interactions. Unveiling the molecular mechanisms of cellular plasticity and cell-cell interactions is imperative to elucidating lung regenerative capacity and aberrant repair in pulmonary fibrosis. By mining publicly available RNA-Seq data sets, we identified loss of CCAAT enhancer-binding protein alpha (CEBPA) as a candidate contributor to idiopathic pulmonary fibrosis (IPF). We used conditional KO mice, scRNA-Seq, lung organoids, small-molecule inhibition, and potentially novel gene manipulation methods to investigate the role of CEBPA in lung fibrosis and repair. Long-term (6 months or more) of Cebpa loss in AT2 cells caused spontaneous fibrosis and increased susceptibility to bleomycin-induced fibrosis. Cebpa knockout (KO) in these mice significantly decreased AT2 cell numbers in the lung and reduced expression of surfactant homeostasis genes, while increasing inflammatory cell recruitment as well as upregulating S100a8/a9 in AT2 cells. In vivo treatment with an S100A8/A9 inhibitor alleviated experimental lung fibrosis. Restoring CEBPA expression in lung organoids ex vivo and during experimental lung fibrosis in vivo rescued CEBPA deficiency-mediated phenotypes. Our study establishes a direct mechanistic link between CEBPA repression, impaired AT2 cell identity, disrupted tissue homeostasis, and lung fibrosis.
Collapse
Affiliation(s)
- Qi Tan
- The Hormel Institute, University of Minnesota, Austin, Minnesota, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - Jack H. Wellmerling
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - Shengren Song
- The Hormel Institute, University of Minnesota, Austin, Minnesota, USA
| | - Sara R. Dresler
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - Jeffrey A. Meridew
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - Kyoung M. Choi
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - Yong Li
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - Y.S. Prakash
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Daniel J. Tschumperlin
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
45
|
Tan JTM, Price KJ, Fanshaw SR, Bilu C, Pham QT, Pham A, Sandeman L, Nankivell VA, Solly EL, Kronfeld-Schor N, Bursill CA. Exercise Reduces Glucose Intolerance, Cardiac Inflammation and Adipose Tissue Dysfunction in Psammomys obesus Exposed to Short Photoperiod and High Energy Diet. Int J Mol Sci 2024; 25:7756. [PMID: 39062999 PMCID: PMC11277119 DOI: 10.3390/ijms25147756] [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: 06/02/2024] [Revised: 07/09/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
Circadian disruption causes glucose intolerance, cardiac fibrosis, and adipocyte dysfunction in sand rats (Psammomys obesus). Exercise intervention can improve glucose metabolism, insulin sensitivity, adipose tissue function and protect against inflammation. We investigated the influence of exercise on male P. obesus exposed to a short photoperiod (5 h light:19 h dark) and high-energy diet. Exercise reduced glucose intolerance. Exercise reduced cardiac expression of inflammatory marker Ccl2 and Bax:Bcl2 apoptosis ratio. Exercise increased heart:body weight ratio and hypertrophy marker Myh7:Myh6, yet reduced Gata4 expression. No phenotypic changes were observed in perivascular fibrosis and myocyte area. Exercise reduced visceral adipose expression of inflammatory transcription factor Rela, adipogenesis marker Ppard and browning marker Ppargc1a, but visceral adipocyte size was unaffected. Conversely, exercise reduced subcutaneous adipocyte size but did not affect any molecular mediators. Exercise increased ZT7 Bmal1 and Per2 in the suprachiasmatic nucleus and subcutaneous Per2. Our study provides new molecular insights and histological assessments on the effect of exercise on cardiac inflammation, adipose tissue dysfunction and circadian gene expression in P. obesus exposed to short photoperiod and high-energy diet. These findings have implications for the protective benefits of exercise for shift workers in order to reduce the risk of diabetes and cardiovascular disease.
Collapse
Affiliation(s)
- Joanne T. M. Tan
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia; (K.J.P.); (S.-R.F.); (Q.T.P.); (A.P.); (L.S.); (V.A.N.); (E.L.S.)
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5005, Australia
| | - Kiara J. Price
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia; (K.J.P.); (S.-R.F.); (Q.T.P.); (A.P.); (L.S.); (V.A.N.); (E.L.S.)
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5005, Australia
| | - Sarah-Rose Fanshaw
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia; (K.J.P.); (S.-R.F.); (Q.T.P.); (A.P.); (L.S.); (V.A.N.); (E.L.S.)
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5005, Australia
| | - Carmel Bilu
- School of Zoology, Tel Aviv University, Tel Aviv 69978, Israel; (C.B.); (N.K.-S.)
| | - Quang Tuan Pham
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia; (K.J.P.); (S.-R.F.); (Q.T.P.); (A.P.); (L.S.); (V.A.N.); (E.L.S.)
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5005, Australia
| | - Anthony Pham
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia; (K.J.P.); (S.-R.F.); (Q.T.P.); (A.P.); (L.S.); (V.A.N.); (E.L.S.)
| | - Lauren Sandeman
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia; (K.J.P.); (S.-R.F.); (Q.T.P.); (A.P.); (L.S.); (V.A.N.); (E.L.S.)
| | - Victoria A. Nankivell
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia; (K.J.P.); (S.-R.F.); (Q.T.P.); (A.P.); (L.S.); (V.A.N.); (E.L.S.)
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5005, Australia
| | - Emma L. Solly
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia; (K.J.P.); (S.-R.F.); (Q.T.P.); (A.P.); (L.S.); (V.A.N.); (E.L.S.)
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5005, Australia
| | - Noga Kronfeld-Schor
- School of Zoology, Tel Aviv University, Tel Aviv 69978, Israel; (C.B.); (N.K.-S.)
| | - Christina A. Bursill
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia; (K.J.P.); (S.-R.F.); (Q.T.P.); (A.P.); (L.S.); (V.A.N.); (E.L.S.)
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5005, Australia
| |
Collapse
|
46
|
Kaimala S, Lootah SS, Mehra N, Kumar CA, Marzooqi SA, Sampath P, Ansari SA, Emerald BS. The Long Non-Coding RNA Obesity-Related (Obr) Contributes To Lipid Metabolism Through Epigenetic Regulation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401939. [PMID: 38704700 PMCID: PMC11234455 DOI: 10.1002/advs.202401939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Indexed: 05/07/2024]
Abstract
Obesity is a multifactorial disease that is part of today's epidemic and also increases the risk of other metabolic diseases. Long noncoding RNAs (lncRNAs) provide one tier of regulatory mechanisms to maintain metabolic homeostasis. Although lncRNAs are a significant constituent of the mammalian genome, studies aimed at their metabolic significance, including obesity, are only beginning to be addressed. Here, a developmentally regulated lncRNA, termed as obesity related (Obr), whose expression in metabolically relevant tissues such as skeletal muscle, liver, and pancreas is altered in diet-induced obesity, is identified. The Clone 9 cell line and high-fat diet-induced obese Wistar rats are used as a model system to verify the function of Obr. By using stable expression and antisense oligonucleotide-mediated downregulation of the expression of Obr followed by different molecular biology experiments, its role in lipid metabolism is verified. It is shown that Obr associates with the cAMP response element-binding protein (Creb) and activates different transcription factors involved in lipid metabolism. Its association with the Creb histone acetyltransferase complex, which includes the cAMP response element-binding protein (CBP) and p300, positively regulates the transcription of genes involved in lipid metabolism. In addition, Obr is regulated by Pparγ in response to lipid accumulation.
Collapse
Affiliation(s)
- Suneesh Kaimala
- Department of Anatomy, College of Medicine and Health Sciences, UAE University, Al Ain, P.O. Box 15551, UAE
| | - Shareena Saeed Lootah
- Department of Anatomy, College of Medicine and Health Sciences, UAE University, Al Ain, P.O. Box 15551, UAE
| | - Neha Mehra
- Department of Anatomy, College of Medicine and Health Sciences, UAE University, Al Ain, P.O. Box 15551, UAE
| | - Challagandla Anil Kumar
- Department of Anatomy, College of Medicine and Health Sciences, UAE University, Al Ain, P.O. Box 15551, UAE
| | - Saeeda Al Marzooqi
- Department of Pathology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, P.O. Box 15551, UAE
| | - Prabha Sampath
- A*STAR Skin Research Laboratory, Agency for Science Technology & Research (A*STAR), Singapore, 138648, Singapore
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore
- Genome Institute of Singapore, Agency for Science Technology & Research (A*STAR), Singapore, 138672, Singapore
| | - Suraiya Anjum Ansari
- Department of Biochemistry and Molecular Biology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, P.O. Box 15551, UAE
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, P.O. Box 15551, UAE
- ASPIRE Precision Medicine, Research Institute Abu Dhabi, Al Ain, Abu Dhabi, P.O. Box 15551, UAE
| | - Bright Starling Emerald
- Department of Anatomy, College of Medicine and Health Sciences, UAE University, Al Ain, P.O. Box 15551, UAE
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, P.O. Box 15551, UAE
- ASPIRE Precision Medicine, Research Institute Abu Dhabi, Al Ain, Abu Dhabi, P.O. Box 15551, UAE
| |
Collapse
|
47
|
Tan JTM, Cheney CV, Bamhare NES, Hossin T, Bilu C, Sandeman L, Nankivell VA, Solly EL, Kronfeld-Schor N, Bursill CA. Female Psammomys obesus Are Protected from Circadian Disruption-Induced Glucose Intolerance, Cardiac Fibrosis and Adipocyte Dysfunction. Int J Mol Sci 2024; 25:7265. [PMID: 39000372 PMCID: PMC11242371 DOI: 10.3390/ijms25137265] [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] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 06/27/2024] [Accepted: 06/28/2024] [Indexed: 07/16/2024] Open
Abstract
Circadian disruption increases the development of cardiovascular disease and diabetes. We found that circadian disruption causes glucose intolerance, cardiac fibrosis and adipocyte tissue dysfunction in male sand rats, Psammomys obesus. Whether these effects occur in female P. obesus is unknown. Male and female P. obesus were fed a high energy diet and exposed to a neutral (12 light:12 dark, control) or short (5 light:19 dark, circadian disruption) photoperiod for 20 weeks. Circadian disruption impaired glucose tolerance in males but not females. It also increased cardiac perivascular fibrosis and cardiac expression of inflammatory marker Ccl2 in males, with no effect in females. Females had reduced proapoptotic Bax mRNA and cardiac Myh7:Myh6 hypertrophy ratio. Cardiac protection in females occurred despite reductions in the clock gene Per2. Circadian disruption increased adipocyte hypertrophy in both males and females. This was concomitant with a reduction in adipocyte differentiation markers Pparg and Cebpa in males and females, respectively. Circadian disruption increased visceral adipose expression of inflammatory mediators Ccl2, Tgfb1 and Cd68 and reduced browning marker Ucp1 in males. However, these changes were not observed in females. Collectively, our study show that sex differentially influences the effects of circadian disruption on glucose tolerance, cardiac function and adipose tissue dysfunction.
Collapse
Affiliation(s)
- Joanne T M Tan
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5005, Australia
| | - Cate V Cheney
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5005, Australia
| | - Nicole E S Bamhare
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5005, Australia
| | - Tasnim Hossin
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5005, Australia
| | - Carmel Bilu
- School of Zoology, Tel Aviv University, Tel Aviv 69978, Israel
| | - Lauren Sandeman
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5005, Australia
| | - Victoria A Nankivell
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5005, Australia
| | - Emma L Solly
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5005, Australia
| | | | - Christina A Bursill
- Vascular Research Centre, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5005, Australia
| |
Collapse
|
48
|
Won YS, Bak SG, Chandimali N, Park EH, Lim HJ, Kwon HS, Park SI, Lee SJ. 7-MEGA™ inhibits adipogenesis in 3T3-L1 adipocytes and suppresses obesity in high-fat-diet-induced obese C57BL/6 mice. Lipids Health Dis 2024; 23:192. [PMID: 38909257 PMCID: PMC11193219 DOI: 10.1186/s12944-024-02175-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 06/04/2024] [Indexed: 06/24/2024] Open
Abstract
BACKGROUND Overweight, often known as obesity, is the abnormal and excessive accumulation of fat that exposes the health of a person at risk by increasing the likelihood that they may experience many chronic conditions. Consequently, obesity has become a global health threat, presenting serious health issues, and attracting a lot of attention in the healthcare profession and the scientific community. METHOD This study aims to explore the anti-adipogenic properties of 7-MEGA™ in an attempt to address obesity, using both in vitro and in vivo research. The effects of 7MEGA™ at three distinct concentrations were investigated in obese mice who were given a high-fat diet (HFD) and 3T3-L1 adipocytes. RESULTS 7MEGA™ decreased the total fat mass, overall body weight, and the perirenal and subcutaneous white adipose tissue (PWAT and SWAT) contents in HFD mice. Additionally, 7MEGA™ showed promise in improving the metabolic health of individuals with obesity and regulate the levels of insulin hormone, pro-inflammatory cytokines and adipokines. Furthermore, Peroxisome proliferator-activated receptors (PPAR) α and γ, Uncoupling Protein 1 (UCP-1), Sterol Regulatory Element-Binding Protein 1 (SREBP-1), Fatty Acid-Binding Protein 4 (FABP4), Fatty Acid Synthase (FAS), Acetyl-CoA Carboxylase (ACC), Stearoyl-CoA Desaturase-1 (SCD-1) and CCAAT/Enhancer-Binding Protein (C/EBPα) were among the adipogenic regulators that 7MEGA™ could regulate. CONCLUSION In summary, this study uncovered that 7MEGA™ demonstrates anti-adipogenic and anti-obesity effects, suggesting its potential in combating obesity.
Collapse
Affiliation(s)
- Yeong-Seon Won
- Division of Research Management, Department of Bioresource Industrialization, Honam National Institute of Biological Resource, Mokpo, 58762, Republic of Korea
| | - Seon-Gyeong Bak
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, 181 Ipsin-Gil, Jeongeup, 56212, Republic of Korea
| | - Nisansala Chandimali
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, 181 Ipsin-Gil, Jeongeup, 56212, Republic of Korea
- Department of Applied Biotechnology, University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Eun Hyun Park
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, 181 Ipsin-Gil, Jeongeup, 56212, Republic of Korea
- Department of Veterinary Pathology, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Hyung-Jin Lim
- Scripps Korea Antibody Institute, Chuncheon, 24341, Republic of Korea
| | - Hyuck Se Kwon
- R&D Team, Food & Supplement Health Claims, Vitech, Jeonju, 55365, Republic of Korea
| | - Sang-Ik Park
- Department of Veterinary Pathology, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, Republic of Korea.
| | - Seung Jae Lee
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, 181 Ipsin-Gil, Jeongeup, 56212, Republic of Korea.
- Department of Applied Biotechnology, University of Science and Technology (UST), Daejeon, 34113, Republic of Korea.
| |
Collapse
|
49
|
Park SY, Shin D, Yoon YS, Park S, Im SS, Kim Y, Kim YS, Choi C, Hur MW. TAT38 and TAT38 mimics potently inhibit adipogenesis by repressing C/EBPα, PPARγ, Pi-PPARγ, and SREBP1 expression. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2024; 1867:195030. [PMID: 38670485 DOI: 10.1016/j.bbagrm.2024.195030] [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: 02/01/2024] [Revised: 04/17/2024] [Accepted: 04/18/2024] [Indexed: 04/28/2024]
Abstract
Antiretroviral therapy-naive people living with HIV possess less fat than people without HIV. Previously, we found that HIV-1 transactivator of transcription (TAT) decreases fat in ob/ob mice. The TAT38 (a.a. 20-57) is important in the inhibition of adipogenesis and contains three functional domains: Cys-ZF domain (a.a. 20-35 TACTNCYCAKCCFQVC), core-domain (a.a. 36-46, FITKALGISYG), and protein transduction domain (PTD)(a.a. 47-57, RAKRRQRRR). Interestingly, the TAT38 region interacts with the Cyclin T1 of the P-TEFb complex, of which expression increases during adipogenesis. The X-ray crystallographic structure of the complex showed that the Cys-ZF and the core domain bind to the Cyclin T1 via hydrophobic interactions. To prepare TAT38 mimics with structural and functional similarities to TAT38, we replaced the core domain with a hydrophobic aliphatic amino acid (from carbon numbers 5 to 8). The TAT38 mimics with 6-hexanoic amino acid (TAT38 Ahx (C6)) and 7-heptanoic amino acid (TAT38 Ahp (C7)) inhibited adipogenesis of 3T3-L1 potently, reduced cellular triglyceride content, and decreased body weight of diet-induced obese (DIO) mice by 10.4-11 % in two weeks. The TAT38 and the TAT38 mimics potently repressed the adipogenic transcription factors genes, C/EBPα, PPARγ, and SREBP1. Also, they inhibit the phosphorylation of PPARγ. The TAT peptides may be promising candidates for development into a drug against obesity or diabetes.
Collapse
Affiliation(s)
- Sun-Young Park
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, 134, ShinChon-Dong, SeoDaeMoon-Ku, Seoul 120-752, Republic of Korea
| | - Dongyoon Shin
- Proteomics Research Team, CHA Institute of Future Medicine, Seongnam, Republic of Korea; Department of Medical Science, School of Medicine, CHA University, Seongnam, Republic of Korea
| | - Young So Yoon
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, 134, ShinChon-Dong, SeoDaeMoon-Ku, Seoul 120-752, Republic of Korea
| | - Sujin Park
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, 134, ShinChon-Dong, SeoDaeMoon-Ku, Seoul 120-752, Republic of Korea
| | - Seung-Soon Im
- Department of Physiology, Keimyung University School of Medicine, 1095 Dalgubeol-daero, Dalseo-gu, Daegu 42601, Republic of Korea
| | - Yeongshin Kim
- Proteomics Research Team, CHA Institute of Future Medicine, Seongnam, Republic of Korea; Department of Medical Science, School of Medicine, CHA University, Seongnam, Republic of Korea
| | - Young-Soo Kim
- Proteomics Research Team, CHA Institute of Future Medicine, Seongnam, Republic of Korea; Department of Medical Science, School of Medicine, CHA University, Seongnam, Republic of Korea
| | - CheolSoo Choi
- Korea Mouse Metabolic Phenotyping Center, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea; Division of Endocrinology & Metabolism, Department of Internal Medicine, Gil Medical Center, Gachon University College of Medicine, Incheon 21565, Republic of Korea
| | - Man-Wook Hur
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, 134, ShinChon-Dong, SeoDaeMoon-Ku, Seoul 120-752, Republic of Korea.
| |
Collapse
|
50
|
Zeng X, Wang TW, Yamaguchi K, Hatakeyama S, Yamazaki S, Shimizu E, Imoto S, Furukawa Y, Johmura Y, Nakanishi M. M2 macrophage-derived TGF-β induces age-associated loss of adipogenesis through progenitor cell senescence. Mol Metab 2024; 84:101943. [PMID: 38657734 PMCID: PMC11079528 DOI: 10.1016/j.molmet.2024.101943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 04/04/2024] [Accepted: 04/16/2024] [Indexed: 04/26/2024] Open
Abstract
OBJECTIVES Adipose tissue is an endocrine and energy storage organ composed of several different cell types, including mature adipocytes, stromal cells, endothelial cells, and a variety of immune cells. Adipose tissue aging contributes to the pathogenesis of metabolic dysfunction and is likely induced by crosstalk between adipose progenitor cells (APCs) and immune cells, but the underlying molecular mechanisms remain largely unknown. In this study, we revealed the biological role of p16high senescent APCs, and investigated the crosstalk between each cell type in the aged white adipose tissue. METHODS We performed the single-cell RNA sequencing (scRNA-seq) analysis on the p16high adipose cells sorted from aged p16-CreERT2/Rosa26-LSL-tdTomato mice. We also performed the time serial analysis on the age-dependent bulk RNA-seq datasets of human and mouse white adipose tissues to infer the transcriptome alteration of adipogenic potential within aging. RESULTS We show that M2 macrophage-derived TGF-β induces APCs senescence which impairs adipogenesis in vivo. p16high senescent APCs increase with age and show loss of adipogenic potential. The ligand-receptor interaction analysis reveals that M2 macrophages are the donors for TGF-β and the senescent APCs are the recipients. Indeed, treatment of APCs with TGF-β1 induces senescent phenotypes through mitochondrial ROS-mediated DNA damage in vitro. TGF-β1 injection into gonadal white adipose tissue (gWAT) suppresses adipogenic potential and induces fibrotic genes as well as p16 in APCs. A gWAT atrophy is observed in cancer cachexia by APCs senescence, whose induction appeared to be independent of TGF-β induction. CONCLUSIONS Our results suggest that M2 macrophage-derived TGF-β induces age-related lipodystrophy by APCs senescence. The TGF-β treatment induced DNA damage, mitochondrial ROS, and finally cellular senescence in APCs.
Collapse
Affiliation(s)
- Xinyi Zeng
- Division of Cancer Cell Biology, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Teh-Wei Wang
- Division of Cancer Cell Biology, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan.
| | - Kiyoshi Yamaguchi
- Division of Clinical Genome Research, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Seira Hatakeyama
- Division of Clinical Genome Research, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Satoshi Yamazaki
- Division of Stem Cell Biology, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Eigo Shimizu
- Division of Health Medical Intelligence, Human Genome Center, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Seiya Imoto
- Division of Health Medical Intelligence, Human Genome Center, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Yoichi Furukawa
- Division of Clinical Genome Research, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Yoshikazu Johmura
- Division of Cancer and Senescence Biology, Cancer Research Institute, Institute for Frontier Science Initiative, Kanazawa University, Kanazawa, Japan
| | - Makoto Nakanishi
- Division of Cancer Cell Biology, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan.
| |
Collapse
|