1
|
Oh JM, Kim G, Jeong J, Chun S. Compound K promotes thermogenic signature and mitochondrial biogenesis via the UCP1-SIRT3-PGC1α signaling pathway. Biomed Pharmacother 2025; 183:117838. [PMID: 39799670 DOI: 10.1016/j.biopha.2025.117838] [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: 11/08/2024] [Revised: 12/28/2024] [Accepted: 01/09/2025] [Indexed: 01/15/2025] Open
Abstract
Compound K (CK), an active ingredient in ginseng, has anti-cancer, anti-inflammatory, and antioxidant properties. However, its effects on thermogenesis and mitochondrial dynamics in white adipose tissue (WAT) adipocytes are not well understood. This study explores CK's impact on thermogenesis and mitochondrial metabolism in cold-exposed mice and mouse stromal vascular fraction (SVF) cells. CK increased the expression of UCP1 and other brown/beige adipocyte markers (Cd137, Cytb, Letm1, Pgc1α, Prdm16, Tbp1, Tbx1, Uqcrc1) and mitochondrial biogenesis/dynamics factors (Cidea, Cox8b, Cycs, Dio2, Drp1, Fis1, Fgf21, Nrf1, Sirt3, Tfam) in 3T3-L1/iWAT SVF cells. CK enhanced mitochondrial respiration, reduced mitochondrial ROS levels, and restored MMP in iWAT SVF cells, leading to the differentiation of WAT into beige adipocytes, and that was also observed in cold-exposed subcutaneous tissue. CK administration to cold-exposed mice reduced fat droplet size and increased the number of mitochondria. Additionally, CK stimulated non-shivering thermogenesis, indicated by the upregulation of thermogenic and mitochondrial division proteins. The browning effect of CK was nullified by SIRT3 knockdown, suggesting that CK induces beige remodeling of WAT by regulating mitochondrial dynamics and SIRT3 expression. These findings suggest CK's potential as a therapeutic agent for obesity and metabolic disorders that promotes the transformation of WAT into a metabolically active beige phenotype.
Collapse
Affiliation(s)
- Jung-Mi Oh
- Department of Physiology, Institute for Medical Sciences, Jeonbuk National University Medical School, Jeonju, Jeollabuk-do 54907, South Korea
| | - Geonhyeong Kim
- Department of Orthopaedic Surgery, Seogwipo Medical Center, Seogwipo-si, Jeju-do 63585, South Korea
| | - Jiho Jeong
- Department of Orthopaedic Surgery, Seogwipo Medical Center, Seogwipo-si, Jeju-do 63585, South Korea
| | - Sungkun Chun
- Department of Physiology, Institute for Medical Sciences, Jeonbuk National University Medical School, Jeonju, Jeollabuk-do 54907, South Korea.
| |
Collapse
|
2
|
Miro C, Menale C, Acampora L, Nappi A, Sagliocchi S, Restolfer F, Torabinejad S, Stornaiuolo M, Dentice M, Cicatiello AG. Muscle PGC-1α Overexpression Drives Metabolite Secretion Boosting Subcutaneous Adipocyte Browning. J Cell Physiol 2025; 240:e31480. [PMID: 39676331 PMCID: PMC11733859 DOI: 10.1002/jcp.31480] [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: 08/08/2024] [Revised: 10/17/2024] [Accepted: 10/24/2024] [Indexed: 12/17/2024]
Abstract
Muscle and adipose tissue (AT) are in mutual interaction through the integration of endocrine and biochemical signals, thus regulating whole-body function and physiology. Besides a traditional view of endocrine relationships that imply the release of cytokines and growth factors, it is becoming increasingly clear that a metabolic network involving metabolites as signal molecules also exists between the two tissues. By elevating the number and functionality of mitochondria, a key role in muscle metabolism is played by the master regulator of mitochondrial biogenesis peroxisome-proliferator-activated receptor-γ coactivator-1α (PGC-1α), that induces a fiber type shift from glycolytic to oxidative myofibers. As a consequence, the upregulation of muscle respiratory rate might affect metabolite production and consumption. However, the underlying mechanisms have not yet been fully elucidated. Here, we used a muscle-specific PGC-1α overexpressing mouse model (MCK-PGC-1α) to analyze the metabolite secretion profile of serum and culture medium recovered from MCK-PGC-1α muscle fibers by NMR. We revealed modified levels of different metabolites that might be ascribed to the metabolic activation of the skeletal muscle fibers. Notably, the dysregulated levels of these metabolites affected adipocyte differentiation, as well as the browning process in vitro and in vivo. Interestingly such effect was exacerbated in the subcutaneous WAT, while only barely present in the visceral WAT. Our data confirm a prominent role of PGC-1α as a trigger of mitochondrial function in skeletal muscle and propose a novel function of this master regulator gene in modulating the metabolite production in turn affecting the activation of WAT and its conversion toward the browning.
Collapse
Affiliation(s)
- Caterina Miro
- Department of Clinical Medicine and SurgeryUniversity of Naples “Federico II”NaplesItaly
| | - Ciro Menale
- Department of Clinical Medicine and SurgeryUniversity of Naples “Federico II”NaplesItaly
| | - Lucia Acampora
- Department of Clinical Medicine and SurgeryUniversity of Naples “Federico II”NaplesItaly
| | - Annarita Nappi
- Department of Clinical Medicine and SurgeryUniversity of Naples “Federico II”NaplesItaly
| | - Serena Sagliocchi
- Department of Clinical Medicine and SurgeryUniversity of Naples “Federico II”NaplesItaly
| | - Federica Restolfer
- Department of Clinical Medicine and SurgeryUniversity of Naples “Federico II”NaplesItaly
| | - Sepehr Torabinejad
- Department of Clinical Medicine and SurgeryUniversity of Naples “Federico II”NaplesItaly
| | | | - Monica Dentice
- Department of Clinical Medicine and SurgeryUniversity of Naples “Federico II”NaplesItaly
- CEINGE‐Biotecnologie Avanzate Franco SalvatoreNaplesItaly
| | | |
Collapse
|
3
|
Cortellino S, D'Angelo M, Quintiliani M, Giordano A. Cancer knocks you out by fasting: Cachexia as a consequence of metabolic alterations in cancer. J Cell Physiol 2025; 240:e31417. [PMID: 39245862 DOI: 10.1002/jcp.31417] [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: 04/28/2024] [Revised: 07/18/2024] [Accepted: 08/09/2024] [Indexed: 09/10/2024]
Abstract
Neoplastic transformation reprograms tumor and surrounding host cell metabolism, increasing nutrient consumption and depletion in the tumor microenvironment. Tumors uptake nutrients from neighboring normal tissues or the bloodstream to meet energy and anabolic demands. Tumor-induced chronic inflammation, a high-energy process, also consumes nutrients to sustain its dysfunctional activities. These tumor-related metabolic and physiological changes, including chronic inflammation, negatively impact systemic metabolism and physiology. Furthermore, the adverse effects of antitumor therapy and tumor obstruction impair the endocrine, neural, and gastrointestinal systems, thereby confounding the systemic status of patients. These alterations result in decreased appetite, impaired nutrient absorption, inflammation, and shift from anabolic to catabolic metabolism. Consequently, cancer patients often suffer from malnutrition, which worsens prognosis and increases susceptibility to secondary adverse events. This review explores how neoplastic transformation affects tumor and microenvironment metabolism and inflammation, leading to poor prognosis, and discusses potential strategies and clinical interventions to improve patient outcomes.
Collapse
Affiliation(s)
- Salvatore Cortellino
- Laboratory of Molecular Oncology, Responsible Research Hospital, Campobasso, Italy
- Scuola Superiore Meridionale (SSM), School for Advanced Studies, Federico II University, Naples, Italy
- SHRO Italia Foundation ETS, Candiolo, Turin, Italy
| | - Margherita D'Angelo
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | | | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania, USA
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| |
Collapse
|
4
|
Sulaiman U, Vaughan R, Siegel P, Liu D, Gilbert E, Cline M. Embryonic Thermal Programming and Dietary Baicalein Supplementation Post-Hatch: Effects on Broiler Adipose Tissue Deposition. Animals (Basel) 2024; 14:3563. [PMID: 39765466 PMCID: PMC11672455 DOI: 10.3390/ani14243563] [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: 10/16/2024] [Revised: 11/25/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
Optimization of growth performance and fat metabolism in broilers are critical for meat quality and overall production efficiency. This experiment investigated the effects of dietary baicalein supplementation and embryonic heat conditioning (EHC) on the growth performance and adipose tissue metabolism of 10-day old broilers. Fertile eggs were divided into control and EHC groups, with EHC eggs exposed to intermittent heating (39.5 °C) from day 7 to day 16 of incubation. Hatched chicks were further divided into four groups: CC (control control), CT (control treatment with baicalein), EC (embryonic heat control), and ET (embryonic heat treatment with baicalein), and were fed ad libitum. On day 10 post-hatch, blood and adipose tissue samples were collected for analysis. C/EBPα mRNA was lower in the ET group compared to the EC group and higher in the CT group compared to the CC group. PPARγ and HSL mRNAs were elevated in both the ET and CT groups relative to their controls. Additionally, plasma non-esterified fatty acid (NEFA) levels were significantly higher in the CT group compared to the CC group. These results indicate that baicalein supplementation, particularly when combined with embryonic heat conditioning, can modulate fat metabolism and potentially improve the growth performance of broilers, thereby offering insights into strategies for enhancing poultry production.
Collapse
Affiliation(s)
- Usman Sulaiman
- School of Animal Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA; (U.S.); (P.S.)
| | - Reagan Vaughan
- Department of Human Nutrition, Foods and Exercise, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA; (R.V.); (D.L.)
| | - Paul Siegel
- School of Animal Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA; (U.S.); (P.S.)
| | - Dongmin Liu
- Department of Human Nutrition, Foods and Exercise, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA; (R.V.); (D.L.)
| | - Elizabeth Gilbert
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA;
| | - Mark Cline
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA;
| |
Collapse
|
5
|
Ogawa M, Oshiro H, Tamura Y, Ishido M, Okamoto T, Hata J. Characteristics of T2* and anisotropy parameters in inguinal and epididymal adipose tissues after cold exposure in mice. Sci Rep 2024; 14:29491. [PMID: 39604392 PMCID: PMC11603128 DOI: 10.1038/s41598-024-78655-1] [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: 01/18/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024] Open
Abstract
White adipose tissue (WAT) in mice undergoes browning in response to cold exposure. Brown and beige adipocytes contain multilocular lipid droplets and abundant iron-containing mitochondria expressing uncoupling protein 1 (UCP-1). Cold exposure-induced browning WAT is accompanied by increased density of blood vessels and sympathetic nerve fibres. A previous study reported a more than threefold increase in sympathetic nerve dendritic tone in inguinal white adipose tissue (iWAT) after cold exposure. Therefore, we hypothesized that water molecule diffusion would be more restricted in brown and beige adipocytes compared to white adipocytes. The characteristics of T2* values and anisotropy parameters by diffusion tensor imaging (DTI) in browning WAT are unclear. The aim of the present study was to investigate the effect of cold exposure on T2* values and anisotropy parameters (fractional anisotropy [FA], apparent diffusion coefficient [ADC], radial diffusivity [RD] and eigenvalues λ1, λ2, λ3) in brown adipose tissue (BAT), iWAT and epididymal white adipose tissue (epiWAT). Furthermore, these parameters were investigated in vivo through additional validation experiments in three control mice. Mice in the cold exposure (CE) group were exposed to a cold environment at 4 °C for 10 days, while these in the control (C) group were maintained at 22 °C throughout the experiment. T2* values, FA, ADC, RD and eigenvalues (λ1, λ2, λ3) were measured in BAT, iWAT and epiWAT using a 9.4T magnetic resonance scanner (Bruker Biospin AG). T2* values of epiWAT in the C group were significantly higher than these of BAT in the C group and iWAT in the CE group. No significant differences were observed between groups for FA, ADC, RD, λ1 and λ2 of iWAT and epiWAT. However, the λ3 values of iWAT and epiWAT in the CE group were significantly higher than these of iWAT, epiWAT and BAT in the C group. Compared to ex vivo measurements, in vivo measurements in control mice showed higher T2* values with reduced intertissue variability while maintaining tissue-specific patterns. These results suggest that T2* values and anisotropy parameters might serve as potential markers for the assessment of adipose tissue plasticity. Further studies are required to investigate their utility as non-invasive indicators of browning WAT.
Collapse
Affiliation(s)
- Madoka Ogawa
- Institute for Liberal Arts, Environment and Society, Institute of Science Tokyo, 2-12-1 Ookayama, Meguro-ku, Tokyo, 152-8550, Japan.
- Nippon Sport Science University, Tokyo, Japan.
| | - Hinako Oshiro
- Graduate School of Human Health Science, Tokyo Metropolitan University, Tokyo, Japan
- Center for Brain Science, RIKEN, Saitama, Japan
| | - Yuki Tamura
- Nippon Sport Science University, Tokyo, Japan
| | | | | | - Junichi Hata
- Graduate School of Human Health Science, Tokyo Metropolitan University, Tokyo, Japan
- Center for Brain Science, RIKEN, Saitama, Japan
| |
Collapse
|
6
|
Kim S, Park SG, Kim J, Hong S, Cho SM, Lim SY, Kim EK, Ju S, Lee SB, Kim SP, Jeong TY, Oh Y, Han S, Kim HR, Lee TC, Kim HC, Yoon WK, An TH, Oh KJ, Nam KH, Lee S, Kim K, Seong JK, Lee H. Comprehensive phenotypic assessment of nonsense mutations in mitochondrial ND5 in mice. Exp Mol Med 2024; 56:2395-2408. [PMID: 39482535 PMCID: PMC11612467 DOI: 10.1038/s12276-024-01333-9] [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: 01/04/2024] [Revised: 07/07/2024] [Accepted: 07/30/2024] [Indexed: 11/03/2024] Open
Abstract
Mitochondrial dysfunction induced by mitochondrial DNA (mtDNA) mutations has been implicated in various human diseases. A comprehensive analysis of mitochondrial genetic disorders requires suitable animal models for human disease studies. While gene knockout via premature stop codons is a powerful method for investigating the unique functions of target genes, achieving knockout of mtDNA has been rare. Here, we report the genotypes and phenotypes of heteroplasmic MT-ND5 gene-knockout mice. These mutant mice presented damaged mitochondrial cristae in the cerebral cortex, hippocampal atrophy, and asymmetry, leading to learning and memory abnormalities. Moreover, mutant mice are susceptible to obesity and thermogenetic disorders. We propose that these mtDNA gene-knockdown mice could serve as valuable animal models for studying the MT-ND5 gene and developing therapies for human mitochondrial disorders in the future.
Collapse
Affiliation(s)
- Sanghun Kim
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Seul Gi Park
- Laboratory Animal Resource and Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, 28116, Republic of Korea
| | - Jieun Kim
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Seongho Hong
- Laboratory of Developmental Biology and Genomics, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
- Korea Model animal Priority Center, Seoul National University, Seoul, 08826, Republic of Korea
| | - Sang-Mi Cho
- Laboratory Animal Resource and Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, 28116, Republic of Korea
| | - Soo-Yeon Lim
- Korea Model animal Priority Center, Seoul National University, Seoul, 08826, Republic of Korea
| | - Eun-Kyoung Kim
- Laboratory Animal Resource and Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, 28116, Republic of Korea
| | - Sungjin Ju
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, 02841, Republic of Korea
- Department of Physiology, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Su Bin Lee
- Korea Model animal Priority Center, Seoul National University, Seoul, 08826, Republic of Korea
| | - Sol Pin Kim
- Laboratory of Developmental Biology and Genomics, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
- Korea Model animal Priority Center, Seoul National University, Seoul, 08826, Republic of Korea
| | - Tae Young Jeong
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, 02841, Republic of Korea
- Department of Physiology, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Yeji Oh
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Seunghun Han
- Laboratory Animal Resource and Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, 28116, Republic of Korea
| | - Hae-Rim Kim
- Laboratory Animal Resource and Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, 28116, Republic of Korea
| | - Taek Chang Lee
- Laboratory Animal Resource and Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, 28116, Republic of Korea
| | - Hyoung-Chin Kim
- Laboratory Animal Resource and Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, 28116, Republic of Korea
| | - Won Kee Yoon
- Laboratory Animal Resource and Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, 28116, Republic of Korea
| | - Tae Hyeon An
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34141, Republic of Korea
| | - Kyoung-Jin Oh
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, 34141, Republic of Korea
| | - Ki-Hoan Nam
- Laboratory Animal Resource and Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, 28116, Republic of Korea.
| | - Seonghyun Lee
- Department of MetaBioHealth, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea.
- Department of Precision Medicine, School of Medicine, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea.
| | - Kyoungmi Kim
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, 02841, Republic of Korea.
- Department of Physiology, Korea University College of Medicine, Seoul, 02841, Republic of Korea.
| | - Je Kyung Seong
- Laboratory of Developmental Biology and Genomics, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea.
- Korea Model animal Priority Center, Seoul National University, Seoul, 08826, Republic of Korea.
| | - Hyunji Lee
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, 02841, Republic of Korea.
- Laboratory Animal Resource and Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, 28116, Republic of Korea.
- Department of Convergence Medicine, Korea University College of Medicine, Seoul, 02708, Republic of Korea.
| |
Collapse
|
7
|
Stephens CJ, Kobayashi R, Berry DC, Butcher JT. The Role of Matrix Stiffness And Viscosity on Lipid Phenotype And Fat Lineage Potential. Tissue Eng Part A 2024. [PMID: 39165245 DOI: 10.1089/ten.tea.2024.0149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/22/2024] Open
Abstract
Autologous fat transfer is a common procedure that patients undergo to rejuvenate large soft tissue defects. However, these surgeries are complicated by limited tissue sources, donor-site morbidity, and necrosis. While the biofabrication of fat tissue can serve as a clinical option for reconstructive surgery, the influence of matrix mechanics, specifically stiffness and viscosity, on adipogenesis requires further elucidation. Additionally, the effects of these mechanical parameters on metabolic and thermogenic fat potential have yet to be investigated. In this study, gelatin methacryloyl (GelMA) polymers with varying degrees of methacrylation (DoM) were fabricated to create matrices with different stiffnesses and viscosities. Human adipose-derived mesenchymal stem cells were then encapsulated in mechanically tunable GelMA and underwent adipogenesis to investigate the effects of matrix mechanics on lipid phenotype and fat potential. Mechanical testing confirmed that GelMA stiffness was regulated by DoM and weight composition, whereas viscosity was determined by the latter. Further work revealed that while lipid phenotype became more enriched as matrix stiffness and viscosity declined, the potential toward metabolic and thermogenic fat appeared to be more viscous dependent rather than stiffness dependent. In addition, fatty acid binding protein 4 and uncoupling protein 1 gene expression exhibited viscous-dependent behavior despite comparable levels of peroxisome proliferator-activated receptor gamma. However, despite the superior role of viscosity, lipid quantity and mitochondrial abundance demonstrated stiffness-dependent behavior. Overall, this work revealed that matrix viscosity played a more superior role than stiffness in driving adipogenesis and distinguishing between metabolic and thermogenic fat potential. Ultimately, this differentiation in fat production is important for engineering ideal adipose tissue for large soft tissue defects.
Collapse
Affiliation(s)
- Chelsea J Stephens
- Cornell University, Nancy E. and Peter C. Meinig School of Biomedical Engineering, Ithaca, New York, USA
| | - Reina Kobayashi
- Cornell University, Nancy E. and Peter C. Meinig School of Biomedical Engineering, Ithaca, New York, USA
| | - Daniel C Berry
- College of Human Ecology, Division of Nutrition Science, Cornell University, Ithaca, New York, USA
| | - Jonathan T Butcher
- Cornell University, Nancy E. and Peter C. Meinig School of Biomedical Engineering, Ithaca, New York, USA
| |
Collapse
|
8
|
Hu Y, Huang Y, Jiang Y, Weng L, Cai Z, He B. The Different Shades of Thermogenic Adipose Tissue. Curr Obes Rep 2024; 13:440-460. [PMID: 38607478 DOI: 10.1007/s13679-024-00559-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/12/2024] [Indexed: 04/13/2024]
Abstract
PURPOSE OF REVIEW By providing a concise overview of adipose tissue types, elucidating the regulation of adipose thermogenic capacity in both physiological contexts and chronic wasting diseases (a protracted hypermetabolic state that precipitates sustained catabolism and consequent progressive corporeal atrophy), and most importantly, delving into the ongoing discourse regarding the role of adipose tissue thermogenic activation in chronic wasting diseases, this review aims to provide researchers with a comprehensive understanding of the field. RECENT FINDINGS Adipose tissue, traditionally classified as white, brown, and beige (brite) based on its thermogenic activity and potential, is intricately regulated by complex mechanisms in response to exercise or cold exposure. This regulation is adipose depot-specific and dependent on the duration of exposure. Excessive thermogenic activation of adipose tissue has been observed in chronic wasting diseases and has been considered a pathological factor that accelerates disease progression. However, this conclusion may be confounded by the detrimental effects of excessive lipolysis. Recent research also suggests that such activation may play a beneficial role in the early stages of chronic wasting disease and provide potential therapeutic effects. A more comprehensive understanding of the changes in adipose tissue thermogenesis under physiological and pathological conditions, as well as the underlying regulatory mechanisms, is essential for the development of novel interventions to improve health and prevent disease.
Collapse
Affiliation(s)
- Yunwen Hu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Yijie Huang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Yangjing Jiang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Lvkan Weng
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| | - Zhaohua Cai
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| | - Ben He
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| |
Collapse
|
9
|
Lee K, Kim HJ, Kim JY, Shim JJ, Lee JH. A Mixture of Lactobacillus HY7601 and KY1032 Regulates Energy Metabolism in Adipose Tissue and Improves Cholesterol Disposal in High-Fat-Diet-Fed Mice. Nutrients 2024; 16:2570. [PMID: 39125449 PMCID: PMC11314552 DOI: 10.3390/nu16152570] [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: 07/05/2024] [Revised: 07/30/2024] [Accepted: 08/03/2024] [Indexed: 08/12/2024] Open
Abstract
We aimed to characterize the anti-obesity and anti-atherosclerosis effects of Lactobacillus curvatus HY7601 and Lactobacillus plantarum KY1032 using high-fat diet (HFD)-fed obese C57BL/6 mice. We divided the mice into control (CON), HFD, HFD with 108 CFU/kg/day probiotics (HFD + KL, HY7301:KY1032 = 1:1), and HFD with 109 CFU/kg/day probiotics (HFD + KH, HY7301:KY1032 = 1:1) groups and fed/treated them during 7 weeks. The body mass, brown adipose tissue (BAT), inguinal white adipose tissue (iWAT), and epididymal white adipose tissue (eWAT) masses and the total cholesterol and triglyceride concentrations were remarkably lower in probiotic-treated groups than in the HFD group in a dose-dependent manner. In addition, the expression of uncoupling protein 1 in the BAT, iWAT, and eWAT was significantly higher in probiotic-treated HFD mice than in the HFD mice, as demonstrated by immunofluorescence staining and Western blotting. We also measured the expression of cholesterol transport genes in the liver and jejunum and found that the expression of those encoding liver-X-receptor α, ATP-binding cassette transporters G5 and G8, and cholesterol 7α-hydroxylase were significantly higher in the HFD + KH mice than in the HFD mice. Thus, a Lactobacillus HY7601 and KY1032 mixture with 109 CFU/kg/day concentration can assist with body weight regulation through the management of lipid metabolism and thermogenesis.
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.); (J.-H.L.)
| | | | | |
Collapse
|
10
|
Chen S, Yuan W, Huang Q, Xiong X, Wang C, Zeng W, Wang L, Huang Y, Liu Y, Wang Y, Huang Q. Asprosin contributes to pathogenesis of obesity by adipocyte mitophagy induction to inhibit white adipose browning in mice. Int J Obes (Lond) 2024; 48:913-922. [PMID: 38374247 DOI: 10.1038/s41366-024-01495-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 01/25/2024] [Accepted: 02/05/2024] [Indexed: 02/21/2024]
Abstract
BACKGROUND Asprosin (ASP) is a newly discovered adipokine secreted by white adipose tissue (WAT), which can regulate the homeostasis of glucose and lipid metabolism. However, it is not clear whether it can regulate the browning of WAT and mitophagy during the browning process. Accordingly, this study aims to investigate the effects and possible mechanisms of ASP on the browning of WAT and mitophagy in vivo and in vitro. METHODS In in vivo experiments, some mouse models were used including adipose tissue ASP-specific deficiency (ASP-/-), high fat diet (HFD)-induced obesity and white adipose browning; in in vitro experiments, some cell models were also established and used, including ASP-deficient 3T3-L1 preadipocyte (ASP-/-) and CL-316243 (CL, 1 µM)-induced browning. Based on these models, the browning of WAT and mitophagy were evaluated by morphology, functionality and molecular markers. RESULTS Our in vivo data show that adipose tissue-specific deletion of ASP contributes to weight loss in mice; supplementation of ASP inhibits the expressions of browning-related proteins including UCP1, PRDM16 and PGC1ɑ during the cold exposure-induced browning, and promotes the expressions of mitophagy-related proteins including PINK1 and Parkin under the conditions of whether normal diet (ND) or HFD. Similarly, our in vitro data also show that the deletion of ASP in 3T3-L1 cells significantly increases the expressions of the browning-related proteins and decreases the expressions of the mitophagy-related proteins. CONCLUSIONS These data demonstrate that ASP deletion can facilitate the browning and inhibit mitophagy in WAT. The findings will lay an experimental foundation for the development of new drugs targeting ASP and the clinical treatment of metabolic diseases related to obesity.
Collapse
Affiliation(s)
- Sheng Chen
- Provincial Key Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, 330006, P.R. China
- Department of Pharmacology, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, P.R. China
| | - Wanwan Yuan
- Provincial Key Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, 330006, P.R. China
- Department of Pharmacology, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, P.R. China
| | - Qianqian Huang
- Provincial Key Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, 330006, P.R. China
- Department of Pharmacology, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, P.R. China
| | - Xiaowei Xiong
- Provincial Key Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, 330006, P.R. China
- Department of Pharmacology, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, P.R. China
| | - Chaowen Wang
- Provincial Key Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, 330006, P.R. China
- Department of Pharmacology, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, P.R. China
| | - Wenjing Zeng
- Provincial Key Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, 330006, P.R. China
- Department of Pharmacology, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, P.R. China
| | - Li Wang
- Provincial Key Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, 330006, P.R. China
- Department of Pharmacology, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, P.R. China
| | - Yijun Huang
- Department of Pharmacology, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, P.R. China
| | - Yeyi Liu
- Department of Pharmacology, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, P.R. China
| | - Yan Wang
- Department of Pharmacology, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, P.R. China
| | - Qiren Huang
- Provincial Key Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, 330006, P.R. China.
- Department of Pharmacology, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, P.R. China.
| |
Collapse
|
11
|
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
|
12
|
Swan J, Szabó Z, Peters J, Kummu O, Kemppi A, Rahtu-Korpela L, Konzack A, Hakkola J, Pasternack A, Ritvos O, Kerkelä R, Magga J. Inhibition of activin receptor 2 signalling ameliorates metabolic dysfunction-associated steatotic liver disease in western diet/L-NAME induced cardiometabolic disease. Biomed Pharmacother 2024; 175:116683. [PMID: 38705130 DOI: 10.1016/j.biopha.2024.116683] [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: 01/09/2024] [Revised: 04/19/2024] [Accepted: 04/29/2024] [Indexed: 05/07/2024] Open
Abstract
OBJECTIVE Blockade of activin 2 receptor (ACVR2) signaling has been shown to improve insulin sensitivity and aid in weight loss. Inhibition of ACVR2 signaling restores cardiac function in multiple heart failure models. However, its potential in the treatment of obesity-related cardiometabolic disease remains unknown. Here, we investigated targeting ACVR2 signaling in cardiometabolic disease manifested with metabolic dysfunction-associated steatotic liver disease (MASLD). METHODS Mice were fed a high-fat, high-sugar diet combined with the administration of nitric oxide synthase inhibitor L-NAME in drinking water, which causes hypertensive stress. For the last eight weeks, the mice were treated with the soluble ACVR2B decoy receptor (sACVR2B-Fc). RESULTS sACVR2B-Fc protected against the development of comorbidities associated with cardiometabolic disease. This was most pronounced in the liver where ACVR2 blockade attenuated the development of MASLD including cessation of pro-fibrotic activation. It also significantly reduced total plasma cholesterol levels, impeded brown adipose tissue whitening, and improved cardiac diastolic function. In vitro, ACVR2 ligands activin A, activin B and GDF11 induced profibrotic signaling and the proliferation of human cardiac fibroblasts. CONCLUSIONS Blockade of ACVR2B exerts broad beneficial effects for therapy of cardiometabolic disease. By reducing obesity, ameliorating cardiovascular deterioration and restraining MASLD, blockade of ACVR2B signaling proves a potential target in MASLD and its comorbidities.
Collapse
Affiliation(s)
- Julia Swan
- Research Unit of Biomedicine and Internal Medicine, University of Oulu, Aapistie 5, Oulu 90220, Finland; Biocenter Oulu, University of Oulu, Aapistie 5, Oulu 90220, Finland.
| | - Zoltán Szabó
- Research Unit of Biomedicine and Internal Medicine, University of Oulu, Aapistie 5, Oulu 90220, Finland
| | - Juliana Peters
- Research Unit of Biomedicine and Internal Medicine, University of Oulu, Aapistie 5, Oulu 90220, Finland
| | - Outi Kummu
- Research Unit of Biomedicine and Internal Medicine, University of Oulu, Aapistie 5, Oulu 90220, Finland; Biocenter Oulu, University of Oulu, Aapistie 5, Oulu 90220, Finland; Medical Research Centre Oulu, Oulu University Hospital and University of Oulu, Aapistie 5, Oulu 90220, Finland
| | - Anna Kemppi
- Research Unit of Biomedicine and Internal Medicine, University of Oulu, Aapistie 5, Oulu 90220, Finland
| | - Lea Rahtu-Korpela
- Research Unit of Biomedicine and Internal Medicine, University of Oulu, Aapistie 5, Oulu 90220, Finland
| | - Anja Konzack
- Research Unit of Biomedicine and Internal Medicine, University of Oulu, Aapistie 5, Oulu 90220, Finland; Biocenter Oulu, University of Oulu, Aapistie 5, Oulu 90220, Finland; Medical Research Centre Oulu, Oulu University Hospital and University of Oulu, Aapistie 5, Oulu 90220, Finland
| | - Jukka Hakkola
- Research Unit of Biomedicine and Internal Medicine, University of Oulu, Aapistie 5, Oulu 90220, Finland; Biocenter Oulu, University of Oulu, Aapistie 5, Oulu 90220, Finland; Medical Research Centre Oulu, Oulu University Hospital and University of Oulu, Aapistie 5, Oulu 90220, Finland
| | - Arja Pasternack
- Department of Physiology, Faculty of Medicine, University of Helsinki, Haartmaninkatu 8, Helsinki 00014, Finland
| | - Olli Ritvos
- Department of Physiology, Faculty of Medicine, University of Helsinki, Haartmaninkatu 8, Helsinki 00014, Finland
| | - Risto Kerkelä
- Research Unit of Biomedicine and Internal Medicine, University of Oulu, Aapistie 5, Oulu 90220, Finland; Biocenter Oulu, University of Oulu, Aapistie 5, Oulu 90220, Finland; Medical Research Centre Oulu, Oulu University Hospital and University of Oulu, Aapistie 5, Oulu 90220, Finland
| | - Johanna Magga
- Research Unit of Biomedicine and Internal Medicine, University of Oulu, Aapistie 5, Oulu 90220, Finland; Biocenter Oulu, University of Oulu, Aapistie 5, Oulu 90220, Finland.
| |
Collapse
|
13
|
Song M, Tang Y, Cao K, Qi L, Xie K. Unveiling the role of interleukin-6 in pancreatic cancer occurrence and progression. Front Endocrinol (Lausanne) 2024; 15:1408312. [PMID: 38828409 PMCID: PMC11140100 DOI: 10.3389/fendo.2024.1408312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 05/06/2024] [Indexed: 06/05/2024] Open
Abstract
Pancreatic cancer is difficult to diagnose early and progresses rapidly. Researchers have found that a cytokine called Interleukin-6 (IL-6) is involved in the entire course of pancreatic cancer, promoting its occurrence and development. From the earliest stages of pancreatic intraepithelial neoplasia to the invasion and metastasis of pancreatic cancer cells and the appearance of tumor cachexia, IL-6 drives oncogenic signal transduction pathways and immune escape that accelerate disease progression. IL-6 is considered a biomarker for pancreatic cancer diagnosis and prognosis, as well as a potential target for treatment. IL-6 antibodies are currently being explored as a hot topic in oncology. This article aims to systematically explain how IL-6 induces the deterioration of normal pancreatic cells, with the goal of finding a breakthrough in pancreatic cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Meihui Song
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
- Division of Gastroenterology, Institute of Digestive Disease, Qingyuan People’s Hospital, The Affiliated Qingyuan Hospital of Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Ying Tang
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Kaimei Cao
- Division of Gastroenterology, Institute of Digestive Disease, Qingyuan People’s Hospital, The Affiliated Qingyuan Hospital of Guangzhou Medical University, Qingyuan, Guangdong, China
- School of Pharmaceutical Sciences, Dali University, Dali, Yunnan, China
| | - Ling Qi
- Division of Gastroenterology, Institute of Digestive Disease, Qingyuan People’s Hospital, The Affiliated Qingyuan Hospital of Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Keping Xie
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| |
Collapse
|
14
|
Shin J, Lee Y, Ju SH, Jung YJ, Sim D, Lee SJ. Unveiling the Potential of Natural Compounds: A Comprehensive Review on Adipose Thermogenesis Modulation. Int J Mol Sci 2024; 25:4915. [PMID: 38732127 PMCID: PMC11084502 DOI: 10.3390/ijms25094915] [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: 04/04/2024] [Revised: 04/19/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024] Open
Abstract
The process of adipocyte browning has recently emerged as a novel therapeutic target for combating obesity and obesity-related diseases. Non-shivering thermogenesis is the process of biological heat production in mammals and is primarily mediated via brown adipose tissue (BAT). The recruitment and activation of BAT can be induced through chemical drugs and nutrients, with subsequent beneficial health effects through the utilization of carbohydrates and fats to generate heat to maintain body temperature. However, since potent drugs may show adverse side effects, nutritional or natural substances could be safe and effective as potential adipocyte browning agents. This review aims to provide an extensive overview of the natural food compounds that have been shown to activate brown adipocytes in humans, animals, and in cultured cells. In addition, some key genetic and molecular targets and the mechanisms of action of these natural compounds reported to have therapeutic potential to combat obesity are discussed.
Collapse
Affiliation(s)
- Jaeeun Shin
- Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02855, Republic of Korea; (J.S.); (Y.L.); (S.H.J.); (Y.J.J.); (D.S.)
| | - Yeonho Lee
- Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02855, Republic of Korea; (J.S.); (Y.L.); (S.H.J.); (Y.J.J.); (D.S.)
| | - Seong Hun Ju
- Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02855, Republic of Korea; (J.S.); (Y.L.); (S.H.J.); (Y.J.J.); (D.S.)
| | - Young Jae Jung
- Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02855, Republic of Korea; (J.S.); (Y.L.); (S.H.J.); (Y.J.J.); (D.S.)
| | - Daehyeon Sim
- Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02855, Republic of Korea; (J.S.); (Y.L.); (S.H.J.); (Y.J.J.); (D.S.)
| | - Sung-Joon Lee
- Department of Food Bioscience and Technology, College of Life Sciences and Biotechnology, Korea University, Seoul 02855, Republic of Korea
- Interdisciplinary Program in Precision Public Health, BK21 Four Institute of Precision Public Health, Korea University, Seoul 02846, Republic of Korea
| |
Collapse
|
15
|
Ghesmati Z, Rashid M, Fayezi S, Gieseler F, Alizadeh E, Darabi M. An update on the secretory functions of brown, white, and beige adipose tissue: Towards therapeutic applications. Rev Endocr Metab Disord 2024; 25:279-308. [PMID: 38051471 PMCID: PMC10942928 DOI: 10.1007/s11154-023-09850-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/30/2023] [Indexed: 12/07/2023]
Abstract
Adipose tissue, including white adipose tissue (WAT), brown adipose tissue (BAT), and beige adipose tissue, is vital in modulating whole-body energy metabolism. While WAT primarily stores energy, BAT dissipates energy as heat for thermoregulation. Beige adipose tissue is a hybrid form of adipose tissue that shares characteristics with WAT and BAT. Dysregulation of adipose tissue metabolism is linked to various disorders, including obesity, type 2 diabetes, cardiovascular diseases, cancer, and infertility. Both brown and beige adipocytes secrete multiple molecules, such as batokines, packaged in extracellular vesicles or as soluble signaling molecules that play autocrine, paracrine, and endocrine roles. A greater understanding of the adipocyte secretome is essential for identifying novel molecular targets in treating metabolic disorders. Additionally, microRNAs show crucial roles in regulating adipose tissue differentiation and function, highlighting their potential as biomarkers for metabolic disorders. The browning of WAT has emerged as a promising therapeutic approach in treating obesity and associated metabolic disorders. Many browning agents have been identified, and nanotechnology-based drug delivery systems have been developed to enhance their efficacy. This review scrutinizes the characteristics of and differences between white, brown, and beige adipose tissues, the molecular mechanisms involved in the development of the adipocytes, the significant roles of batokines, and regulatory microRNAs active in different adipose tissues. Finally, the potential of WAT browning in treating obesity and atherosclerosis, the relationship of BAT with cancer and fertility disorders, and the crosstalk between adipose tissue with circadian system and circadian disorders are also investigated.
Collapse
Affiliation(s)
- Zeinab Ghesmati
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohsen Rashid
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shabnam Fayezi
- Department of Gynecologic Endocrinology and Fertility Disorders, Women's Hospital, Ruprecht-Karls University of Heidelberg, Heidelberg, Germany
| | - Frank Gieseler
- Division of Experimental Oncology, Department of Hematology and Oncology, University Medical Center Schleswig-Holstein, Campus Lübeck, 23538, Lübeck, Germany
| | - Effat Alizadeh
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Masoud Darabi
- Division of Experimental Oncology, Department of Hematology and Oncology, University Medical Center Schleswig-Holstein, Campus Lübeck, 23538, Lübeck, Germany.
| |
Collapse
|
16
|
Abbasi M, Fan Z, Dawson JA, Wang S. Anti-obesity and metabolic benefits of metformin: Comparison of different delivery routes. J Drug Deliv Sci Technol 2024; 91:105110. [PMID: 38188941 PMCID: PMC10768944 DOI: 10.1016/j.jddst.2023.105110] [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] [Indexed: 01/09/2024]
Abstract
Obesity is a severe public health problem. Healthy lifestyle interventions are commonly recommended for fighting obesity. But they are hard to follow and have low efficacy. Pharmacotherapy and surgery are of high efficacy but are beset with side effects. Browning subcutaneous white adipose tissue (WAT) is a practical and efficient approach for combating obesity. Metformin, a commonly used FDA-approved antidiabetic drug, is potent to induce browning of WAT through phosphorylation and activation of AMP-activated protein kinase. However, oral administration of metformin has low oral bioavailability, fast renal clearance, and low target specificity that limit metformin's application in browning WAT. Local and transdermal delivery of metformin directly to subcutaneous WAT using injection or microneedle (MN) in combination with iontophoresis (INT) may solve these problems. In this paper, we administered metformin to C57BL/6J obese mice using the following three routes: transdermal delivery (MN and INT), local injection into inguinal WAT (IgWAT, a type of subcutaneous WAT in mice), and oral gavage. The anti-obesity and metabolic effects of metformin via these delivery routes were determined and compared. As compared to local IgWAT injection and oral gavage delivery, transdermal delivery of metformin using MN and INT resulted in 9% lower body weight and 7% decrease in body fat% accompanied by improved energy metabolism and decreased inflammation through browning IgWAT in obese C57BL/6J mice. Transdermal delivery of metformin using MN and INT is an effective approach in browning subcutaneous WAT for combating obesity and improving metabolic health.
Collapse
Affiliation(s)
- Mehrnaz Abbasi
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas 79409, USA
- College of Human Sciences, Auburn University, Auburn, AL, 36830, USA
| | - Zhaoyang Fan
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas 79409, USA
- School of Electrical, Computer and Energy Engineering, Arizona State University, Tempe, Arizona 85281, USA
| | - John A. Dawson
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas 79409, USA
- Department of Economics, Applied Statistics, and International, New Mexico State University, Las Cruces, New Mexico 88003, USA
| | - Shu Wang
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas 79409, USA
- College of Health Solutions, Arizona State University, Phoenix, Arizona, 85004, USA
| |
Collapse
|
17
|
Nucera S, Scarano F, Macrì R, Mollace R, Gliozzi M, Carresi C, Ruga S, Serra M, Tavernese A, Caminiti R, Coppoletta A, Cardamone A, Montalcini T, Pujia A, Palma E, Muscoli C, Barillà F, Musolino V, Mollace V. The Effect of an Innovative Combination of Bergamot Polyphenolic Fraction and Cynara cardunculus L. Extract on Weight Gain Reduction and Fat Browning in Obese Mice. Int J Mol Sci 2023; 25:191. [PMID: 38203362 PMCID: PMC10779365 DOI: 10.3390/ijms25010191] [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: 11/23/2023] [Revised: 12/16/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
Obesity is one of the world's most serious public health issues, with a high risk of developing a wide range of diseases. As a result, focusing on adipose tissue dysfunction may help to prevent the metabolic disturbances commonly associated with obesity. Nutraceutical supplementation may be a crucial strategy for improving WAT inflammation and obesity and accelerating the browning process. The aim of this study was to perform a preclinical "proof of concept" study on Bergacyn®, an innovative formulation originating from a combination of bergamot polyphenolic fraction (BPF) and Cynara cardunculus (CyC), for the treatment of adipose tissue dysfunction. In particular, Bergacyn® supplementation in WD/SW-fed mice at doses of 50 mg/kg given orally for 12 weeks, was able to reduce body weight and total fat mass in the WD/SW mice, in association with an improvement in plasma biochemical parameters, including glycemia, total cholesterol, and LDL levels. In addition, a significant reduction in serum ALT levels was highlighted. The decreased WAT levels corresponded to an increased weight of BAT tissue, which was associated with a downregulation of PPARγ as compared to the vehicle group. Bergacyn® was able to restore PPARγ levels and prevent NF-kB overexpression in the WAT of mice fed a WD/SW diet, suggesting an improved oxidative metabolism and inflammatory status. These results were associated with a significant potentiation of the total antioxidant status in WD/SW mice. Finally, our data show, for the first time, that Bergacyn® supplementation may be a valuable approach to counteract adipose tissue dysfunction and obesity-associated effects on cardiometabolic risk.
Collapse
Affiliation(s)
- Saverio Nucera
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (S.N.); (F.S.); (R.M.); (M.G.); (S.R.); (M.S.); (A.T.); (R.C.); (A.C.); (A.C.); (C.M.)
| | - Federica Scarano
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (S.N.); (F.S.); (R.M.); (M.G.); (S.R.); (M.S.); (A.T.); (R.C.); (A.C.); (A.C.); (C.M.)
| | - Roberta Macrì
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (S.N.); (F.S.); (R.M.); (M.G.); (S.R.); (M.S.); (A.T.); (R.C.); (A.C.); (A.C.); (C.M.)
| | - Rocco Mollace
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (S.N.); (F.S.); (R.M.); (M.G.); (S.R.); (M.S.); (A.T.); (R.C.); (A.C.); (A.C.); (C.M.)
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy;
| | - Micaela Gliozzi
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (S.N.); (F.S.); (R.M.); (M.G.); (S.R.); (M.S.); (A.T.); (R.C.); (A.C.); (A.C.); (C.M.)
| | - Cristina Carresi
- Veterinary Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (C.C.); (E.P.)
| | - Stefano Ruga
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (S.N.); (F.S.); (R.M.); (M.G.); (S.R.); (M.S.); (A.T.); (R.C.); (A.C.); (A.C.); (C.M.)
| | - Maria Serra
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (S.N.); (F.S.); (R.M.); (M.G.); (S.R.); (M.S.); (A.T.); (R.C.); (A.C.); (A.C.); (C.M.)
| | - Annamaria Tavernese
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (S.N.); (F.S.); (R.M.); (M.G.); (S.R.); (M.S.); (A.T.); (R.C.); (A.C.); (A.C.); (C.M.)
| | - Rosamaria Caminiti
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (S.N.); (F.S.); (R.M.); (M.G.); (S.R.); (M.S.); (A.T.); (R.C.); (A.C.); (A.C.); (C.M.)
| | - Annarita Coppoletta
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (S.N.); (F.S.); (R.M.); (M.G.); (S.R.); (M.S.); (A.T.); (R.C.); (A.C.); (A.C.); (C.M.)
| | - Antonio Cardamone
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (S.N.); (F.S.); (R.M.); (M.G.); (S.R.); (M.S.); (A.T.); (R.C.); (A.C.); (A.C.); (C.M.)
| | - Tiziana Montalcini
- Clinical Nutrition Unit, Department of Clinical and Experimental Medicine, University Magna of Græcia of Catanzaro, 88100 Catanzaro, Italy;
| | - Arturo Pujia
- Department of Medical and Surgical Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy;
| | - Ernesto Palma
- Veterinary Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (C.C.); (E.P.)
| | - Carolina Muscoli
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (S.N.); (F.S.); (R.M.); (M.G.); (S.R.); (M.S.); (A.T.); (R.C.); (A.C.); (A.C.); (C.M.)
| | - Francesco Barillà
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy;
| | - Vincenzo Musolino
- Pharmaceutical Biology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy;
| | - Vincenzo Mollace
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (S.N.); (F.S.); (R.M.); (M.G.); (S.R.); (M.S.); (A.T.); (R.C.); (A.C.); (A.C.); (C.M.)
- Renato Dulbecco Institute, Lamezia Terme, 88046 Catanzaro, Italy
| |
Collapse
|
18
|
Lee HS, Choi SM, Lim SH, Choi CI. Betanin from Beetroot ( Beta vulgaris L.) Regulates Lipid Metabolism and Promotes Fat Browning in 3T3-L1 Adipocytes. Pharmaceuticals (Basel) 2023; 16:1727. [PMID: 38139853 PMCID: PMC10748323 DOI: 10.3390/ph16121727] [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: 11/08/2023] [Revised: 12/06/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023] Open
Abstract
Fat browning, which converts white adipose tissue to brown, has attracted attention as a promising strategy for the treatment of obesity. Betanin (BT) has been reported to have potential anti-obesity activity. 3T3-L1 cells were differentiated for 7 days during BT treatment. The BT concentration range for the study was determined using an MTT assay, and lipid accumulation was evaluated by Oil-Red-O staining. The expression of protein level was analyzed by Western blot. Immunofluorescence images were performed with confocal microscopy to visually show the amount and location of thermogenesis factor uncoupling protein1 (UCP1) and mitochondria. qRT-PCR was performed to evaluate mRNA expression. BT inhibited lipid accumulation and increased the expression of UCP1, peroxisome-proliferator-activated receptor gamma (PPARγ), and PPARγ coactivator-1 alpha (PGC-1α). In addition, the increases in beige adipocyte-specific markers were observed, supporting BT-mediated browning of the fat tissue. The UCP1 was localized in the inner membrane of the mitochondria, and its expression was associated with mitochondrial activation. Consistent with this, the mRNA expression of mitochondrial biogenesis markers increased in 3T3-L1 cells after BT treatment. Immunofluorescence staining also indicated an increased number of mitochondria and UCP1, respectively. Moreover, BT inhibited lipogenesis and enhanced lipolysis and fatty acid oxidation. This mechanism has been suggested to be mediated by an adenosine monophosphate-activated protein kinase (AMPK) pathway. BT induces fat browning and regulates lipid metabolism via the AMPK-mediated pathway in 3T3-L1 cells, suggesting that BT can be a promising candidate for controlling obesity.
Collapse
Affiliation(s)
| | | | | | - Chang-Ik Choi
- Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang 10326, Republic of Korea; (H.S.L.); (S.M.C.); (S.H.L.)
| |
Collapse
|
19
|
Chen S, Fu Y, Wang T, Chen Z, Zhao P, Huang X, Qiao M, Li T, Song L. Effect of 2'-Fucosyllactose on Beige Adipocyte Formation in 3T3-L1 Adipocytes and C3H10T1/2 Cells. Foods 2023; 12:4137. [PMID: 38002194 PMCID: PMC10670332 DOI: 10.3390/foods12224137] [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: 10/25/2023] [Revised: 11/11/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
2'-Fucosyllactose (2'-FL), the functional oligosaccharide naturally present in milk, has been shown to exert health benefits. This study was aimed to investigate the effect of 2'-fucosyllactose (2'-FL) on the browning of white adipose tissue in 3T3-L1 adipocytes and C3H10T1/2 cells. The results revealed that 2'-FL decreased lipid accumulations with reduced intracellular triglyceride contents in vitro. 2'-FL intervention increased the mitochondria density and the proportion of UCP1-positive cells. The mRNA expressions of the mitochondrial biogenesis-related and browning markers (Cox7a, Cyto C, Tfam, Ucp1, Pgc1α, Prdm16, Cidea, Elovl3, Pparα, CD137, and Tmem26) were increased after 2'-FL intervention to some extent. Similarly, the protein expression of the browning markers, including UCP1, PGC1α, and PRDM16, was up-regulated in the 2'-FL group. Additionally, an adenosine monophosphate-activated protein kinase (AMPK) inhibitor, compound C (1 μM), significantly decreased the induction of thermogenic proteins expressions mediated by 2'-FL, indicating that the 2'-FL-enhanced beige cell formation was partially dependent on the AMPK pathway. In conclusion, 2'-FL effectively promoted the browning of white adipose in vitro.
Collapse
Affiliation(s)
- Siru Chen
- Henan Engineering Technology Research Center of Food Processing and Circulation Safety Control, College of Food Science and Technology, Henan Agricultural University, Zhengzhou 450002, China; (S.C.); (Z.C.)
| | - Yankun Fu
- Henan Engineering Technology Research Center of Food Processing and Circulation Safety Control, College of Food Science and Technology, Henan Agricultural University, Zhengzhou 450002, China; (S.C.); (Z.C.)
| | - Tianlin Wang
- Henan Engineering Technology Research Center of Food Processing and Circulation Safety Control, College of Food Science and Technology, Henan Agricultural University, Zhengzhou 450002, China; (S.C.); (Z.C.)
| | - Zhenglin Chen
- Henan Engineering Technology Research Center of Food Processing and Circulation Safety Control, College of Food Science and Technology, Henan Agricultural University, Zhengzhou 450002, China; (S.C.); (Z.C.)
| | - Peijun Zhao
- Henan Engineering Technology Research Center of Food Processing and Circulation Safety Control, College of Food Science and Technology, Henan Agricultural University, Zhengzhou 450002, China; (S.C.); (Z.C.)
| | - Xianqing Huang
- Henan Engineering Technology Research Center of Food Processing and Circulation Safety Control, College of Food Science and Technology, Henan Agricultural University, Zhengzhou 450002, China; (S.C.); (Z.C.)
- Henan Technology Innovation Center of Meat Processing and Research, College of Food Science and Technology, Henan Agricultural University, Zhengzhou 450002, China
| | - Mingwu Qiao
- Henan Engineering Technology Research Center of Food Processing and Circulation Safety Control, College of Food Science and Technology, Henan Agricultural University, Zhengzhou 450002, China; (S.C.); (Z.C.)
- Henan Technology Innovation Center of Meat Processing and Research, College of Food Science and Technology, Henan Agricultural University, Zhengzhou 450002, China
| | - Tiange Li
- Henan Engineering Technology Research Center of Food Processing and Circulation Safety Control, College of Food Science and Technology, Henan Agricultural University, Zhengzhou 450002, China; (S.C.); (Z.C.)
| | - Lianjun Song
- Henan Engineering Technology Research Center of Food Processing and Circulation Safety Control, College of Food Science and Technology, Henan Agricultural University, Zhengzhou 450002, China; (S.C.); (Z.C.)
- Henan Technology Innovation Center of Meat Processing and Research, College of Food Science and Technology, Henan Agricultural University, Zhengzhou 450002, China
| |
Collapse
|
20
|
Directo D, Lee SR. Cancer Cachexia: Underlying Mechanisms and Potential Therapeutic Interventions. Metabolites 2023; 13:1024. [PMID: 37755304 PMCID: PMC10538050 DOI: 10.3390/metabo13091024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 09/14/2023] [Accepted: 09/14/2023] [Indexed: 09/28/2023] Open
Abstract
Cancer cachexia, a multifactorial metabolic syndrome developed during malignant tumor growth, is characterized by an accelerated loss of body weight accompanied by the depletion of skeletal muscle mass. This debilitating condition is associated with muscle degradation, impaired immune function, reduced functional capacity, compromised quality of life, and diminished survival in cancer patients. Despite the lack of the known capability of fully reversing or ameliorating this condition, ongoing research is shedding light on promising preclinical approaches that target the disrupted mechanisms in the pathophysiology of cancer cachexia. This comprehensive review delves into critical aspects of cancer cachexia, including its underlying pathophysiological mechanisms, preclinical models for studying the progression of cancer cachexia, methods for clinical assessment, relevant biomarkers, and potential therapeutic strategies. These discussions collectively aim to contribute to the evolving foundation for effective, multifaceted counteractive strategies against this challenging condition.
Collapse
Affiliation(s)
| | - Sang-Rok Lee
- Department of Kinesiology, New Mexico State University, Las Cruces, NM 88003, USA;
| |
Collapse
|
21
|
Li L, Geng J, Yu W, Zhou F, Zheng Z, Fu K, Kong J, Feng X. Inhibition of PPARγ by BZ26, a GW9662 derivate, attenuated obesity-related breast cancer progression by inhibiting the reprogramming of mature adipocytes into to cancer associate adipocyte-like cells. Front Pharmacol 2023; 14:1205030. [PMID: 37649895 PMCID: PMC10462981 DOI: 10.3389/fphar.2023.1205030] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 07/31/2023] [Indexed: 09/01/2023] Open
Abstract
Obesity has been associated with the development of 13 different types of cancers, including breast cancer. Evidence has indicated that cancer-associated adipocytes promote the proliferation, invasion, and metastasis of cancer. However, the mechanisms that link CAAs to the progression of obesity-related cancer are still unknown. Here, we found the mature adipocytes in the visceral fat of HFD-fed mice have a CAAs phenotype but the stromal vascular fraction of the visceral fat has not. Importantly, we found the derivate of the potent PPARγ antagonist GW9662, BZ26 inhibited the reprogramming of mature adipocytes in the visceral fat of HFD-fed mice into CAA-like cells and inhibited the proliferation and invasion of obesity-related breast cancer. Further study found that it mediated the browning of visceral, subcutaneous and perirenal fat and attenuated inflammation of adipose tissue and metabolic disorders. For the mechanism, we found that BZ26 bound and inhibited PPARγ by acting as a new modulator. Therefore, BZ26 serves as a novel modulator of PPARγ activity, that is, capable of inhibiting obesity-related breast cancer progression by inhibiting of CAA-like cell formation, suggesting that inhibiting the reprogramming of mature adipocytes into CAAs or CAA-like cells may be a potential therapeutic strategy for obesity-related cancer treatment.
Collapse
Key Words
|