Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (158)

Search Parameters:
Keywords = human brown adipose tissue

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
17 pages, 8756 KiB  
Article
A Diet Rich in Essential Amino Acids Inhibits the Growth of HCT116 Human Colon Cancer Cell In Vitro and In Vivo
by Giovanni Corsetti, Claudia Romano, Silvia Codenotti, Evasio Pasini, Alessandro Fanzani, Tiziano Scarabelli and Francesco S. Dioguardi
Int. J. Mol. Sci. 2025, 26(14), 7014; https://doi.org/10.3390/ijms26147014 - 21 Jul 2025
Viewed by 337
Abstract
The metabolic hyperactivity of tumor cells demands a substantial amount of energy and molecules to build new cells and expand the tumor, diverting these resources from healthy cells. Amino acids (AAs) are the only totipotent and essential molecules for protein construction. Previous in [...] Read more.
The metabolic hyperactivity of tumor cells demands a substantial amount of energy and molecules to build new cells and expand the tumor, diverting these resources from healthy cells. Amino acids (AAs) are the only totipotent and essential molecules for protein construction. Previous in vitro studies in human and murine cancer cells, along with in vivo studies in mice, have shown that an excess of essential amino acids (EAAs) exerts an inhibitory effect on tumor proliferation by promoting apoptosis and autophagy. In this study, both in vitro and in vivo, we evaluated whether a mixture based on EAA can influence the development of human colon cancer (HCT116). To this end, in vitro, we assessed the proliferation of HCT116 cells treated with a special mix of EAA. In vivo, immunosuppressed athymic nude mice, injected with HCT116 cells subcutaneously (s.c.) or intraperitoneally (i.p.), were given a modified EAAs-rich diet (EAARD) compared to the standard laboratory diet (StD). In vitro data showed that the EAA mix impairs cancer growth by inducing apoptosis and autophagy. In vivo, the results demonstrated that EAARD-fed mice developed s.c. tumors significantly smaller than those of StD-fed mice (total mass 3.24 vs. 6.09 g, respectively). Mice injected i.p. and fed with EAARD showed a smaller and more limited number of intra-peritoneal tumors than StD-fed mice (total mass 0.79 vs. 4.77 g, respectively). EAAs prevents the growth of HCT116 cells by inducing autophagy and apoptosis, increasing endoplasmic reticulum stress, and inhibiting inflammation and neo-vascularization. In addition, the EAARD-fed mice, maintained muscle mass and white and brown adipose tissues. A diet with an excess of EAAs affects the survival and proliferative capacity of human colon cancer cells, maintaining anabolic stimuli in muscular cells. Full article
(This article belongs to the Special Issue Innovative Research on Nutrition and Epigenetics in Cancer)
Show Figures

Graphical abstract

26 pages, 1132 KiB  
Review
GLP-1 and Its Role in Glycogen Production: A Narrative Review
by Joseph Lotosky, Xavier Jean, Anungoo Altankhuyag, Saqib Khan, Ashley Bernotas, Alireza Sharafshah, Kenneth Blum, Alan Posner and Panayotis K. Thanos
Biomedicines 2025, 13(7), 1610; https://doi.org/10.3390/biomedicines13071610 - 30 Jun 2025
Viewed by 1213
Abstract
Glucagon-like peptide-1 (GLP-1) has emerged as a pivotal regulator in the management of glucose homeostasis, glycogen metabolism, and energy balance, positioning it as a critical therapeutic target for addressing obesity, metabolic syndrome, and type 2 diabetes mellitus (T2DM). GLP-1 receptor agonists (GLP-1RAs) have [...] Read more.
Glucagon-like peptide-1 (GLP-1) has emerged as a pivotal regulator in the management of glucose homeostasis, glycogen metabolism, and energy balance, positioning it as a critical therapeutic target for addressing obesity, metabolic syndrome, and type 2 diabetes mellitus (T2DM). GLP-1 receptor agonists (GLP-1RAs) have shown promise for improving glycemic control and reducing weight through appetite regulation, delayed gastric emptying, and energy expenditure modulation. This narrative review explores the mechanisms of GLP-1-mediated glycogen metabolism and energy expenditure, particularly in key tissues—pancreas, liver, skeletal muscle, and adipose tissue. In the pancreas, GLP-1 enhances insulin secretion and beta-cell function. In the liver, it promotes glycogen synthesis via insulin-dependent and potential insulin-independent pathways, involving protein kinase B (AKT) and AMP-activated protein kinase (AMPK) signaling. Skeletal muscle benefits from GLP-1 through increased glucose uptake, AMPK activation, and mitochondrial function, facilitating glycogen storage. In adipose tissue, GLP-1 stimulates brown adipose tissue (BAT) thermogenesis and energy expenditure, contributing to weight loss. This increase in energy expenditure, along with enhanced glycogen metabolism, is a plausible mechanism for the weight loss observed with GLP-1RAs. Despite these advances, significant knowledge gaps remain, particularly regarding the direct hepatic effects of GLP-1, the extent to which it modulates glycogen metabolism in vivo, and its impact on thermogenesis in humans. Future research focusing on both the tissue-specific actions of GLP-1 and its systemic role in energy homeostasis and metabolic regulation will be essential for optimizing its therapeutic potential. Full article
(This article belongs to the Section Endocrinology and Metabolism Research)
Show Figures

Figure 1

18 pages, 4077 KiB  
Article
Exosome-Derived miR-11987 in Bovine Milk Inhibits Obesity Through Browning of White Fat
by In-Seon Bae and Sang Hoon Kim
Int. J. Mol. Sci. 2025, 26(13), 6006; https://doi.org/10.3390/ijms26136006 - 23 Jun 2025
Viewed by 419
Abstract
The global obese population accounts for approximately 30% of the total population and continues to increase. White adipocytes, which accumulate in the body for energy storage, are associated with obesity. Mechanisms that activate browning of white adipocytes are an attractive therapeutic target for [...] Read more.
The global obese population accounts for approximately 30% of the total population and continues to increase. White adipocytes, which accumulate in the body for energy storage, are associated with obesity. Mechanisms that activate browning of white adipocytes are an attractive therapeutic target for obesity and metabolic disorders. Exosomes are nano-sized biovesicles that play a role in cell-to-cell communication though the transfer of cargos such as microRNAs. Although milk exosomes contain many endogenous microRNA molecules, the role of microRNAs in milk exosomes is limited. Therefore, the aim of this study was to investigate the effects of milk exosomes on the browning of white adipocyte. Mouse pre-adipocytes (3T3-L1) and human adipose-derived stem cells (hADSCs) were differentiated and exposed to milk exosomes. Compared to control, milk exosomes promoted the expression of thermogenic genes and cellular mitochondrial energy metabolism in both 3T3-L1 cells and hADSCs. Additionally, milk exosomes were orally administered to mice fed a high-fat diet. As the intake of milk exosomes increased, the mice’s body weight decreased. Milk exosomes also increased the protein levels of thermogenic genes and mitochondrial-related genes in mouse adipose tissue. The overexpression of miR-11987, which is abundant in milk exosomes, in both 3T3-L1 cells and hADSCs led to the increased expression of thermogenic genes and mitochondrial activity. Our results support that bovine-specific miR-11987 in milk exosomes promotes the browning of white adipocytes. Therefore, milk exosome and milk exosomal miR-11987 could have significant clinical implications for obesity and metabolic syndrome. Full article
(This article belongs to the Special Issue Molecular Research on Diabetes and Obesity)
Show Figures

Figure 1

11 pages, 497 KiB  
Communication
Acute Downregulation of Zinc α2-Glycoprotein: Evidence from Human and HepG2 Cell Studies
by Èlia Navarro-Masip, David M. Selva, Cristina Hernández, Andreea Ciudin, Blanca Salinas-Roca, Julia Cabrera-Serra, Rafael Simó and Albert Lecube
Int. J. Mol. Sci. 2025, 26(12), 5438; https://doi.org/10.3390/ijms26125438 - 6 Jun 2025
Viewed by 443
Abstract
Zinc-alpha2-glycoprotein (ZAG) is a soluble glycoprotein primarily produced in adipocytes and the liver, with key roles in lipid metabolism, including lipolysis and the browning of adipose tissue. Despite extensive studies on its role in rodents, the relationship between ZAG and insulin in humans [...] Read more.
Zinc-alpha2-glycoprotein (ZAG) is a soluble glycoprotein primarily produced in adipocytes and the liver, with key roles in lipid metabolism, including lipolysis and the browning of adipose tissue. Despite extensive studies on its role in rodents, the relationship between ZAG and insulin in humans remains unclear. Given the emerging interest in ZAG’s involvement in metabolic diseases such as metabolic-dysfunction-associated steatotic liver disease, this study aimed to investigate the acute effects of insulin on ZAG levels both in vivo and in vitro. We recruited 24 healthy, individuals who were non-obese and assessed the impact of oral glucose overload, a standardized liquid nutritional supplement, and intravenous glucagon on circulating ZAG levels. In parallel, we explored the effects of insulin on ZAG production in cultured HepG2 cells. Our findings revealed a consistent acute reduction in serum ZAG levels following all in vivo tests, coinciding with increased insulin levels. In vitro, insulin rapidly downregulated ZAG protein and mRNA levels in HepG2 cells, with significant reductions observed within 15 min, followed by partial recovery after 2 h. These results suggest a potential acute inhibitory effect of insulin on ZAG production, supporting its role in promoting energy storage by suppressing lipolysis postprandially. This study provides new insights into the complex interplay between insulin and ZAG in regulating energy balance and highlights the potential of ZAG as a therapeutic target in metabolic diseases. Full article
Show Figures

Figure 1

19 pages, 3635 KiB  
Article
Protective Effects of a Standardized Water Extract from the Stem of Ipomoea batatas L. Against High-Fat Diet-Induced Obesity
by Chae-Won Lee, Ye Seul Yoon, Young-Seo Yoon, Kyung-Sook Chung, Mi-ju Kim, Geonha Park, Minsik Choi, Young-Pyo Jang and Kyung-Tae Lee
Nutrients 2025, 17(10), 1643; https://doi.org/10.3390/nu17101643 - 12 May 2025
Viewed by 796
Abstract
Background/Objectives: Obesity is a major health concern that can lead to various chronic diseases. Little is known about the anti-obesity effect of a standardized hot water extract from the stems of Ipomoea batatas (WIB). This study aimed to evaluate the therapeutic potential of [...] Read more.
Background/Objectives: Obesity is a major health concern that can lead to various chronic diseases. Little is known about the anti-obesity effect of a standardized hot water extract from the stems of Ipomoea batatas (WIB). This study aimed to evaluate the therapeutic potential of WIB as a natural alternative to conventional anti-obesity treatments by assessing its effects on body weight, fat accumulation, and key metabolic biomarkers in a high-fat diet-induced obesity model. Methods: A high-fat diet (HFD) induced obesity in C57BL/6 mice. The mice were then treated orally with either orlistat (positive control) or WIB. Changes in body weight, food intake, and fat weight were measured, along with blood lipid profiles and adipokines. Western blot analyses were conducted to determine protein levels in each tissue. H&E staining in white adipose tissue and liver, and the gut microbiota composition were analyzed. Results: WIB treatment significantly reduced body weight and fat mass compared to the HFD group and demonstrated comparable effects to orlistat. WIB improved blood lipid profiles and adipokine levels. H&E staining revealed reduced fat accumulation in the white adipose tissue and liver. Also in those tissues, WIB restored expression levels of sterol regulatory element-binding protein-1 (SREBP-1) and CCAAT/enhancer-binding protein α (C/EBPα) and increased AMP-activated protein kinase (AMPK) phosphorylation. In brown adipose tissue, WIB enhanced AMPK phosphorylation and upregulated thermogenic-related proteins, including peroxisome proliferator-activated receptor-gamma coactivator-1α (PGC-1α), peroxisome proliferator-activated receptor α (PPARα), sirtuin 1 (SIRT1), uncoupling protein-1 (UCP-1), and cytochrome C oxidase subunit 4 (COX-IV). Analysis of gut microbiota revealed that WIB normalized β-diversity and reversed HFD-induced phyla imbalances (notably in Bacteroidetes, Firmicutes, and Proteobacteria). Conclusions: By reducing adiposity under the conditions tested in a murine model, improving metabolic markers, and favorably modulating gut microbiota, WIB demonstrates potential in mitigating obesity-related risks. These findings suggest that WIB may serve as a promising natural substance for the management of obesity. Further studies are warranted to confirm its efficacy and explore the potential underlying mechanisms in overweight or obese humans as a health supplement to help manage or prevent obesity. Full article
Show Figures

Figure 1

23 pages, 1549 KiB  
Review
Meteorin-β: A Novel Biomarker and Therapeutic Target on Its Way to the Regulation of Human Diseases
by Bei Wang, Xiao Li and Xun Gao
Int. J. Mol. Sci. 2025, 26(10), 4485; https://doi.org/10.3390/ijms26104485 - 8 May 2025
Viewed by 687
Abstract
The novel secreted protein Meteorin-β (Metrnβ) is a homologous protein of the neurotrophic regulator Meteorin, which is widely expressed in the skin, mucous membranes, and white adipose tissue upon stimulation by a variety of inflammatory mediators, including cytokines and chemokines, while, at the [...] Read more.
The novel secreted protein Meteorin-β (Metrnβ) is a homologous protein of the neurotrophic regulator Meteorin, which is widely expressed in the skin, mucous membranes, and white adipose tissue upon stimulation by a variety of inflammatory mediators, including cytokines and chemokines, while, at the same time Metrnβ may also regulate the expression of these cytokines and chemokines. As a small secreted protein with low tissue specificity, Metrnβ plays vital roles in energy metabolism, insulin sensitivity regulation, neurodevelopment, white fat browning, and inflammatory response. Specifically, Metrnβ may act as an adipokine, myokine, neurotrophic factor, and cytokine, thereby being involved in the pathological and physiological processes of various human diseases, including metabolic, autoimmune and infectious/allergic diseases, and certain types of tumors. This review aims to systematically introduce the current research progress on Metrnβ, including its expression and distribution profiles, biological functions, and immunomodulatory roles in the process of human diseases. Additionally, we also discuss its potential as a biomarker, as well as a therapeutic/preventive agent for human diseases. Full article
Show Figures

Figure 1

16 pages, 3791 KiB  
Article
Effects of 17,18-Epoxyeicosatetraenoic Acid and 19,20-Epoxydocosapentaenoic Acid Combined with Soluble Epoxide Hydrolase Inhibitor t-TUCB on Brown Adipogenesis and Mitochondrial Respiration
by Yang Yang, Haoying Wu, Xinyun Xu, Christophe Morisseau, Kin Sing Stephen Lee, Bruce D. Hammock, Jiangang Chen and Ling Zhao
Nutrients 2025, 17(6), 936; https://doi.org/10.3390/nu17060936 - 7 Mar 2025
Viewed by 1093
Abstract
Background/Objectives: 17,18-epoxyeicosatetraenoic acid (17,18-EEQ) and 19,20-epoxydocosapentaenoic acid (19,20-EDP) are bioactive metabolites produced from eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), respectively, by CYP450s. These metabolites are unstable and quickly metabolized by auto-oxidation, esterification, β-oxidation, or hydrolysis by soluble epoxide hydrolase (sEH). 17,18-EEQ [...] Read more.
Background/Objectives: 17,18-epoxyeicosatetraenoic acid (17,18-EEQ) and 19,20-epoxydocosapentaenoic acid (19,20-EDP) are bioactive metabolites produced from eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), respectively, by CYP450s. These metabolites are unstable and quickly metabolized by auto-oxidation, esterification, β-oxidation, or hydrolysis by soluble epoxide hydrolase (sEH). 17,18-EEQ or 19,20-EDP combined with a potent sEH inhibitor t-TUCB differentially activated brown adipose tissue in diet-induced obesity. In the current study, we investigated whether these n-3 epoxy fatty acids with t-TUCB directly promote brown adipocyte differentiation and their thermogenic capacities. Methods: Murine brown preadipocytes were treated with 17,18-EEQ or 19,20-EDP with t-TUCB during and post differentiation. Brown marker protein expression and mitochondrial respiration were measured. In addition, the activation of PPARγ and suppression of NFκB reporter by 17,18-EEQ or 19,20-EDP alone or with t-TUCB were assessed, and the roles of PPARγ were evaluated with PPARγ knockdown and GW9662. Results: 17,18-EEQ or 19,20-EDP with t-TUCB promoted brown adipogenesis and mitochondrial respiration and uncoupling. Moreover, with t-TUCB, both epoxides improved mitochondrial respiration, but only 17,18-EEQ with t-TUCB significantly increased mitochondrial uncoupling (and heat production) in the differentiated adipocytes. PPARγ may be required for the effects of epoxides on differentiation but not on the thermogenic function post differentiation. Conclusions: The results demonstrate that, with t-TUCB, 17,18-EEQ and 19,20-EDP promote brown adipogenesis and mitochondrial respiration and uncoupling. 17,18-EEQ also promotes thermogenesis in differentiated brown adipocytes. Together, the results suggest thermogenic potentials of tested n-3 epoxides, especially 17,18-EEQ with t-TUCB. Translational studies of these n-3 epoxides on human brown adipocyte differentiation and functions are warranted. Full article
(This article belongs to the Section Nutrition and Metabolism)
Show Figures

Figure 1

26 pages, 4223 KiB  
Article
CTHRC1 Expression Results in Secretion-Mediated, SOX9-Dependent Suppression of Adipogenesis: Implications for the Regulatory Role of Newly Identified CTHRC1+/PDGFR-Alpha+ Stromal Cells of Adipose
by Matthew E. Siviski, Rachel Bercovitch, Kathleen Pyburn, Christian Potts, Shivangi R. Pande, Carlos A. Gartner, William Halteman, Doreen Kacer, Barbara Toomey, Calvin Vary, Robert Koza, Lucy Liaw, Sergey Ryzhov, Volkhard Lindner and Igor Prudovsky
Int. J. Mol. Sci. 2025, 26(5), 1804; https://doi.org/10.3390/ijms26051804 - 20 Feb 2025
Viewed by 876
Abstract
Adipogenesis is regulated by the coordinated activity of adipogenic transcription factors including PPAR-gamma and C/EBP alpha, while dysregulated adipogenesis can predispose adipose tissues to adipocyte hypertrophy and hyperplasia. We have previously reported that Cthrc1-null mice have increased adiposity compared to wildtype mice, [...] Read more.
Adipogenesis is regulated by the coordinated activity of adipogenic transcription factors including PPAR-gamma and C/EBP alpha, while dysregulated adipogenesis can predispose adipose tissues to adipocyte hypertrophy and hyperplasia. We have previously reported that Cthrc1-null mice have increased adiposity compared to wildtype mice, supporting the notion that CTHRC1 regulates body composition. Herein, we derived conditioned medium from 3T3-L1 cells expressing human CTHRC1 and investigated its anti-adipogenic activity. This constituent significantly reduced 3T3-L1 cell adipogenic differentiation commensurate to the marked suppression of Cebpa and Pparg gene expression. It also increased the expression of the anti-adipogenic transcription factor SOX9 and promoted its nuclear translocation. Importantly, Sox9 gene knockdown demonstrated that the anti-adipogenic effect produced by this conditioned medium is dependent on SOX9 expression, while its ability to positively regulate SOX9 was attenuated by the application of Rho and Rac1 signaling pathway inhibitors. We also identified the selective expression of CTHRC1 in PDGFRA-expressing cell populations in human white adipose tissue, but not brown or perivascular adipose tissues. Congruently, flow cytometry revealed CTHRC1 expression in PDGFR-alpha+ stromal cells of mouse white adipose tissue, thus defining a novel stromal cell population that could underpin the ability of CTHRC1 to regulate adiposity. Full article
(This article belongs to the Section Biochemistry)
Show Figures

Figure 1

15 pages, 5376 KiB  
Article
Butyrate Prevents Obesity Accompanied by HDAC9-Mediated Browning of White Adipose Tissue
by Jing Yang, Guoli Li, Shan Wang, Mingqian He, Sijing Dong, Ting Wang, Binyin Shi, Patrick C. N. Rensen and Yanan Wang
Biomedicines 2025, 13(2), 260; https://doi.org/10.3390/biomedicines13020260 - 21 Jan 2025
Cited by 1 | Viewed by 2223
Abstract
Background/Objectives: Mounting evidence indicates that the short-chain fatty acid butyrate protects against obesity and associated comorbidities, partially through the induction of adipose tissue thermogenesis. However, the effects of butyrate on white adipose tissue (WAT) browning and its molecular mechanism are still elusive. The [...] Read more.
Background/Objectives: Mounting evidence indicates that the short-chain fatty acid butyrate protects against obesity and associated comorbidities, partially through the induction of adipose tissue thermogenesis. However, the effects of butyrate on white adipose tissue (WAT) browning and its molecular mechanism are still elusive. The objective of this study was to investigate butyrate-induced thermogenesis in white adipose tissue and its underlying mechanism. Methods: We studied the effects of butyrate on diet-induced obesity in the humanized APOE*3-Leiden.CETP transgenic mouse model and explored factors related to white adipose browning. Specifically, mice were challenged with a high-fat diet supplemented with butyrate. Adiposity was measured to assess obesity development. Energy metabolism was detected using an indirect calorimetry system. RNA-seq analysis was conducted to analyze the transcription landscape of WAT and responsible targets. Furthermore, the revealed molecular mechanism was verified in vitro. Results: Butyrate alleviated high-fat diet-induced obesity and promoted energy expenditure accompanied by brown adipose tissue activation and WAT browning. Mechanistically, RNA-seq analysis revealed that butyrate downregulated HDAC9 in WAT. Additionally, butyrate decreased HDAC9 while increasing thermogenesis in vitro. Inhibition of HDAC9 with TMP269 promoted thermogenic gene expression, mimicking the effects of butyrate. Conclusions: Butyrate protects against diet-induced obesity accompanied by decreasing the expression of HDAC9 in white adipose tissue and inducing browning. This study reveals a new mechanism whereby butyrate activates adaptive thermogenesis and provides new insights for the development of weight-loss drugs targeting adipose HDAC9. Full article
Show Figures

Figure 1

20 pages, 2300 KiB  
Article
Human miR-1 Stimulates Metabolic and Thermogenic-Related Genes in Adipocytes
by Ester Díez-Sainz, Fermín I. Milagro, Paula Aranaz, José I. Riezu-Boj, Pierre-Louis Batrow, Laura Contu, Nadine Gautier, Ez-Zoubir Amri, Isabelle Mothe-Satney and Silvia Lorente-Cebrián
Int. J. Mol. Sci. 2025, 26(1), 276; https://doi.org/10.3390/ijms26010276 - 31 Dec 2024
Cited by 2 | Viewed by 1284
Abstract
MicroRNAs play a pivotal role in the regulation of adipose tissue function and have emerged as promising therapeutic candidates for the management of obesity and associated comorbidities. Among them, miR-1 could be a potential biomarker for metabolic diseases and contribute to metabolic homeostasis. [...] Read more.
MicroRNAs play a pivotal role in the regulation of adipose tissue function and have emerged as promising therapeutic candidates for the management of obesity and associated comorbidities. Among them, miR-1 could be a potential biomarker for metabolic diseases and contribute to metabolic homeostasis. However, thorough research is required to fully elucidate the impact of miR-1 on human adipocyte thermogenesis and metabolism. This study aimed to explore the effect of miR-1 on human adipocyte browning, a process whose activation has been linked to obesity protection and counteraction. Human multipotent adipose-derived stem cells, hMADS cells, were differentiated into white and brown-like adipocytes and transfected with miR-1 mimics for gene expression and western blotting analyses. miR-1 inhibited the expression of its previously validated target PTK9/TWF1 and modulated the expression profile of key genes involved in thermogenesis and adipocyte browning (increased UCP1 at mRNA and protein level, increased CPT1M, decreased HIF3A), adipocyte differentiation and metabolism (decreased PLIN1, FASN, RXRA, PPARG, FABP4, MAPKAPK2), as well as genes related to the cytoskeleton (decreased ACTB) and extracellular matrix (decreased COL1A1). These findings suggest that miR-1 can modulate the expression of adipocyte human genes associated with thermogenesis and metabolism, which could hold value for eventual therapeutic potential in obesity. Full article
(This article belongs to the Special Issue The Role of microRNA in Human Diseases: 3rd Edition)
Show Figures

Graphical abstract

28 pages, 1398 KiB  
Review
Spotlight on the Mechanism of Action of Semaglutide
by Ilias Papakonstantinou, Konstantinos Tsioufis and Vasiliki Katsi
Curr. Issues Mol. Biol. 2024, 46(12), 14514-14541; https://doi.org/10.3390/cimb46120872 - 23 Dec 2024
Cited by 7 | Viewed by 12302
Abstract
Initially intended to control blood glucose levels in patients with type 2 diabetes, semaglutide, a potent glucagon-like peptide 1 analogue, has been established as an effective weight loss treatment by controlling appetite. Integrating the latest clinical trials, semaglutide in patients with or without [...] Read more.
Initially intended to control blood glucose levels in patients with type 2 diabetes, semaglutide, a potent glucagon-like peptide 1 analogue, has been established as an effective weight loss treatment by controlling appetite. Integrating the latest clinical trials, semaglutide in patients with or without diabetes presents significant therapeutic efficacy in ameliorating cardiometabolic risk factors and physical functioning, independent of body weight reduction. Semaglutide may modulate adipose tissue browning, which enhances human metabolism and exhibits possible benefits in skeletal muscle degeneration, accelerated by obesity and ageing. This may be attributed to anti-inflammatory, mitochondrial biogenesis, antioxidant and autophagy-regulating effects. However, most of the supporting evidence on the mechanistic actions of semaglutide is preclinical, demonstrated in rodents and not actually confirmed in humans, therefore warranting caution in the interpretation. This article aims to explore potential innovative molecular mechanisms of semaglutide action in restoring the balance of several interlinking aspects of metabolism, pointing to distinct functions in inflammation and oxidative stress in insulin-sensitive musculoskeletal and adipose tissues. Moreover, possible applications in protection from infections and anti-aging properties are discussed. Semaglutide enhancement of the core molecular mechanisms involved in the progress of obesity and diabetes, although mostly preclinical, may provide a framework for future research applications in human diseases overall. Full article
(This article belongs to the Section Molecular Medicine)
Show Figures

Figure 1

16 pages, 3113 KiB  
Article
Silymarin-Enriched Extract from Milk Thistle Activates Thermogenesis in a Preclinical Model of High-Fat-Diet-Induced Obesity to Relieve Systemic Meta-Inflammation
by Marina Reguero, Guillermo Reglero, José Carlos Quintela, Ricardo Ramos-Ruiz, Ana Ramírez de Molina and Marta Gómez de Cedrón
Nutrients 2024, 16(23), 4166; https://doi.org/10.3390/nu16234166 - 30 Nov 2024
Viewed by 2461
Abstract
Background: Obesity and aging are associated with the progressive loss of brown adipose tissue (BAT), an increase in visceral white adipose tissue (vWAT), and a reduction in subcutaneous white adipose tissue (sWAT). The progressive expansion of visceral obesity promotes a low grade of [...] Read more.
Background: Obesity and aging are associated with the progressive loss of brown adipose tissue (BAT), an increase in visceral white adipose tissue (vWAT), and a reduction in subcutaneous white adipose tissue (sWAT). The progressive expansion of visceral obesity promotes a low grade of systemic chronic inflammation (meta-inflammation), contributing to the onset of comorbidities such as type 2 diabetes mellitus (T2DM), metabolic syndrome, and even cancer. Thus, preserving the thermogenic activity of adipose tissue and improving the metabolic flexibility of sWAT could be an effective strategy to prevent the development of metabolic chronic diseases and promote healthy aging. Precision nutrition has emerged as a complementary approach to control the metabolic alterations associated with unhealthy obesity and aging. In a previous work, we described that a silymarin-enriched extract from milk thistle (Mthistle) increased markers of browning and thermogenesis in vitro in human differentiated adipocytes (SGBS). Objectives/Methods: Therefore, this study aims to evaluate the potential of Mthistle to activate thermogenesis in a preclinical model of high-fat diet (HFD)-induced obesity (DIO). Results: Our results demonstrate that Mthistle increases systemic energy expenditure (EE), preserves body temperature after cold exposure, improves insulin resistance, and reduces inflammatory markers in WAT. Conclusions: Based on these results, silymarin-enriched extract from Mthistle may be proposed as a nutraceutical for the management of metabolic chronic diseases and/or accelerated aging. Full article
(This article belongs to the Section Phytochemicals and Human Health)
Show Figures

Graphical abstract

12 pages, 592 KiB  
Article
Dietary Intake Is Similar Among Adult Men with Different Levels of Cold-Induced Brown Adipose Tissue Activation
by Andres E. Carrillo, Petros C. Dinas, Argyro Krase, Eleni Nintou, Alexandros Georgakopoulos, Marinos Metaxas, Edward J. Ryan, Maria Vliora, Panagiotis Georgoulias, Sofia Chatziioannou and Andreas D. Flouris
Nutrients 2024, 16(21), 3697; https://doi.org/10.3390/nu16213697 - 30 Oct 2024
Viewed by 1820
Abstract
Background/Objectives: Brown adipose tissue (BAT) activation has important metabolic health implications, yet the relationship between habitual dietary intake and BAT activity in humans remains to be fully understood. Methods: We compared dietary intake among adult men with (BATpositive, age: 34.8 ± [...] Read more.
Background/Objectives: Brown adipose tissue (BAT) activation has important metabolic health implications, yet the relationship between habitual dietary intake and BAT activity in humans remains to be fully understood. Methods: We compared dietary intake among adult men with (BATpositive, age: 34.8 ± 5.4 years, BMI: 28.2 ± 5.3 kg/m2, n = 12) and without (BATnegative, age: 39.1 ± 4.1 years, BMI: 31.1 ± 6.7 kg/m2, n = 11) cold-induced BAT activation. Activation of BAT was measured immediately following 2 h of cold exposure using 18F fluorodeoxyglucose positron emission tomography and computed tomography reported as maximum standardized uptake (SUVmax). Participants categorized as BATpositive had an SUVmax > 1.5 g/mL that was normalized to lean body mass (SUVlean) for analysis. Shivering intensity was recorded every 15 min during cold exposure and dietary intake was estimated from 7 consecutive 24 h dietary recalls. Results: The BATnegative group was significantly older than the BATpositive group (p = 0.046). Although BATnegative participants consumed an average of 281.2 kcal/day more than BATpositive, there were no significant differences in dietary intake between groups (p ≥ 0.202). Further, no statistically significant associations between SUVlean and dietary intake among BATpositive participants were observed (p ≥ 0.175). Participants who shivered (n = 9) during cold exposure tended to be shorter (p = 0.056) and have a lower waist-to-hip ratio (p = 0.097) but did not differ in dietary intake (p ≥ 0.204) or BAT activity (p = 0.964) when compared to the non-shivering (n = 11) group. Conclusions: Our results indicate that BAT activity and shivering during cold exposure are more strongly related to variables such as age and body size or composition rather than habitual dietary intake. We conclude that habitual dietary intake likely has a negligible influence on BAT activity among adult men. Full article
(This article belongs to the Special Issue Food Intake and Adipose Tissue Metabolism)
Show Figures

Figure 1

15 pages, 2088 KiB  
Article
Mesenchymal Stem Cell-Derived Exosomes Attenuate Hepatic Steatosis and Insulin Resistance in Diet-Induced Obese Mice by Activating the FGF21-Adiponectin Axis
by Bobae Kim, Rwubuzizi Ronaldo, Beet-Na Kweon, Solhee Yoon, Yein Park, Jea-Hyun Baek, Jung Min Lee and Chang-Kee Hyun
Int. J. Mol. Sci. 2024, 25(19), 10447; https://doi.org/10.3390/ijms251910447 - 27 Sep 2024
Viewed by 2355
Abstract
Exosomes derived from mesenchymal stem cells have shown promise in treating metabolic disorders, yet their specific mechanisms remain largely unclear. This study investigates the protective effects of exosomes from human umbilical cord Wharton’s jelly mesenchymal stem cells (hWJMSCs) against adiposity and insulin resistance [...] Read more.
Exosomes derived from mesenchymal stem cells have shown promise in treating metabolic disorders, yet their specific mechanisms remain largely unclear. This study investigates the protective effects of exosomes from human umbilical cord Wharton’s jelly mesenchymal stem cells (hWJMSCs) against adiposity and insulin resistance in high-fat diet (HFD)-induced obese mice. HFD-fed mice treated with hWJMSC-derived exosomes demonstrated improved gut barrier integrity, which restored immune balance in the liver and adipose tissues by reducing macrophage infiltration and pro-inflammatory cytokine expression. Furthermore, these exosomes normalized lipid metabolism including lipid oxidation and lipogenesis, which alleviate lipotoxicity-induced endoplasmic reticulum (ER) stress, thereby decreasing fat accumulation and chronic tissue inflammation in hepatic and adipose tissues. Notably, hWJMSC-derived exosomes also promoted browning and thermogenic capacity of adipose tissues, which was linked to reduced fibroblast growth factor 21 (FGF21) resistance and increased adiponectin production. This process activated the AMPK-SIRT1-PGC-1α pathway, highlighting the role of the FGF21–adiponectin axis. Our findings elucidate the molecular mechanisms through which hWJMSC-derived exosomes counteract HFD-induced metabolic dysfunctions, supporting their potential as therapeutic agents for metabolic disorders. Full article
Show Figures

Figure 1

21 pages, 1756 KiB  
Article
Plant miR6262 Modulates the Expression of Metabolic and Thermogenic Genes in Human Hepatocytes and Adipocytes
by Ester Díez-Sainz, Fermín I. Milagro, Paula Aranaz, José I. Riezu-Boj and Silvia Lorente-Cebrián
Nutrients 2024, 16(18), 3146; https://doi.org/10.3390/nu16183146 - 18 Sep 2024
Cited by 1 | Viewed by 1655
Abstract
Background: Edible plants have been linked to the mitigation of metabolic disturbances in liver and adipose tissue, including the decrease of lipogenesis and the enhancement of lipolysis and adipocyte browning. In this context, plant microRNAs could be key bioactive molecules underlying the cross-kingdom [...] Read more.
Background: Edible plants have been linked to the mitigation of metabolic disturbances in liver and adipose tissue, including the decrease of lipogenesis and the enhancement of lipolysis and adipocyte browning. In this context, plant microRNAs could be key bioactive molecules underlying the cross-kingdom beneficial effects of plants. This study sought to explore the impact of plant-derived microRNAs on the modulation of adipocyte and hepatocyte genes involved in metabolism and thermogenesis. Methods: Plant miR6262 was selected as a candidate from miRBase for the predicted effect on the regulation of human metabolic genes. Functional validation was conducted after transfection with plant miRNA mimics in HepG2 hepatocytes exposed to free fatty acids to mimic liver steatosis and hMADs cells differentiated into brown-like adipocytes. Results: miR6262 decreases the expression of the predicted target RXRA in the fatty acids-treated hepatocytes and in brown-like adipocytes and affects the expression profile of critical genes involved in metabolism and thermogenesis, including PPARA, G6PC, SREBF1 (hepatocytes) and CIDEA, CPT1M and PLIN1 (adipocytes). Nevertheless, plant miR6262 mimic transfections did not decrease hepatocyte lipid accumulation or stimulate adipocyte browning. Conclusions: these findings suggest that plant miR6262 could have a cross-kingdom regulation relevance through the modulation of human genes involved in lipid and glucose metabolism and thermogenesis in adipocytes and hepatocytes. Full article
(This article belongs to the Section Nutrigenetics and Nutrigenomics)
Show Figures

Graphical abstract

Back to TopTop