1
|
Zambrano-Vásquez OR, Cortés-Camacho F, Castañeda-Sánchez JI, Aréchaga-Ocampo E, Valle-Velázquez E, Cabrera-Angeles JC, Sánchez-Gloria JL, Sánchez-Muñoz F, Arellano-Buendia AS, Sánchez-Lozada LG, Osorio-Alonso H. Update in non-alcoholic fatty liver disease management: role of sodium-glucose cotransporter 2 inhibitors. Life Sci 2025; 372:123638. [PMID: 40246191 DOI: 10.1016/j.lfs.2025.123638] [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: 12/18/2024] [Revised: 03/28/2025] [Accepted: 04/09/2025] [Indexed: 04/19/2025]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is characterized by excessive lipid accumulation in hepatocytes without significant alcohol consumption. It is closely associated with sedentarism, hypercaloric diets, obesity, dyslipidemia, insulin resistance, type 2 diabetes mellitus, and genetic predisposition. NAFLD comprises a spectrum of liver disorders, from simple steatosis to non-alcoholic (NASH) and liver cirrhosis. The complex etiological mechanisms include oxidative stress, inflammation, apoptosis, and fibrosis; therefore, its management is challenging. Sodium-glucose cotransporter type 2 inhibitors (SGLT2i), a class of antidiabetic drugs, have emerged as promising therapeutic agents due to their ability to improve key metabolic parameters, including obesity, dyslipidemia, insulin resistance, and hyperglycemia. This review explores the cellular mechanisms by which SGLT2i, either as monotherapy or combined with other treatments, modulate signaling pathways involved in lipid and carbohydrate metabolism. Additionally, we examine their effects on oxidative stress, inflammation, fibrosis, and apoptosis, which are critical drivers of NAFLD progression. This review is intended to summarize the multiple benefits of SGLT2 inhibitors and to educate healthcare providers on the therapeutic potential of these drugs in order to foster their incorporation into effective NAFLD management plans.
Collapse
Affiliation(s)
- Oscar R Zambrano-Vásquez
- Doctorado en Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Ciudad de México 04960, Mexico; Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, México City 14080, Mexico
| | - Fernando Cortés-Camacho
- Doctorado en Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana, Ciudad de México 04960, Mexico; Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, México City 14080, Mexico
| | - Jorge I Castañeda-Sánchez
- Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana, Unidad Xochimilco, México City 04960, Mexico
| | - Elena Aréchaga-Ocampo
- Departamento de Ciencias Naturales, Universidad Autónoma Metropolitana, Unidad Cuajimalpa, México City 05348, Mexico
| | - Estefanía Valle-Velázquez
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, México City 14080, Mexico
| | - Juan C Cabrera-Angeles
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, México City, Mexico
| | - José L Sánchez-Gloria
- Department of Internal Medicine, Division of Nephrology, Rush University Medical Center, Chicago, IL 60612, USA
| | - Fausto Sánchez-Muñoz
- Departamento de Fisiología, Instituto Nacional de Cardiología Ignacio Chávez, México City 14080, Mexico
| | - Abraham S Arellano-Buendia
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, México City 14080, Mexico
| | - Laura G Sánchez-Lozada
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, México City 14080, Mexico
| | - Horacio Osorio-Alonso
- Departamento de Fisiopatología Cardio-Renal, Instituto Nacional de Cardiología Ignacio Chávez, México City 14080, Mexico.
| |
Collapse
|
2
|
Vratarić M, Teofilović A, Milutinović DV, Veličković N, Vučićević L, Đmura G, Djordjevic A. Changes in lipid metabolism in the visceral rather than the subcutaneous adipose tissue depot attenuate metabolic disturbances in obesity-resistant mice fed a high-fat diet. J Nutr Biochem 2025; 141:109912. [PMID: 40174754 DOI: 10.1016/j.jnutbio.2025.109912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 03/12/2025] [Accepted: 03/27/2025] [Indexed: 04/04/2025]
Abstract
Obesity is characterized by an enlargement of white adipose tissue caused by caloric excess. The depot-specific adaptation of white adipose tissue in individuals resistant to obesity despite a high-calorie diet is crucial for understanding the pathogenesis of obesity and related metabolic disorders. Our aim was to characterize the metabolic and morphological state of obesity resistance and to investigate depot-specific changes in signaling pathways in epididymal visceral (eVAT) and inguinal subcutaneous (iSAT) white adipose tissue of C57BL/6J male mice on a high-fat diet (60 kcal% fats). After 14 weeks, the mice were categorized as obese (at least 30% higher body mass compared to the control group) or obesity-resistant (weight gain below 30%). Biochemical and morphological parameters, as well as histology, and signaling pathways involved in lipid metabolism, inflammation, and insulin sensitivity were investigated in eVAT and iSAT. The results showed unaltered body, total VAT and iSAT mass in obesity-resistant mice despite increased caloric intake. Leptin levels and glucose homeostasis were improved in these animals compared to the obese mice. In both eVAT and iSAT of the obesity-resistant mice, adipocyte size and lipolytic capacity were retained at control levels, while compared to the obese mice, preserved capacity for adipogenesis, improved local insulin sensitivity and the absence of inflammation were observed only in the eVAT. In conclusion, metabolic adaptation of eVAT rather than iSAT may have a substantial impact on the maintenance of the obesity-resistant phenotype with fewer metabolic complications, which could contribute to the improvement of existing obesity therapies.
Collapse
Affiliation(s)
- Miloš Vratarić
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Ana Teofilović
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia.
| | - Danijela Vojnović Milutinović
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Nataša Veličković
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Ljubica Vučićević
- Department of Neurophysiology, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Goran Đmura
- Animal Facility, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Ana Djordjevic
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
3
|
Li Y, Song Q, Guo R, Qian Y, Jiang Y, Song Z. Glucose metabolism through the hexosamine biosynthetic pathway drives hepatic de novo lipogenesis via promoting N-linked protein glycosylation. Am J Physiol Gastrointest Liver Physiol 2025; 328:G746-G759. [PMID: 40331866 DOI: 10.1152/ajpgi.00056.2025] [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] [Received: 02/19/2025] [Revised: 03/17/2025] [Accepted: 03/30/2025] [Indexed: 05/08/2025]
Abstract
De novo lipogenesis (DNL) converts excess glucose into lipids, whereas the hexosamine biosynthetic pathway (HBP), a glycolytic branch, generates UDP-N-acetylglucosamine for protein glycosylation, including O-GlcNAcylation and N-linked glycosylation. Both pathways are active in hepatocytes and integral to glucose metabolism; however, their functional interplay remains unclear. Here, we investigated the role of HBP in hepatic DNL activation using both in vitro and in vivo models. AML12 hepatocytes were cultured in low- and high-glucose media with or without HBP blockade, both pharmacologically and genetically. For in vivo studies, male C57BL/6J mice were subjected to a fasting-refeeding regimen with or without intraperitoneal administration of azaserine, a competitive inhibitor of glutamine-fructose-6-phosphate transaminase 1 (GFPT1), the rate-limiting enzyme of the HBP. Our results demonstrated that, in AML12 cells, glucose exposure activated both DNL and HBP, leading to triacylglycerol (TAG) accumulation, whereas HBP inhibition ameliorated DNL and TAG accumulation. In mice, refeeding after a 24-h fasting induced hepatic DNL, which was abolished by HBP inhibition, indicating its mechanistic involvement in glucose-driven lipogenesis. Mechanistically, we identified ATF4 as a key regulator of GFPT1 upregulation under high-glucose conditions. As expected, both glucose-treated hepatocytes and livers from fasting-refed mice exhibited increased protein glycosylation. Notably, blocking N-linked glycosylation, but not O-GlcNAcylation, abolished glucose-induced DNL activation, indicating that HBP is essential for glucose-induced DNL pathway activation via promoting N-linked glycosylation, independent of O-GlcNAcylation. In conclusion, our findings establish that an intact HBP is required for glucose-induced hepatic DNL activation, primarily through promoting protein N-linked glycosylation.NEW & NOTEWORTHY High-glucose exposure activates both hepatic HBP and DNL pathways. The glucose metabolism into HBP is essential for the activation of the DNL pathway. ATF4 activation plays a mechanistic role in high glucose-induced HBP activation. HBP drives high glucose-induced hepatic DNL activation via promoting N-linked protein glycosylation.
Collapse
Affiliation(s)
- Yanhui Li
- Department of Kinesiology and Nutrition, University of Illinois Chicago, Chicago, Illinois, United States
| | - Qing Song
- Department of Kinesiology and Nutrition, University of Illinois Chicago, Chicago, Illinois, United States
| | - Rui Guo
- Department of Kinesiology and Nutrition, University of Illinois Chicago, Chicago, Illinois, United States
| | - Yanyu Qian
- Department of Physiology and Biophysics, University of Illinois Chicago, Chicago, Illinois, United States
| | - Yuwei Jiang
- Department of Physiology and Biophysics, University of Illinois Chicago, Chicago, Illinois, United States
| | - Zhenyuan Song
- Department of Kinesiology and Nutrition, University of Illinois Chicago, Chicago, Illinois, United States
| |
Collapse
|
4
|
Rong P, Mu Y, Wang M, Chen L, Liu F, Jin Y, Feng W, Zhou K, Liang H, Wang HY, Chen S. Targeting IGF1 to alleviate obesity through regulating energy expenditure and fat deposition. SCIENCE CHINA. LIFE SCIENCES 2025; 68:1662-1675. [PMID: 39843847 DOI: 10.1007/s11427-024-2768-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 10/31/2024] [Indexed: 01/24/2025]
Abstract
Insulin-like growth factor 1 (IGF1) is a regulator of both cellular hypertrophy and lipogenesis, which are two key processes for pathogenesis of obesity. However, the in vivo role of IGF1 in the development of obesity remains unclear. Here, we show that IGF1 expression is increased in adipose tissue in obese human patients and animal models. Elevation of IGF1 is associated with increased lipogenic gene expression and decreased energy expenditure. Genetic down-regulation of IGF1 normalizes lipogenic gene expression, restores aberrant energy metabolism and alleviates obese phenotype of a genetic mouse model with IGF1-hypersecretion. Importantly, genetic down-regulation of IGF1 exerts similar effects on development of diet-induced obesity. Furthermore, berberine that is an AMP-activated protein kinase (AMPK) activator in medicinal herbs inhibits IGF1 secretion, decreases lipogenic gene expression and alleviates diet-induced adiposity. Collectively, our findings demonstrate that hypersecretion of IGF1 is a critical factor for the development of obesity and can be targeted using AMPK activators to alleviate obesity.
Collapse
Affiliation(s)
- Ping Rong
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing, 210061, China
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, 210061, China
| | - Yinqiu Mu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing, 210061, China
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, 210061, China
| | - Meiqin Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing, 210061, China
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, 210061, China
| | - Liang Chen
- College of Life Sciences, Anhui Medical University, Hefei, 230032, China
| | - Fangtong Liu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing, 210061, China
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, 210061, China
| | - Yuxin Jin
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing, 210061, China
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, 210061, China
| | - Weikuan Feng
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing, 210061, China
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, 210061, China
| | - Kun Zhou
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing, 210061, China
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, 210061, China
| | - Hui Liang
- Department of General Surgery, First Affiliated Hospital, Nanjing Medical University, Nanjing, 210029, China
| | - Hong-Yu Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing, 210061, China.
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, 210061, China.
| | - Shuai Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Endocrinology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Model Animal Research Center, Nanjing University, Nanjing, 210061, China.
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, School of Medicine, Nanjing University, Nanjing, 210061, China.
| |
Collapse
|
5
|
Ning B, Wang SA, Young MJ, Chen YC, Hung Y, Huong TT, Chang WC, Wang YC, Yu ML, Hsu KC, Hung JJ. USP24 upregulation stabilizes PKA-Cα to promote lipogenesis, inflammation, and fibrosis during MASH progression. J Biomed Sci 2025; 32:54. [PMID: 40448065 DOI: 10.1186/s12929-025-01148-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Accepted: 05/20/2025] [Indexed: 06/02/2025] Open
Abstract
BACKGROUND Ubiquitin-specific peptidase 24 (USP24), a deubiquitinating enzyme, regulates protein stability by removing ubiquitin. This study investigates the role of UPS24 in lipid metabolism, inflammation, and fibrosis. It also explores the effect of targeting USP24 on metabolic disorders, focusing on high-fat diet (HFD)-induced obesity and liver diseases. METHODS This study utilized CRISPR/Cas9 to create functional knockout mice (USP24C1695A) and treated HFD-fed mice with USP24 inhibitor (USP24-i-101). The effects of USP24 inhibition or knockout on 3T3-L1 derived adipocytes, primary hepatocytes, hepatic stellate cells, and murine hepatocyte cell line AML12 (alpha mouse liver 12) cells were assessed with RNA-sequencing. Molecular mechanisms and the interaction between USP24 and PKA-Cα were studied with co-immunoprecipitation. Downstream signaling pathways involving CREB, SREBP1, PPARγ, and C/EBPβ, as well as USP24 role in liver inflammation and fibrosis, were studied using western blot and real-time PCR. Clinical and animal tissue samples were examined with immunohistochemistry to identify the correlations between USP24 and metabolic-associated liver diseases. RESULTS Knockout or inhibition of USP24 reduced body weight, lipid accumulation, inflammation, and fibrosis in HFD-fed mice. The expression of genes related to lipogenesis, inflammation, and fibrosis was downregulated in USP24C1695A mice and those treated with USP24 inhibitor (USP24-i-101). USP24 inhibition decreased lipid droplet accumulation in adipocytes and hepatocytes, suppressed inflammation in hepatocytes and AML12 cells, and reduced fibrosis in hepatic stellate cells. Mechanistically, USP24 expression was upregulated by PKA activation during adipocyte differentiation, leading to increased PKA-Cα stability and CREB phosphorylation, which promoted lipogenic gene expression. Free fatty acids (FFA) increased USP24 expression, activating NF-κB and TGFβ pathways to induce inflammation (Cox2) and fibrosis (α-SMA). USP24 was highly expressed in patients with metabolic dysfunction-associated steatohepatitis (MASH) and correlated with Cox2 and α-SMA levels.
Collapse
Affiliation(s)
- Beh Ning
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan, 701, Taiwan
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Shao-An Wang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ming-Jer Young
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan, 701, Taiwan
| | - Yung-Ching Chen
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan, 701, Taiwan
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yun Hung
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Tran Thu Huong
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan, 701, Taiwan
| | - Wen-Chang Chang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yi-Ching Wang
- Institute of Pharmacology, National Cheng Kung University, Tainan, Taiwan
| | - Ming-Lung Yu
- Center of Excellence for Metabolic Associated Fatty Liver Disease, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Kai-Cheng Hsu
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Jan-Jong Hung
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan, 701, Taiwan.
| |
Collapse
|
6
|
Cai WF, Chen QC, Ni Q, Liu L, Liu Q, Yi YK, Jiang CP, Shen CY. p-Synephrine ameliorates non-alcoholic fatty liver disease by regulating liver-adipose axis via AMPK/NF-kappa B pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 348:119890. [PMID: 40294666 DOI: 10.1016/j.jep.2025.119890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 02/20/2025] [Accepted: 04/25/2025] [Indexed: 04/30/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Citrus aurantium L. var. amara Engl. is a folk medicine and dietary supplement popularly used in alleviating indigestion due to food retention and obesity. p-Synephrine, a principal proto-alkaloid in Citrus aurantium L. var. amara Engl., is extensively utilized due to its numerous benefits, particularly its potential to ameliorate obesity. Previous studies of our research demonstrated that p-synephrine had the potential to alleviate insulin resistance (IR) and liver lipid accumulation caused by high-fat diet (HFD), as well as enlargement of cells in adipose tissue. However, the effects of p-synephrine in ameliorating non-alcoholic fatty liver disease (NAFLD) were still unclear. AIM OF STUDY To explore the effects of p-synephrine on HFD-induced NAFLD and its mechanisms. METHODS NAFLD mice were developed by HFD feeding and treated with p-synephrine once a day for 21 weeks. The protective effects of p-synephrine against NAFLD and its mechanisms were evaluated by OGTT, ITT, biochemical index measurements, H&E, immunofluorescence, Sirius red staining, oil red O staining, immunohistochemistry, RT-qPCR, Western blot, network pharmacology, and molecular docking assays. RESULTS The results of network pharmacology suggested that AMPK-α1 might be the core target, and AMPK and insulin signaling pathways might be the key regulatory pathways of p-synephrine to alleviate NAFLD. Molecular docking confirmed AMPK-α1 as a probable direct molecular target. p-Synephrine significantly reduced HFD-induced weight gain of the body, liver, and iWAT. It improved glucose tolerance, insulin tolerance and lipid metabolism disorders caused by HFD. Serum levels of NO, TNF-α, and IL-6 in NAFLD mice were suppressed. AST, ALT, and HYP levels in serum and liver were inhibited. Morphological observation showed p-synephrine alleviated hepatic steatosis and fibrosis. p-Synephrine administration also significantly inhibited hepatic de novo lipogenesis (DNL), as evidenced by its regulation of non-esterified fatty acid (NEFA) and TG contents, as well as SREBP-1c, FASN, and ACC1 mRNA expression levels. p-Synephrine also reversed HFD-induced histopathological changes in iWAT, promoted iWAT browning by increasing UCP1 and PGC-1α expression. Simultaneously, p-synephrine intervention markedly increased phosphorylation levels of IRS-1, PI3K, and Akt, and protein expression of GLUT-4 in iWAT and liver. Expression of TNF-α, IL-6, and IL-1β and NF-κB activation in iWAT and liver were attenuated through the treatment of p-synephrine. Further assays showed that p-synephrine intervention potently regulated AMPK pathway in iWAT and liver of mice. CONCLUSION This investigation proposed that p-synephrine had the potential to ameliorate HFD-induced NAFLD by regulating liver-adipose axis through AMPK/NF-κB pathway.
Collapse
Affiliation(s)
- Wei-Feng Cai
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, 510515, China; Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Guangzhou, 510515, China
| | - Qi-Cong Chen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, 510515, China; Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Guangzhou, 510515, China
| | - Qian Ni
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, 510515, China; Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Guangzhou, 510515, China
| | - Li Liu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, 510515, China; Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Guangzhou, 510515, China
| | - Qiang Liu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, 510515, China; Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Guangzhou, 510515, China
| | - Yan-Kui Yi
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, 510515, China; Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Guangzhou, 510515, China
| | - Cui-Ping Jiang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, 510515, China; Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Guangzhou, 510515, China.
| | - Chun-Yan Shen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, 510515, China; Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Guangzhou, 510515, China.
| |
Collapse
|
7
|
Karim R, Teng W, Behram CD, Lin H. SIRT2-mediated ACSS2 K271 deacetylation suppresses lipogenesis under nutrient stress. eLife 2025; 13:RP97019. [PMID: 40331334 PMCID: PMC12058118 DOI: 10.7554/elife.97019] [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] [Indexed: 05/08/2025] Open
Abstract
De novo lipogenesis is associated with the development of human diseases such as cancer, diabetes, and obesity. At the core of lipogenesis lies acetyl coenzyme A (CoA), a metabolite that plays a crucial role in fatty acid synthesis. One of the pathways contributing to the production of cytosolic acetyl-CoA is mediated by acetyl-CoA synthetase 2 (ACSS2). Here, we reveal that when cells encounter nutrient stress, particularly a deficiency in amino acids, Sirtuin 2 (SIRT2) catalyzes the deacetylation of ACSS2 at the lysine residue K271. This results in K271 ubiquitination and subsequently proteasomal degradation of ACSS2. Substitution of K271 leads to decreased ubiquitination of ACSS2, increased ACSS2 protein level, and thus increased lipogenesis. Our study uncovers a mechanism that cells employ to efficiently manage lipogenesis during periods of nutrient stress.
Collapse
Affiliation(s)
- Rezwana Karim
- Department of Chemistry and Chemical Biology, Cornell UniversityIthacaUnited States
| | - Wendi Teng
- Department of Chemistry and Chemical Biology, Cornell UniversityIthacaUnited States
| | - Cameron D Behram
- Department of Chemistry and Chemical Biology, Cornell UniversityIthacaUnited States
| | - Hening Lin
- Department of Chemistry and Chemical Biology, Cornell UniversityIthacaUnited States
- Howard Hughes Medical Institute; Department of Chemistry and Chemical Biology; Department of Molecular Biology and Genetics, Cornell UniversityIthacaUnited States
- Howard Hughes Medical Institute; Department of Medicine and Department of Chemistry, The University of ChicagoChicagoUnited States
| |
Collapse
|
8
|
Xiao Y, Ai M, Miao J, Yan S, Du Y, Zhang J, Tang C, Zhang K. Effects of chili meal supplementation on productive performance, intestinal health, and liver lipid metabolism of laying hens fed low-protein diets. Poult Sci 2025; 104:105001. [PMID: 40073638 PMCID: PMC11950995 DOI: 10.1016/j.psj.2025.105001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 03/01/2025] [Accepted: 03/05/2025] [Indexed: 03/14/2025] Open
Abstract
This study aimed to explore the effects of chili meal (CM), a by-product of chili pepper oil extraction, on the productive performance, intestinal health, and lipid metabolism of laying hens fed low-protein (LP) diets. A total of 384 Hy-Line brown laying hens (32 weeks old) were divided into six groups: control (CON) diet with 16.5 % crude protein (CP), LP diet with 15 % CP, and LP diets supplemented with 3 %, 5 %, 7 %, and 9 % CM. Results showed that dietary CM supplementation of up to 5 % did not negatively affect the productive performance of laying hens fed LP diets. However, the groups receiving 7 % and 9 % CM exhibited a significant increase in the feed-to-egg ratio (P < 0.05). Additionally, dietary CM supplementation effectively enhanced egg yolk color in a dose-dependent manner (P < 0.05). Intestinal morphology analysis indicated that the 5 % CM group had a higher villus height-to-crypt depth ratio than the LP and 9 % CM groups (P < 0.05), with no significant differences among the other groups. Dietary supplementation with 3 %-7 % CM did not significantly affect serum and jejunal antioxidant capacity, and the 9 % CM group exhibited the highest levels of serum and jejunal malondialdehyde among the groups (P < 0.05). Dietary CM supplementation significantly increased anti-inflammatory cytokines (IL-4 and IL-10) and decreased pro-inflammatory cytokines (IL-1β, IL-6, and TNF-α) in the serum and jejunal tissue of laying hens (P < 0.05). Moreover, CM supplementation significantly altered the cecal microbiota composition in laying hens, increasing the abundance of beneficial bacteria, such as Desulfovibrio and Megamonas. Furthermore, dietary CM supplementation significantly decreased serum triglyceride levels; downregulated liver mRNA levels of ACC, FAS, and SREBP-1C/2; and upregulated the mRNA levels of ACOX1, PPAR-α, Apob, and CPT in laying hens fed LP diets. In conclusion, CM supplementation should not exceed 5 % to avoid negative impacts on performance while supporting intestinal health and lipid metabolism.
Collapse
Affiliation(s)
- Yudi Xiao
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, PR China; State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China
| | - Mingming Ai
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, PR China
| | - Junhong Miao
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, PR China
| | - Shuhui Yan
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, PR China
| | - Yifan Du
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, PR China
| | - Junmin Zhang
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China
| | - Chaohua Tang
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China.
| | - Kai Zhang
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, PR China.
| |
Collapse
|
9
|
Ma X, Ye Z, Li M, Wei W, Chen J, Zhang L. Cold-Induced DHRS4 Promotes Thermogenesis via Enhanced Fatty Acid β-Oxidation in Porcine Subcutaneous Adipocytes. Animals (Basel) 2025; 15:1190. [PMID: 40362005 PMCID: PMC12071078 DOI: 10.3390/ani15091190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 04/19/2025] [Accepted: 04/21/2025] [Indexed: 05/15/2025] Open
Abstract
Adipose tissue exhibits remarkable plasticity in adapting to thermal stress, yet the epigenetic mechanisms coordinating metabolic reprogramming in large mammals-particularly in livestock species lacking classical brown adipose tissue (BAT) such as swine-remain elusive. Using a porcine cold exposure model, we investigated adipose adaptation mechanisms through integrated single-cell RNA sequencing and bulk transcriptomic analyses of subcutaneous adipose tissue (subWAT). We identified a cold-induced thermogenic adipocyte subpopulation, characterized by upregulated DHRS4 expression. Mechanistically, cold exposure induced hypomethylation at the DHRS4 promoter locus, enhancing its expression to potentiate fatty acid β-oxidation, accompanied by thermogenic capacity upregulation. Our findings establish DHRS4 as an epigenetic-metabolic switch governing cold adaptation and a potential target for improving cold resistance in swine production systems.
Collapse
Affiliation(s)
| | | | | | | | | | - Lifan Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (X.M.); (Z.Y.); (M.L.); (W.W.); (J.C.)
| |
Collapse
|
10
|
Sharma S, Tiwari N, Tanwar SS. The current findings on the gut-liver axis and the molecular basis of NAFLD/NASH associated with gut microbiome dysbiosis. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04069-z. [PMID: 40202676 DOI: 10.1007/s00210-025-04069-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 03/17/2025] [Indexed: 04/10/2025]
Abstract
Recent research has highlighted the complex relationship between gut microbiota, metabolic pathways, and nonalcoholic fatty liver disease (NAFLD) progression. Gut dysbiosis, commonly observed in NAFLD patients, impairs intestinal permeability, leading to the translocation of bacterial products like lipopolysaccharides, short-chain fatty acids, and ethanol to the liver. These microbiome-associated mechanisms contribute to intestinal and hepatic inflammation, potentially advancing NAFLD to NASH. Dietary habits, particularly those rich in saturated fats and fructose, can modify the microbiome composition, leading to dysbiosis and fatty liver development. Metabolomic approaches have identified unique profiles in NASH patients, with specific metabolites like ethanol linked to disease progression. While bariatric surgery has shown promise in preventing NAFLD progression, the role of gut microbiome and metabolites in this improvement remains to be proven. Understanding these microbiome-related pathways may provide new diagnostic and therapeutic targets for NAFLD and NASH. A comprehensive review of current literature was conducted using multiple medical research databases, including PubMed, Scopus, Web of Science, Embase, Cochrane Library, ClinicalTrials.gov, ScienceDirect, Medline, ProQuest, and Google Scholar. The review focused on studies that examine the relationship between gut microbiota composition, metabolic pathways, and NAFLD progression. Key areas of interest included microbial dysbiosis, endotoxin production, and the influence of diet on gut microbiota. The analysis revealed that gut dysbiosis contributes to NAFLD through several mechanisms, diet significantly influences gut microbiota composition, which in turn affects liver function through the gut-liver axis. High-fat diets can lead to dysbiosis, altering microbial metabolic activities and promoting liver inflammation. Specifically, gut microbiota-mediated generation of saturated fatty acids, such as palmitic acid, can activate liver macrophages and increase TNF-α expression, contributing to NASH development. Different dietary components, including cholesterol, fiber, fat, and carbohydrates, can modulate the gut microbiome and influence NAFLD progression. This gut-liver axis plays a crucial role in maintaining immune homeostasis, with the liver responding to gut-derived bacteria by activating innate and adaptive immune responses. Microbial metabolites, such as bile acids, tryptophan catabolites, and branched-chain amino acids, regulate adipose tissue and intestinal homeostasis, contributing to NASH pathogenesis. Additionally, the microbiome of NASH patients shows an elevated capacity for alcohol production, suggesting similarities between alcoholic steatohepatitis and NASH. These findings indicate that targeting the gut microbiota may be a promising approach for NASH treatment and prevention. Recent research highlights the potential of targeting gut microbiota for managing nonalcoholic fatty liver disease (NAFLD). The gut-liver axis plays a crucial role in NAFLD pathophysiology, with dysbiosis contributing to disease progression. Various therapeutic approaches aimed at modulating gut microbiota have shown promise, including probiotics, prebiotics, synbiotics, fecal microbiota transplantation, and dietary interventions. Probiotics have demonstrated efficacy in human randomized controlled trials, while other interventions require further investigation in clinical settings. These microbiota-targeted therapies may improve NAFLD outcomes through multiple mechanisms, such as reducing inflammation and enhancing metabolic function. Although lifestyle modifications remain the primary recommendation for NAFLD management, microbiota-focused interventions offer a promising alternative for patients struggling to achieve weight loss targets.
Collapse
Affiliation(s)
- Seema Sharma
- Department of Pharmacy, Shri Vaishnav Vidyapeeth Vishwavidyalaya, Indore, M.P, India
| | - Nishant Tiwari
- Acropolis Institute of Pharmaceutical Education and Research, Indore, M.P, India
| | - Sampat Singh Tanwar
- Department of Pharmacy, Shri Vaishnav Vidyapeeth Vishwavidyalaya, Indore, M.P, India.
| |
Collapse
|
11
|
Khaliq A, Badshah H, Shah Y. Combination therapy with vitamin E and ertugliflozin in patients with non-alcoholic fatty liver disease and type 2 diabetes mellitus: a randomized clinical trial. Ir J Med Sci 2025:10.1007/s11845-025-03945-0. [PMID: 40202706 DOI: 10.1007/s11845-025-03945-0] [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: 07/12/2024] [Accepted: 03/25/2025] [Indexed: 04/10/2025]
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) is a common comorbidity in type 2 diabetes mellitus (T2DM), with shared pathophysiological mechanisms, including insulin resistance, oxidative stress, and inflammation. OBJECTIVES This study evaluates the effects of vitamin E and ertugliflozin, individually and in combination, alongside standard pioglitazone therapy, on hepatic and metabolic parameters in patients with NAFLD and T2DM. METHODS A 24-week, double-blind, randomized, controlled clinical trial on 173 patients with NAFLD and T2DM was assigned into four groups: vitamin E (n = 42), pioglitazone (n = 43), ertugliflozin (n = 44), and vitamin E + ertugliflozin (n = 44) combination therapy. The primary outcome was to monitor changes in liver steatosis assessed via ultrasound. Secondary outcomes included evaluation of liver enzymes, glycemic control, fibrosis markers, and lipid profiles. RESULTS Combination therapy of vitamin E + ertugliflozin showed the highest decrease in liver fat content, with 11 participants achieving successful Grade 0 (p < 0.001). Significant improvements were also observed in glycemic control, HbA1c, triglycerides, and liver enzymes. Ertugliflozin monotherapy showed significant efficacy in improving liver enzymes, glycemic parameters, and fibrosis markers. Pioglitazone improved the initial stage of NAFLD but had a limited impact on advanced fibrosis. Ertugliflozin, in combination with vitamin E, decreases oxidative stress; however, vitamin E by itself has no impact on the metabolic and fibrosis index. CONCLUSION The ertugliflozin and vitamin E combination is a very effective treatment for patients with NAFLD and T2DM. It improves hepatic steatosis and metabolic indicators. Exploration is required for combination therapy in order to assess the prolonged efficacy and safety of the treatment.
Collapse
Affiliation(s)
- Adil Khaliq
- Department of Pharmacy, Abdul Wali Khan University, Mardan, Pakistan
| | - Haroon Badshah
- Department of Pharmacy, Abdul Wali Khan University, Mardan, Pakistan.
| | - Yasar Shah
- Department of Pharmacy, Abdul Wali Khan University, Mardan, Pakistan
| |
Collapse
|
12
|
Ding Z, Wang L, Sun J, Zheng L, Tang Y, Tang H. Hepatocellular carcinoma: pathogenesis, molecular mechanisms, and treatment advances. Front Oncol 2025; 15:1526206. [PMID: 40265012 PMCID: PMC12011620 DOI: 10.3389/fonc.2025.1526206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 03/21/2025] [Indexed: 04/24/2025] Open
Abstract
Hepatocellular Carcinoma (HCC), a highly prevalent malignancy, poses a significant global health challenge. Its pathogenesis is intricate and multifactorial, involving a complex interplay of environmental and genetic factors. Viral hepatitis, excessive alcohol consumption, and cirrhosis are known to significantly elevate the risk of developing HCC. The underlying biological processes driving HCC are equally complex, encompassing aberrant activation of molecular signaling pathways, dysregulation of hepatocellular differentiation and angiogenesis, and immune dysfunction. This review delves into the multifaceted nature of HCC, exploring its etiology and the intricate molecular signaling pathways involved in its development. We examine the role of immune dysregulation in HCC progression and discuss the potential of emerging therapeutic strategies, including immune-targeted therapy and tumor-associated macrophage interventions. Additionally, we explore the potential of traditional Chinese medicine (TCM) monomers in inhibiting tumor growth. By elucidating the complex interplay of factors contributing to HCC, this review aims to provide a comprehensive understanding of the disease and highlight promising avenues for future research and therapeutic development.
Collapse
Affiliation(s)
- Zhixian Ding
- General Clinical Research Center, Wanbei Coal-Electricity Group General Hospital, Suzhou, China
- Laboratory of Inflammation and Repair of Liver Injury and Tumor Immunity, Wanbei Coal-Electricity Group General Hospital, Hefei, China
| | - Lusheng Wang
- General Clinical Research Center, Wanbei Coal-Electricity Group General Hospital, Suzhou, China
- Laboratory of Inflammation and Repair of Liver Injury and Tumor Immunity, Wanbei Coal-Electricity Group General Hospital, Hefei, China
| | - Jiting Sun
- General Clinical Research Center, Wanbei Coal-Electricity Group General Hospital, Suzhou, China
- Laboratory of Inflammation and Repair of Liver Injury and Tumor Immunity, Wanbei Coal-Electricity Group General Hospital, Hefei, China
| | - Lijie Zheng
- General Clinical Research Center, Wanbei Coal-Electricity Group General Hospital, Suzhou, China
- Laboratory of Inflammation and Repair of Liver Injury and Tumor Immunity, Wanbei Coal-Electricity Group General Hospital, Hefei, China
| | - Yu Tang
- General Clinical Research Center, Wanbei Coal-Electricity Group General Hospital, Suzhou, China
- Laboratory of Inflammation and Repair of Liver Injury and Tumor Immunity, Wanbei Coal-Electricity Group General Hospital, Hefei, China
| | - Heng Tang
- General Clinical Research Center, Wanbei Coal-Electricity Group General Hospital, Suzhou, China
- Laboratory of Inflammation and Repair of Liver Injury and Tumor Immunity, Wanbei Coal-Electricity Group General Hospital, Hefei, China
| |
Collapse
|
13
|
Huang SH, Tulegenov D, Shvets G. Combining quantum cascade lasers and plasmonic metasurfaces to monitor de novo lipogenesis with vibrational contrast microscopy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.30.646207. [PMID: 40236123 PMCID: PMC11996395 DOI: 10.1101/2025.03.30.646207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
The combination of a tunable quantum cascade laser (QCL) and plasmonic mid-infrared (MIR) metasurface is a powerful tool enabling label-free high-content microscopy of hydrated cells using the vibrational contrast of their constituent biomolecules. While the QCL provides a high-brightness source whose frequency can be rapidly tuned to that of the relevant molecular vibration, the metasurface is used to overcome water absorption of MIR light. Here we employ the resulting Metasurface-enabled Inverted Reflected-light Infrared Absorption Microscopy (MIRIAM) tool for non-destructive monitoring of the vital process of de novo lipogenesis (DNL), by which fat tissue cells (adipocytes) synthesize fatty acids from glucose and store them inside lipid droplets. Using 13 C-labeled glucose as a metabolic probe, we produce spatially- and temporally-resolved images of 13 C incorporation into lipids and proteins, observed as red-shifted vibrational peaks in the MIR spectra. These findings demonstrate MIRIAM's capability for studying metabolic pathways with molecular specificity, offering a powerful platform for label-free imaging of cellular metabolism.
Collapse
|
14
|
Zhang Y, Yang J, Min J, Huang S, Li Y, Liu S. The emerging role of E3 ubiquitin ligases and deubiquitinases in metabolic dysfunction-associated steatotic liver disease. J Transl Med 2025; 23:368. [PMID: 40133964 PMCID: PMC11938720 DOI: 10.1186/s12967-025-06255-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 02/17/2025] [Indexed: 03/27/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most common chronic liver disease worldwide, with a prevalence as high as 32.4%. MASLD encompasses a spectrum of liver pathologies, ranging from steatosis to metabolic dysfunction-associated steatohepatitis (MASH), fibrosis, and, in some cases, progression to end-stage liver disease (cirrhosis and hepatocellular carcinoma). A comprehensive understanding of the pathogenesis of this highly prevalent liver disease may facilitate the identification of novel targets for the development of improved therapies. E3 ubiquitin ligases and deubiquitinases (DUBs) are key regulatory components of the ubiquitin‒proteasome system (UPS), which plays a pivotal role in maintaining intracellular protein homeostasis. Emerging evidence implicates that aberrant expression of E3 ligases and DUBs is involved in the progression of MASLD. Here, we review abnormalities in E3 ligases and DUBs by (1) discussing their targets, mechanisms, and functions in MASLD; (2) summarizing pharmacological interventions targeting these enzymes in preclinical and clinical studies; and (3) addressing challenges and future therapeutic strategies. This review synthesizes current evidence to highlight the development of novel therapeutic strategies based on the UPS for MASLD and progressive liver disease.
Collapse
Affiliation(s)
- Yu Zhang
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, CSU-Sinocare Research Center for Nutrition and Metabolic Health, Furong Laboratory, Changsha, Hunan, 410011, China
| | - Jiahui Yang
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, CSU-Sinocare Research Center for Nutrition and Metabolic Health, Furong Laboratory, Changsha, Hunan, 410011, China
| | - Jiali Min
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, CSU-Sinocare Research Center for Nutrition and Metabolic Health, Furong Laboratory, Changsha, Hunan, 410011, China
| | - Shan Huang
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, CSU-Sinocare Research Center for Nutrition and Metabolic Health, Furong Laboratory, Changsha, Hunan, 410011, China
| | - Yuchen Li
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, CSU-Sinocare Research Center for Nutrition and Metabolic Health, Furong Laboratory, Changsha, Hunan, 410011, China
| | - Shanshan Liu
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, CSU-Sinocare Research Center for Nutrition and Metabolic Health, Furong Laboratory, Changsha, Hunan, 410011, China.
| |
Collapse
|
15
|
Wan D, Lee JE, Park YK, Maisto S, Agyapong C, Ozato K, Gavrilova O, Ge K. Histone chaperone HIRA facilitates transcription elongation to regulate insulin sensitivity and obesity-associated adipose expansion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.21.644577. [PMID: 40196683 PMCID: PMC11974756 DOI: 10.1101/2025.03.21.644577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Adipose tissue is essential for maintaining glucose and lipid homeostasis in mammals. The histone chaperone HIRA has been reported to play a lineage- and stage-selective role during development. However, its role in adipose tissue development and function as well as its working mechanism remain unknown. Here we show that tissue-specific knockout of histone chaperone HIRA in mice impairs insulin sensitivity and alleviates adipose tissue expansion during high-fat diet-induced obesity, but only moderately affects embryonic development of adipose tissue. Mechanistically, HIRA is selectively required for expression of genes critical for insulin response and lipogenesis, rather than adipogenesis, in adipose tissue. By acute depletion of HIRA protein and by mapping HIRA genomic localization in adipocytes, we demonstrate that HIRA binds to promoters and enhancers of insulin response and lipogenesis genes and regulates their expression by facilitating transcription elongation. Our findings not only identify HIRA as an epigenomic regulator of insulin sensitivity, lipogenesis, and obesity-associated adipose expansion, but also reveal a novel mechanism by which HIRA regulates transcription.
Collapse
Affiliation(s)
- Danyang Wan
- Adipocyte Biology and Gene Regulation Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ji-Eun Lee
- Adipocyte Biology and Gene Regulation Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Young-Kwon Park
- Adipocyte Biology and Gene Regulation Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Susanna Maisto
- Adipocyte Biology and Gene Regulation Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Christabelle Agyapong
- Adipocyte Biology and Gene Regulation Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Keiko Ozato
- Division of Developmental Biology, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Oksana Gavrilova
- Mouse Metabolism Core, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Kai Ge
- Adipocyte Biology and Gene Regulation Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
16
|
Balbuena-Pecino S, Riera-Heredia N, Sánchez-Moya A, Perelló-Amorós M, Gutiérrez J, Capilla E, Navarro I. Screening the effects of phytoestrogens on lipid metabolism in primary cultured adipocytes from rainbow trout (Oncorhynchus mykiss) and gilthead sea bream (Sparus aurata). FISH PHYSIOLOGY AND BIOCHEMISTRY 2025; 51:71. [PMID: 40131537 PMCID: PMC11937063 DOI: 10.1007/s10695-025-01483-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 03/05/2025] [Indexed: 03/27/2025]
Abstract
Aquafeed formulation has progressively reduced its dependence on fish-derived ingredients over the past decades. Plant-based substitutes have been a major focus, with soybean meal and its derivatives leading the way. However, many plants contain phytoestrogens, which may affect fish physiology. This study aimed to assess in vitro the effects of genistein (GE), daidzein (DZN), glycitein (GLY), and coumestrol (COU) on the lipid metabolism of rainbow trout (Oncorhynchus mykiss) and gilthead sea bream (Sparus aurata). Primary cultured adipocytes were incubated with these phytoestrogens, along with 17β-estradiol, at two doses each (1, 10, or 100 μM). The 100 μM dose of GE and DZN decreased adipocyte viability, and mainly enhanced lipid accumulation in both species, suggesting a hypertrophic condition. However, the reduction in adipocyte number and lipid content with 100 μM DZN in rainbow trout indicated a limiting effect on adipose tissue growth in this species. Interestingly, COU significantly increased cell viability in gilthead sea bream, potentially leading to hyperplastic growth, a more favorable metabolic state. In that species, which proved to be more phytoestrogens-sensitive, lipoprotein lipase was generally downregulated upon treatments. Moreover, 10 µM GE significantly decreased the mRNA levels of fatty acid transport protein 1 and fatty acid synthase, and increased those of fatty acid binding protein 1, suggesting an acceleration of the differentiation process compared to the control cells. This work provides new insights into how dietary phytoestrogens modulate fish lipid metabolism and supports that their presence in plant protein feedstuffs can potentially affect fish health and production performance.
Collapse
Affiliation(s)
- Sara Balbuena-Pecino
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, 08028, Barcelona, Spain
| | - Natàlia Riera-Heredia
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, 08028, Barcelona, Spain
| | - Albert Sánchez-Moya
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, 08028, Barcelona, Spain
| | | | - Joaquim Gutiérrez
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, 08028, Barcelona, Spain
| | - Encarnación Capilla
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, 08028, Barcelona, Spain
| | - Isabel Navarro
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, 08028, Barcelona, Spain.
| |
Collapse
|
17
|
Yi T, Wu S, Yang Y, Li X, Yang S, Zhang Y, Zhang L, Hu Y, Zhang G, Li J, Yang D. Single-nucleus RNA sequencing reveals dynamic changes in the microenvironment of visceral adipose tissue and metabolic characteristics after cold exposure. Front Endocrinol (Lausanne) 2025; 16:1562431. [PMID: 40196457 PMCID: PMC11973077 DOI: 10.3389/fendo.2025.1562431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 03/04/2025] [Indexed: 04/09/2025] Open
Abstract
Introduction Visceral adipose tissue (VAT) plays a crucial role in regulating systemic metabolic balance. Excess accumulation of VAT is closely associated with various metabolic disorders, a process that involves the coordinated actions of multiple cell types within the tissue. Cold exposure, as a potential intervention, has been proposed to improve metabolic dysfunction. However, the heterogeneity of VAT and its comprehensive metabolic characteristics under cold exposure remain unclear. Methods We collected epididymal white adipose tissue (eWAT) of C57BL/6J mice after cold exposure at three different time points for single-nucleus RNA sequencing (snRNA-seq) analysis. Results We successfully identified ten major cell types in eWAT, enabling understanding of the dynamic changes in the eWAT microenvironment and its metabolic features during cold exposure. This study revealed that cold exposure for 1 day reduced cellular metabolic activity and intercellular communication in eWAT including receptor-ligand-based cell communication and metabolite-mediated interactions. However, after 14 days of cold acclimation, the metabolic activity of adipocytes was significantly enhanced, and intercellular metabolic communication was restored. Additionally, prolonged cold exposure promoted the formation of a distinct adipocyte subpopulation that may be associated with UCP1-independent thermogenesis. These changes may be a new homeostatic state established by VAT to adapt to the cold environment. The study also identified the importance of adipocytes, adipose stem and progenitor cells, myeloid cells, and endothelial cells in the process of cold adaptation. Discussion This research provides valuable insights into the cellular heterogeneity, adipocyte remodeling, and metabolic reprogramming in eWAT after cold exposure. It highlights the critical role of transcriptional dynamics in eWAT during cold exposure and provides new perspectives on the prevention and treatment of metabolic diseases.
Collapse
Affiliation(s)
- Ting Yi
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, China
- Academy of Military Medical Sciences, Academy of Military Sciences, Tianjin, China
| | - Shuai Wu
- Academy of Military Medical Sciences, Academy of Military Sciences, Tianjin, China
| | - Yusha Yang
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, China
- Academy of Military Medical Sciences, Academy of Military Sciences, Tianjin, China
| | - Xi Li
- Academy of Military Medical Sciences, Academy of Military Sciences, Tianjin, China
| | - Shuran Yang
- Academy of Military Medical Sciences, Academy of Military Sciences, Tianjin, China
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yongqiang Zhang
- Academy of Military Medical Sciences, Academy of Military Sciences, Tianjin, China
| | - Li Zhang
- Academy of Military Medical Sciences, Academy of Military Sciences, Tianjin, China
| | - Yuyu Hu
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, China
- Academy of Military Medical Sciences, Academy of Military Sciences, Tianjin, China
| | - Guanyu Zhang
- Academy of Military Medical Sciences, Academy of Military Sciences, Tianjin, China
| | - Jun Li
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, China
| | - Danfeng Yang
- Academy of Military Medical Sciences, Academy of Military Sciences, Tianjin, China
| |
Collapse
|
18
|
Zhang Z, Liu Y, Yu T, Liu Z. Unraveling the Complex Nexus of Macrophage Metabolism, Periodontitis, and Associated Comorbidities. J Innate Immun 2025; 17:211-225. [PMID: 40058341 PMCID: PMC11968099 DOI: 10.1159/000542531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 11/07/2024] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND Periodontitis is recognized as one of the most prevalent oral dysbiotic inflammatory diseases, ultimately leading to the irreversible destruction of periodontal tissues. Macrophages play a pivotal role in the development and progression of periodontitis, and the feasibility of targeting them therapeutically has been established. Since metabolic switching significantly contributes to macrophage regulation, conducting an in-depth review of macrophage metabolism in periodontitis may serve as the foundation for developing innovative treatments. SUMMARY This paper has been carefully reviewed to provide a comprehensive overview of the roles played by macrophages in periodontitis and associated comorbidities. Initially, detailed presentations on the metabolic reprogramming of macrophages, including glucose, lipid, and amino acid metabolism, were provided. Subsequently, dominating macrophage phenotype and metabolism under lipopolysaccharide (LPS) stimulation or during periodontitis were presented with emphasize on critical molecules involved. Furthermore, in recognition of the close association between periodontitis and several comorbidities, the interaction among macrophage metabolism, periodontitis, and related metabolic diseases, was thoroughly discussed. KEY MESSAGES Through the examination of current research on macrophage metabolic reprogramming induced by periodontitis, this review provides potential immunometabolic therapeutic targets for the future and raises many important, yet unstudied, subjects for follow-up. BACKGROUND Periodontitis is recognized as one of the most prevalent oral dysbiotic inflammatory diseases, ultimately leading to the irreversible destruction of periodontal tissues. Macrophages play a pivotal role in the development and progression of periodontitis, and the feasibility of targeting them therapeutically has been established. Since metabolic switching significantly contributes to macrophage regulation, conducting an in-depth review of macrophage metabolism in periodontitis may serve as the foundation for developing innovative treatments. SUMMARY This paper has been carefully reviewed to provide a comprehensive overview of the roles played by macrophages in periodontitis and associated comorbidities. Initially, detailed presentations on the metabolic reprogramming of macrophages, including glucose, lipid, and amino acid metabolism, were provided. Subsequently, dominating macrophage phenotype and metabolism under lipopolysaccharide (LPS) stimulation or during periodontitis were presented with emphasize on critical molecules involved. Furthermore, in recognition of the close association between periodontitis and several comorbidities, the interaction among macrophage metabolism, periodontitis, and related metabolic diseases, was thoroughly discussed. KEY MESSAGES Through the examination of current research on macrophage metabolic reprogramming induced by periodontitis, this review provides potential immunometabolic therapeutic targets for the future and raises many important, yet unstudied, subjects for follow-up.
Collapse
Affiliation(s)
- Zihan Zhang
- The State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yi Liu
- The State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China,
| | - Tian Yu
- Department of Stomatology, Nanbu Country People's Hospital, Nanchong, China
| | - Zhen Liu
- Department of Stomatology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| |
Collapse
|
19
|
Lv H, Xu X, Wu Z, Lin Y, Liu Y, Liu M, Xu L, Wang X, Sun N, Abdel-Shafy H, Abdelrahman M, Alsaegh AA, Ahmed AE, Yang L, Hua G. Yes-associated protein 1 is essential for maintaining lactation via regulating mammary epithelial cell dynamics and secretion capacity. Int J Biol Macromol 2025; 293:139290. [PMID: 39743110 DOI: 10.1016/j.ijbiomac.2024.139290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/08/2024] [Accepted: 12/27/2024] [Indexed: 01/04/2025]
Abstract
Understanding the physiology and molecular mechanisms of lactogenesis is crucial for enhancing mammalian milk production. Yes-associated protein 1 (YAP1) regulated mammary epithelial cell survival during pregnancy, but its role in lactation maintenance remains unclear. We found that YAP1 was highly expressed in mammary gland across specie, with elevated expression levels during murine gestation and lactation, particularly localized in alveoli epithelial cells. In vivo administration of a YAP1 inhibitor impaired murine milk yield, mammary gland weight, alveolar structure, and mammary epithelial cell dynamics. In vitro, YAP1 positively affected mammary epithelial cell growth and the synthesis of triglyceride and α-casein. Notably, the primary lactogenesis hormone Prolactin induced cell growth and triglyceride secretion while enhancing YAP1 expression and activity. In contrast, Melatonin inhibited cell growth and triglyceride synthesis, decreasing YAP1 expression and activity. YAP1 knockdown compromised prolactin induced effects, whereas YAP1 overexpression partially rescued cell functions inhibited by melatonin. Finally, Bioinformatics analyses revealed that YAP1 regulated multiple biological processes related to lactogenesis, including cell cycle, apoptosis, endoplasmic reticulum, amino acid transport and biosynthesis, etc. These finding indicated that YAP1 is essential for mammary epithelial cells growth and secretion and played an essential role in the lactating endocrine network by mediating key hormone functions.
Collapse
Affiliation(s)
- Haimiao Lv
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518120, China; Shenzhen Institute of Nutrition and Health, Huazhong Agricultural University, Shenzhen 518038, China
| | - Xiaoling Xu
- Laboratory of Animal Reproduction, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, BJ, China
| | - Zihui Wu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Yuxin Lin
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Yan Liu
- Laboratory of Animal Reproduction, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, BJ, China
| | - Miaoyu Liu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Linghua Xu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Xiaojie Wang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Nan Sun
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Hamdy Abdel-Shafy
- Department of Animal Production, Faculty of Agriculture, Cairo University, El-Gamma Street, 12613 Giza, Egypt
| | - Mohamed Abdelrahman
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; Animal Production Department, Faculty of Agriculture, Assuit University, Asyut, Egypt
| | - Aiman A Alsaegh
- Department of Laboratory Medicine, Faculty of Applied Medical Sciences, Umm Al-Qura University, Saudi Arabia
| | - Ahmed Ezzat Ahmed
- Department of Biology, College of Science, King Khalid University, P.O. Box 9004, 61413 Abha, Saudi Arabia; Department of Theriogenology, Faculty of Veterinary Medicine, South Valley University, 83523 Qena, Egypt
| | - Liguo Yang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; National Center for International Research on Animal Genetics, Breeding and Reproduction, Frontiers Science Center for Animal Breeding and Sustainable Production, Huazhong Agricultural University, Wuhan 430070, China
| | - Guohua Hua
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518120, China; Shenzhen Institute of Nutrition and Health, Huazhong Agricultural University, Shenzhen 518038, China; National Center for International Research on Animal Genetics, Breeding and Reproduction, Frontiers Science Center for Animal Breeding and Sustainable Production, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
20
|
Goodman JH, Berland C, Soni RK, Ferrante AW. Overfeeding induces adipose tissue release of distinct mitochondria. Cell Rep 2025; 44:115318. [PMID: 39970045 DOI: 10.1016/j.celrep.2025.115318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 11/20/2024] [Accepted: 01/24/2025] [Indexed: 02/21/2025] Open
Abstract
Overfeeding animals beyond what they eat ad libitum causes rapid adipose tissue expansion, leading to an unusual form of obesity characterized by low immune cell accumulation in fat and sustained anorexia. To investigate how overfeeding affects adipose tissue, we studied the protein secretome of fat from equally obese overfed and ad libitum-fed mice. Fat from overfed animals secretes lower amounts of immune regulatory proteins. Unexpectedly, fat from overfed mice releases larger amounts of mitochondrial proteins. Microscopy identified mitochondria in the conditioned medium of cultured fat that were found not within extracellular vesicles but rather as free extracellular organelles. The protein profile of released mitochondria was distinct from the mitochondrial protein profile of the whole fat, suggesting that the metabolic stress of overfeeding leads to the release of a mitochondrial subset favoring de novo lipogenesis. These findings add to growing evidence that cells alter their energy profiles through the release of mitochondria.
Collapse
Affiliation(s)
- Joshua H Goodman
- Division of Preventive Medicine & Nutrition, Naomi Berrie Diabetes Center, Columbia University, New York, NY, USA
| | - Chloé Berland
- Division of Preventive Medicine & Nutrition, Naomi Berrie Diabetes Center, Columbia University, New York, NY, USA
| | - Rajesh K Soni
- Proteomics and Macromolecular Crystallography Shared Resource, Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA; Pathology & Cell Biology, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY 10032, USA
| | - Anthony W Ferrante
- Division of Preventive Medicine & Nutrition, Naomi Berrie Diabetes Center, Columbia University, New York, NY, USA.
| |
Collapse
|
21
|
Paoli A. The Influence of Physical Exercise, Ketogenic Diet, and Time-Restricted Eating on De Novo Lipogenesis: A Narrative Review. Nutrients 2025; 17:663. [PMID: 40004991 PMCID: PMC11858292 DOI: 10.3390/nu17040663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/04/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
De novo lipogenesis (DNL) is a metabolic pathway that converts carbohydrates into fatty acids, primarily occurring in the liver and, to a lesser extent, in adipose tissue. While hepatic DNL is highly responsive to dietary carbohydrate intake and regulated by insulin via transcription factors like SREBP-1c, adipose DNL is more modest and less sensitive to dietary overfeeding. Dysregulated DNL contributes to metabolic disorders, including metabolic dysfunction-associated steatotic liver disease (MASLD). Lifestyle interventions, such as physical exercise, ketogenic diets, and time-restricted eating (TRE) offer promising strategies to regulate DNL and improve metabolic health. Physical exercise enhances glucose uptake in muscles, reduces insulin levels, and promotes lipid oxidation, thereby suppressing hepatic DNL. Endurance and resistance training also improve mitochondrial function, further mitigating hepatic triglyceride accumulation. Ketogenic diets shift energy metabolism toward fatty acid oxidation and ketogenesis, lower insulin, and directly downregulate lipogenic enzyme activity in the liver. TRE aligns feeding with circadian rhythms by optimizing AMP-activated protein kinase (AMPK) activation during fasting periods, which suppresses DNL and enhances lipid metabolism. The combined effects of these interventions demonstrate significant potential for improving lipid profiles, reducing hepatic triglycerides, and preventing lipotoxicity. By addressing the distinct roles of the liver and adipose DNL, these strategies target systemic and localized lipid metabolism dysregulation. Although further research is needed to fully understand their long-term impact, these findings highlight the transformative potential of integrating these approaches into clinical practice to manage metabolic disorders and their associated complications.
Collapse
Affiliation(s)
- Antonio Paoli
- Department of Biomedical Sciences, University of Padua, 35100 Padua, Italy;
- Research Center for High Performance Sport, UCAM Catholic University of Murcia, 30107 Murcia, Spain
| |
Collapse
|
22
|
Fu M, Yoon KS, Ha J, Kang I, Choe W. Crosstalk Between Antioxidants and Adipogenesis: Mechanistic Pathways and Their Roles in Metabolic Health. Antioxidants (Basel) 2025; 14:203. [PMID: 40002389 PMCID: PMC11852089 DOI: 10.3390/antiox14020203] [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: 01/05/2025] [Revised: 02/01/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
The interplay between oxidative stress and adipogenesis is a critical factor in the development of obesity and its associated metabolic disorders. Excessive reactive oxygen species (ROS) disrupt key transcription factors such as peroxisome proliferator-activated receptor gamma (PPARγ) and CCAAT/enhancer-binding protein alpha (C/EBPα), impairing lipid metabolism, promoting adipocyte dysfunction, and exacerbating inflammation and insulin resistance. Antioxidants, classified as endogenous (e.g., glutathione, superoxide dismutase, and catalase) and exogenous (e.g., polyphenols, flavonoids, and vitamins C and E), are pivotal in mitigating these effects by restoring redox balance and preserving adipocyte functionality. Endogenous antioxidants neutralize ROS and safeguard cellular structures; however, under heightened oxidative stress, these defenses are often insufficient, necessitating dietary supplementation. Exogenous antioxidants derived from plant-based sources, such as polyphenols and vitamins, act through direct ROS scavenging, upregulation of endogenous antioxidant enzymes, and modulation of key signaling pathways like nuclear factor kappa B (NF-κB) and PPARγ, reducing lipid peroxidation, inflammation, and adipocyte dysfunction. Furthermore, they influence epigenetic regulation and transcriptional networks to restore adipocyte differentiation and limit lipid accumulation. Antioxidant-rich diets, including the Mediterranean diet, are strongly associated with improved metabolic health, reduced obesity rates, and enhanced insulin sensitivity. Advances in personalized antioxidant therapies, guided by biomarkers of oxidative stress and supported by novel delivery systems, present promising avenues for optimizing therapeutic interventions. This review, "Crosstalk Between Antioxidants and Adipogenesis: Mechanistic Pathways and Their Role in Metabolic Health", highlights the mechanistic pathways by which antioxidants regulate oxidative stress and adipogenesis to enhance metabolic health.
Collapse
Affiliation(s)
- Minghao Fu
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.F.); (K.-S.Y.); (J.H.); (I.K.)
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Kyung-Sik Yoon
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.F.); (K.-S.Y.); (J.H.); (I.K.)
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Joohun Ha
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.F.); (K.-S.Y.); (J.H.); (I.K.)
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Insug Kang
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.F.); (K.-S.Y.); (J.H.); (I.K.)
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Wonchae Choe
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.F.); (K.-S.Y.); (J.H.); (I.K.)
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
23
|
Santos HO, Penha-Silva N. Revisiting the concepts of de novo lipogenesis to understand the conversion of carbohydrates into fats: Stop overvaluing and extrapolating the renowned phrase "fat burns in the flame of carbohydrate". Nutrition 2025; 130:112617. [PMID: 39566326 DOI: 10.1016/j.nut.2024.112617] [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: 11/05/2023] [Revised: 09/16/2024] [Accepted: 10/18/2024] [Indexed: 11/22/2024]
Abstract
Carbohydrates can be converted into fatty acids via de novo lipogenesis (DNL). Although DNL is considered inefficient, these endogenous fatty acids contribute substantially to the esterification pathway in adipose tissue, together with fatty acids of feeding. This article revisited the concepts of DNL and aimed to discuss the clinical magnitude of carbohydrate overfeeding and fat mass accumulation. Although fat storage resulting from fat intake is more favorable for fat mass accrual than carbohydrates due to molecule structure and metabolism (e.g., oxidation and thermic effect), carbohydrates can substantially participate in lipogenesis and esterification under excess carbohydrate intake over time. Regarding only monosaccharide overfeeding, glucose and fructose favor the subcutaneous and visceral adipose tissue, respectively. While fructose and sucrose are considered villains in nonalcoholic fatty liver disease, energy surplus from carbohydrates, regardless of sources, can be considered an underlying cause of obesity. Interestingly, some degree of DNL in adipocytes may be favorable to mitigate a high deposition of fatty acids in the liver, conferring a physiological role. Although "fat burns in the flame of carbohydrate" is a praiseworthy phrase that has helped describe basic concepts in biochemistry for many decades, it appears to be overvalued and extrapolated even nowadays. DNL cannot be neglected. It is time to consider DNL an efficient biochemical process in health and disease.
Collapse
Affiliation(s)
- Heitor O Santos
- School of Medicine, Uberlândia Federal University, Uberlândia, MG, Brazil.
| | - Nilson Penha-Silva
- Institute of Biotechnology, Uberlândia Federal University, Uberlândia, MG, Brazil
| |
Collapse
|
24
|
Lian H, Zhang Y, Zhu Z, Wan R, Wang Z, Yang K, Ma S, Wang Y, Xu K, Cheng L, Zhao W, Li Y, Wang L, Yu G. Fatty acid synthase inhibition alleviates lung fibrosis via β-catenin signal in fibroblasts. Life Sci Alliance 2025; 8:e202402805. [PMID: 39567194 PMCID: PMC11579593 DOI: 10.26508/lsa.202402805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 11/11/2024] [Accepted: 11/11/2024] [Indexed: 11/22/2024] Open
Abstract
Idiopathic pulmonary fibrosis is a progressive and lethal interstitial lung disease with an unclear etiology and limited treatment options. Fatty acid synthase (FASN) plays various roles in metabolic-related diseases. This study demonstrates that FASN expression is increased in fibroblasts from the lung tissues of patients with idiopathic pulmonary fibrosis and in bleomycin-treated mice. In MRC-5 cells, the inhibition of FASN using shRNA or the pharmacological inhibitor C75 resulted in the increased mRNA and protein expression of glycogen synthase kinase 3β and Axin1, both negative regulators of the Wnt/β-catenin signaling pathway, and promoted autophagy. This outcome led to a decrease in β-catenin protein and mRNA levels, effectively inhibiting the proliferation, migration, and differentiation of lung fibroblasts into myofibroblasts, while inducing the differentiation of fibroblasts into adipofibroblasts. In vivo experiments showed that C75 alleviated bleomycin-induced lung fibrosis in mice by inhibiting β-catenin. In conclusion, these findings suggest that inhibiting FASN in fibroblasts may diminish the activity of the Wnt/β-catenin signaling pathway, providing a potential therapeutic avenue for pulmonary fibrosis.
Collapse
Affiliation(s)
- Hui Lian
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Pingyuan Laboratory, College of Life Science, Henan Normal University, Xinxiang, China
| | - Yujie Zhang
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Pingyuan Laboratory, College of Life Science, Henan Normal University, Xinxiang, China
| | - Zhao Zhu
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Pingyuan Laboratory, College of Life Science, Henan Normal University, Xinxiang, China
| | - Ruyan Wan
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Pingyuan Laboratory, College of Life Science, Henan Normal University, Xinxiang, China
| | - Zhixia Wang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Kun Yang
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Pingyuan Laboratory, College of Life Science, Henan Normal University, Xinxiang, China
| | - Shuaichen Ma
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Pingyuan Laboratory, College of Life Science, Henan Normal University, Xinxiang, China
| | - Yaxuan Wang
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Pingyuan Laboratory, College of Life Science, Henan Normal University, Xinxiang, China
| | - Kai Xu
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Pingyuan Laboratory, College of Life Science, Henan Normal University, Xinxiang, China
| | - Lianhui Cheng
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Pingyuan Laboratory, College of Life Science, Henan Normal University, Xinxiang, China
| | - Wenyu Zhao
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Pingyuan Laboratory, College of Life Science, Henan Normal University, Xinxiang, China
| | - Yajun Li
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Pingyuan Laboratory, College of Life Science, Henan Normal University, Xinxiang, China
| | - Lan Wang
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Pingyuan Laboratory, College of Life Science, Henan Normal University, Xinxiang, China
| | - Guoying Yu
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Pingyuan Laboratory, College of Life Science, Henan Normal University, Xinxiang, China
| |
Collapse
|
25
|
Abstract
Evolutionary perspectives have yielded profound insights in health and medical sciences. A fundamental recognition is that modern diet and lifestyle practices are mismatched with the human physiological constitution, shaped over eons in response to environmental selective pressures. This Darwinian angle can help illuminate and resolve issues in nutrition, including the contentious issue of fat consumption. In the present paper, the intake of saturated fat in ancestral and contemporary dietary settings is discussed. It is shown that while saturated fatty acids have been consumed by human ancestors across time and space, they do not feature dominantly in the diets of hunter-gatherers or projected nutritional inputs of genetic accommodation. A higher intake of high-fat dairy and meat products produces a divergent fatty acid profile that can increase the risk of cardiovascular and inflammatory disease and decrease the overall satiating-, antioxidant-, and nutrient capacity of the diet. By prioritizing fiber-rich and micronutrient-dense foods, as well as items with a higher proportion of unsaturated fatty acids, and in particular the long-chain polyunsaturated omega-3 fatty acids, a nutritional profile that is better aligned with that of wild and natural diets is achieved. This would help prevent the burdening diseases of civilization, including heart disease, cancer, and neurodegenerative conditions. Saturated fat is a natural part of a balanced diet; however, caution is warranted in a food environment that differs markedly from the one to which we are adapted.
Collapse
Affiliation(s)
- Eirik Garnås
- Institute of Health, Oslo New University College, Ullevålsveien 76, Oslo, 0454, Norway.
| |
Collapse
|
26
|
Barbhuiya PA, Ahmed A, Dutta PP, Sen S, Pathak MP. Mitigating Metabolic Dysfunction-associated Steatotic Liver Disease (MASLD): The Role of Bioactive Phytoconstituents in Indian Culinary Spices. Curr Nutr Rep 2025; 14:20. [PMID: 39841356 DOI: 10.1007/s13668-024-00598-w] [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] [Accepted: 11/11/2024] [Indexed: 01/30/2025]
Abstract
PURPOSE OF REVIEW The term metabolic dysfunction-associated steatotic liver disease (MASLD) refers to a group of progressive steatotic liver conditions that include metabolic dysfunction-associated steatohepatitis (MASH), which has varying degrees of liver fibrosis and may advance to cirrhosis, and independent hepatic steatosis. MASLD has a complex underlying mechanism, with patients exhibiting diverse causes and phases of the disease. India has a pool prevalence of MASLD of 38.6% in adults. In 2023, the term NAFLD has been redefined and changed to MASLD. Currently, there are no drugs approved by the FDA for the treatment of MASLD. This study investigates the potential of bioactive phytoconstituents present in spices as a therapeutic approach for MASLD. Moreover, it offers comprehensive data on several pre-clinical studies of bioactive phytoconstituents derived from spices that primarily focus on treating obesity-associated MASLD. RECENT FINDINGS Spices include a high amount of bioactive chemicals and several research have indicated their diverse pharmacological activities. Bioactive phytoconstituents from common Indian spices like cinnamic acid, eugenol, curcumin, allicin, 6-gingerols, capsaicin, piperine, eucalyptol, trigonelline, and linalool have been reported to exhibit anti-MASLD effects both in-vivo and in-vitro. Bioactive phytoconstituents from different culinary species of India have shown promising potential against MASLD in pre-clinical status. Further clinical studies on a large scale would be beneficial for paving the path to the development of a new drug which is the need of time.
Collapse
Affiliation(s)
- Pervej Alom Barbhuiya
- Faculty of Pharmaceutical Science, Assam down town University, Sankar Madhab Path, Gandhi Nagar, Panikhaiti, Guwahati, Assam, India, PIN - 781026
- Centre for Research On Ethnomedicine, Faculty of Pharmaceutical Science, Assam Down Town University, Sankar Madhab Path, Gandhi Nagar, Panikhaiti, Guwahati, Assam, India, PIN - 781026
| | - Ameena Ahmed
- Faculty of Pharmaceutical Science, Assam down town University, Sankar Madhab Path, Gandhi Nagar, Panikhaiti, Guwahati, Assam, India, PIN - 781026
- Rahman Institute of Pharmaceutical Sciences and Research, Tepesia, Sonapur, Assam, India, PIN - 782402
| | - Partha Pratim Dutta
- Faculty of Pharmaceutical Science, Assam down town University, Sankar Madhab Path, Gandhi Nagar, Panikhaiti, Guwahati, Assam, India, PIN - 781026
- Centre for Research On Ethnomedicine, Faculty of Pharmaceutical Science, Assam Down Town University, Sankar Madhab Path, Gandhi Nagar, Panikhaiti, Guwahati, Assam, India, PIN - 781026
| | - Saikat Sen
- Faculty of Pharmaceutical Science, Assam down town University, Sankar Madhab Path, Gandhi Nagar, Panikhaiti, Guwahati, Assam, India, PIN - 781026
- Centre for Research On Ethnomedicine, Faculty of Pharmaceutical Science, Assam Down Town University, Sankar Madhab Path, Gandhi Nagar, Panikhaiti, Guwahati, Assam, India, PIN - 781026
| | - Manash Pratim Pathak
- Faculty of Pharmaceutical Science, Assam down town University, Sankar Madhab Path, Gandhi Nagar, Panikhaiti, Guwahati, Assam, India, PIN - 781026.
- Centre for Research On Ethnomedicine, Faculty of Pharmaceutical Science, Assam Down Town University, Sankar Madhab Path, Gandhi Nagar, Panikhaiti, Guwahati, Assam, India, PIN - 781026.
| |
Collapse
|
27
|
Kalecký K, Buitrago L, Alarcon JM, Singh A, Bottiglieri T, Kaddurah-Daouk R, Hernández AI, Alzheimer’s Disease Metabolomics Consortium. Rescue of hippocampal synaptic plasticity and memory performance by Fingolimod (FTY720) in APP/PS1 model of Alzheimer's disease is accompanied by correction in metabolism of sphingolipids, polyamines, and phospholipid saturation composition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.17.633452. [PMID: 39868189 PMCID: PMC11761635 DOI: 10.1101/2025.01.17.633452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Previously, our metabolomic, transcriptomic, and genomic studies characterized the ceramide/sphingomyelin pathway as a therapeutic target in Alzheimer's disease, and we demonstrated that FTY720, a sphingosine-1-phospahate receptor modulator approved for treatment of multiple sclerosis, recovers synaptic plasticity and memory in APP/PS1 mice. To further investigate how FTY720 rescues the pathology, we performed metabolomic analysis in brain, plasma, and liver of trained APP/PS1 and wild-type mice. APP/PS1 mice showed area-specific brain disturbances in polyamines, phospholipids, and sphingolipids. Most changes were completely or partially normalized in FTY720-treated subjects, indicating rebalancing the "sphingolipid rheostat", reactivating phosphatidylethanolamine synthesis via mitochondrial phosphatidylserine decarboxylase pathway, and normalizing polyamine levels that support mitochondrial activity. Synaptic plasticity and memory were rescued, with spermidine synthesis in temporal cortex best corresponding to hippocampal CA3-CA1 plasticity normalization. FTY720 effects, also reflected in other pathways, are consistent with promotion of mitochondrial function, synaptic plasticity, and anti-inflammatory environment, while reducing pro-apoptotic and pro-inflammatory signals.
Collapse
Affiliation(s)
- Karel Kalecký
- Center of Metabolomics, Institute of Metabolic Disease, Baylor Scott & White Research Institute, Dallas, TX, USA
| | - Luna Buitrago
- Neural and Behavioral Sciences Program, School of Graduate Studies, Department of Neurology/Pharmacology, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Juan Marcos Alarcon
- Neural and Behavioral Sciences Program, School of Graduate Studies, The Robert F. Furchgott Center for Neural and Behavioral Science, Department of Pathology, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Abanish Singh
- Department of Psychiatry and Behavioral Sciences; and Department of Medicine, Duke University School of Medicine, Durham, Durham, NC, USA
| | - Teodoro Bottiglieri
- Center of Metabolomics, Institute of Metabolic Disease, Baylor Scott & White Research Institute, Dallas, TX, USA
| | - Rima Kaddurah-Daouk
- Department of Psychiatry and Behavioural Sciences, Duke University, Durham, NC, USA
- Duke Institute of Brain Sciences, Duke University, Durham, NC, USA
- Department of Medicine, Duke University, Durham, NC, USA
| | - Alejandro Iván Hernández
- Neural and Behavioral Sciences Program, School of Graduate Studies, The Robert F. Furchgott Center for Neural and Behavioral Science, Department of Pathology, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | | |
Collapse
|
28
|
Sakamoto K, Butera MA, Zhou C, Maurizi G, Chen B, Ling L, Shawkat A, Patlolla L, Thakker K, Calle V, Morgan DA, Rahmouni K, Schwartz GJ, Tahiri A, Buettner C. Overnutrition causes insulin resistance and metabolic disorder through increased sympathetic nervous system activity. Cell Metab 2025; 37:121-137.e6. [PMID: 39437790 PMCID: PMC11711004 DOI: 10.1016/j.cmet.2024.09.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 06/19/2024] [Accepted: 09/23/2024] [Indexed: 10/25/2024]
Abstract
The mechanisms underlying obesity-induced insulin resistance remain incompletely understood, as impaired cellular insulin signaling, traditionally considered the primary driver of insulin resistance, does not always accompany impaired insulin action. Overnutrition rapidly increases plasma norepinephrine (NE), suggesting overactivation of the sympathetic nervous system (SNS). However, the role of the SNS in obesity is controversial, as both increased and decreased SNS activity (SNA) have been reported. Here, we show that reducing catecholamine (CA) release from the SNS protects against overnutrition-induced insulin resistance as well as hyperglucagonemia, adipose tissue dysfunction, and fatty liver disease, as we demonstrate utilizing a mouse model of inducible and peripherally restricted deletion of tyrosine hydroxylase (th; THΔper). A key mechanism through which heightened SNA induces insulin resistance is by triggering adipose tissue lipolysis. Increased SNA emerges as a critical driver in the pathogenesis of overnutrition-induced insulin resistance and metabolic disease independent of cellular insulin signaling.
Collapse
Affiliation(s)
- Kenichi Sakamoto
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA; Department of Medicine and Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mary A Butera
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA; Department of Medicine and Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chunxue Zhou
- Department of Medicine and Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Giulia Maurizi
- Department of Medicine and Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bandy Chen
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA; Department of Medicine and Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Li Ling
- Department of Medicine and Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Adham Shawkat
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Likhitha Patlolla
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Kavira Thakker
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Victor Calle
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Donald A Morgan
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Kamal Rahmouni
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Gary J Schwartz
- Department of Medicine & Neuroscience, Albert Einstein College of Medicine, New York, NY, USA
| | - Azeddine Tahiri
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Christoph Buettner
- Division of Endocrinology, Metabolism & Nutrition, Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA; Department of Medicine and Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
29
|
An H, Jang Y, Choi J, Hur J, Kim S, Kwon Y. New Insights into AMPK, as a Potential Therapeutic Target in Metabolic Dysfunction-Associated Steatotic Liver Disease and Hepatic Fibrosis. Biomol Ther (Seoul) 2025; 33:18-38. [PMID: 39702310 PMCID: PMC11704404 DOI: 10.4062/biomolther.2024.188] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 12/08/2024] [Accepted: 12/10/2024] [Indexed: 12/21/2024] Open
Abstract
AMP-activated protein kinase (AMPK) activators have garnered significant attention for their potential to prevent the progression of metabolic dysfunction-associated steatotic liver disease (MASLD) into liver fibrosis and to fundamentally improve liver function. The broad spectrum of pathways regulated by AMPK activators makes them promising alternatives to conventional liver replacement therapies and the limited pharmacological treatments currently available. In this study, we aim to illustrate the newly detailed multiple mechanisms of MASLD progression based on the multiple-hit hypothesis. This model posits that impaired lipid metabolism, combined with insulin resistance and metabolic imbalance, initiates inflammatory cascades, gut dysbiosis, and the accumulation of toxic metabolites, ultimately promoting fibrosis and accelerating MASLD progression to irreversible hepatocellular carcinoma (HCC). AMPK plays a multifaceted protective role against these pathological conditions by regulating several key downstream signaling pathways. It regulates biological effectors critical to metabolic and inflammatory responses, such as SIRT1, Nrf2, mTOR, and TGF-β, through complex and interrelated mechanisms. Due to these intricate connections, AMPK's role is pivotal in managing metabolic and inflammatory disorders. In this review, we demonstrate the specific roles of AMPK and its related pathways. Several agents directly activate AMPK by binding as agonists, while some others indirectly activate AMPK by modulating upstream molecules, including adiponectin, LKB1, and the AMP: ATP ratio. As AMPK activators can target each stage of MASLD progression, the development of AMPK activators offers immense potential to expand therapeutic strategies for liver diseases such as MASH, MASLD, and liver fibrosis.
Collapse
Affiliation(s)
- Haeun An
- College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Yerin Jang
- College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Jungin Choi
- College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Juhee Hur
- College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Seojeong Kim
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Youngjoo Kwon
- College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| |
Collapse
|
30
|
Feng S, Wang J, Peng Q, Zhang P, Jiang Y, Zhang H, Song X, Li Y, Huang W, Zhang D, Deng C. Schisandra sphenanthera extract modulates sweet taste receptor pathway, IRS/PI3K, AMPK/mTOR pathway and endogenous metabolites against T2DM. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156348. [PMID: 39740377 DOI: 10.1016/j.phymed.2024.156348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 11/20/2024] [Accepted: 12/24/2024] [Indexed: 01/02/2025]
Abstract
BACKGROUND Southern Schisandra is the dried and matured fruit of Schisandra sphenanthera Rehd. et Wils. in the family of Magnoliaceae; Traditional medicine reports that Schisandra sphenanthera has astringent and astringent properties, benefiting qi and promoting the production of body fluid, tranquilising the heart and calming the mind; it is clinically utilized for prolonged cough, thirst due to injury of the body fluid, internal heat and thirst, palpitation and insomnia, etc., and thirst belongs to the category of diabetes mellitus; the literature reports and the preliminary study of our team showed that Schisandra sphenanthera can be used to prevent and control diabetes mellitus. PURPOSE In the research, we investigated the mechanism of action of SDP against T2DM by integrating pharmacodynamics, endogenous metabolite assays and signalling pathways. MATERIALS AND METHODS UPLC-MS/MS was used to identify the chemical constituents. HPLC was utilized to determine the content of eight lignan-like components in SDP. A T2DM rat model was established by the combined induction of high-fat and high-sugar feed and STZ, and the mechanism of action of SDP on T2DM was investigated by using biochemical indices, Western blot analysis of protein expression, mRNA expression, immunohistochemistry and endogenous metabolites. RESULTS The chemical components in SDP were determined by UPLC-MS/MS and HPLC, and biochemical indicators determined that SDP has the effects of lowering blood glucose, anti-glycolipid metabolism, and anti-oxidative stress, and is able to restore pathological damage in the liver and pancreas, activate the PI3K/AKT, AMPK/mTOR, and sweetness receptor signalling pathways, restore the sweetness receptor mRNAs, and modulate the urinary compounds such as malic acid, γ-aminobutyric acid, leucine, N-acetylaspartic acid and other compounds thereby achieving the therapeutic effect of T2DM. CONCLUSION SDP can ameliorate diabetes-induced symptoms related to elevated blood glucose, dyslipidaemia, elevated fasting insulin levels and impaired glucose tolerance in rats; the anti-T2DM of SDP may be through the regulation of the sweet taste receptor pathway, the PI3K/AKT/mTOR and the AMPK/mTOR signalling pathway, which leads to the development of a normal level and exerts an antidiabetic effect.
Collapse
Affiliation(s)
- Shibo Feng
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China
| | - Jiaojiao Wang
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China
| | - Qin Peng
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China
| | - Panpan Zhang
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China
| | - Yi Jiang
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China; College of Pharmacy and Shaanxi Qinling Application Development and Engineering Center of Chinese Herbal Medicine, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China; Shaanxi Key Laboratory of Research and Application of"Taibai Qi Yao", Xianyang 712046, PR China
| | - Huawei Zhang
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China; College of Pharmacy and Shaanxi Qinling Application Development and Engineering Center of Chinese Herbal Medicine, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China; Shaanxi Key Laboratory of Research and Application of"Taibai Qi Yao", Xianyang 712046, PR China
| | - Xiaomei Song
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China; College of Pharmacy and Shaanxi Qinling Application Development and Engineering Center of Chinese Herbal Medicine, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China; Shaanxi Key Laboratory of Research and Application of"Taibai Qi Yao", Xianyang 712046, PR China
| | - Yuze Li
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China; College of Pharmacy and Shaanxi Qinling Application Development and Engineering Center of Chinese Herbal Medicine, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China; Shaanxi Key Laboratory of Research and Application of"Taibai Qi Yao", Xianyang 712046, PR China
| | - Wenli Huang
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China; College of Pharmacy and Shaanxi Qinling Application Development and Engineering Center of Chinese Herbal Medicine, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China; Shaanxi Key Laboratory of Research and Application of"Taibai Qi Yao", Xianyang 712046, PR China
| | - Dongdong Zhang
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China; College of Pharmacy and Shaanxi Qinling Application Development and Engineering Center of Chinese Herbal Medicine, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China; Shaanxi Key Laboratory of Research and Application of"Taibai Qi Yao", Xianyang 712046, PR China
| | - Chong Deng
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China; College of Pharmacy and Shaanxi Qinling Application Development and Engineering Center of Chinese Herbal Medicine, Shaanxi University of Chinese Medicine, Xianyang 712046, PR China; Key Laboratory of Pharmacodynamics and Material Basis of Chinese Medicine, Shaanxi Administration of Traditional Chinese Medicine, Xi'an 712046, PR China; Shaanxi Key Laboratory of Research and Application of"Taibai Qi Yao", Xianyang 712046, PR China.
| |
Collapse
|
31
|
Khan TJ, Semenkovich CF, Zayed MA. De novo lipid synthesis in cardiovascular tissue and disease. Atherosclerosis 2025; 400:119066. [PMID: 39616863 DOI: 10.1016/j.atherosclerosis.2024.119066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 11/14/2024] [Accepted: 11/21/2024] [Indexed: 12/13/2024]
Abstract
Most tissues have the capacity for endogenous lipid synthesis. A crucial foundational pathway for lipid synthesis is de novo lipid synthesis (DNL), a ubiquitous and complex metabolic process that occurs at high levels in the liver, adipose and brain tissue. Under normal physiological conditions, DNL is vital in converting excess carbohydrates into fatty acids. DNL is linked to other pathways, including the endogenous synthesis of phospholipids and sphingolipids. However, abnormal lipid synthesis can contribute to various pathologies and clinical conditions. Experimental studies involving dietary restriction and in vivo genetic modifications provide compelling evidence demonstrating the significance of lipid synthesis in maintaining normal cardiovascular tissue function. Similarly, clinical investigations suggest altered lipid synthesis can harm cardiac and arterial tissues, thereby influencing cardiovascular disease (CVD) development and progression. Consequently, there is increased interest in exploring pharmacological interventions that target lipid synthesis metabolic pathways as potential strategies to alleviate CVD. Here we review the physiological and pathological impact of endogenous lipid synthesis and its implications for CVD. Since lipid synthesis can be targeted pharmacologically, enhancing our understanding of the molecular and biochemical mechanisms underlying lipid generation and cardiovascular function may prompt new insights into CVD and its treatment.
Collapse
Affiliation(s)
- Tariq J Khan
- Washington University School of Medicine, Department of Surgery, Section of Vascular Surgery, St. Louis, MO, USA
| | - Clay F Semenkovich
- Washington University School of Medicine, Department of Medicine, Division of Endocrinology, Metabolism and Lipid Research, St. Louis, MO, USA; Washington University School of Medicine, Department of Cell Biology and Physiology, St. Louis, MO, USA
| | - Mohamed A Zayed
- Washington University School of Medicine, Department of Surgery, Section of Vascular Surgery, St. Louis, MO, USA; Washington University School of Medicine, Department of Surgery, Division of Surgical Sciences, St. Louis, MO, USA; Washington University School of Medicine, Department of Radiology, St. Louis, MO, USA; Washington University School of Medicine, Division of Molecular Cell Biology, St. Louis, MO, USA; Washington University, McKelvey School of Engineering, Department of Biomedical Engineering, St. Louis, MO, USA; Veterans Affairs St. Louis Health Care System, St. Louis, MO, USA.
| |
Collapse
|
32
|
Zhang H, Zhang L, Zhao X, Ma Y, Sun D, Bai Y, Liu W, Liang X, Liang H. Folic Acid Prevents High-Fat Diet-Induced Postpartum Weight Retention in Rats, Which Is Associated with a Reduction in Endoplasmic Reticulum Stress-Mediated Hepatic Lipogenesis. Nutrients 2024; 16:4377. [PMID: 39770997 PMCID: PMC11676124 DOI: 10.3390/nu16244377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 12/13/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Proactively preventing postpartum weight retention (PPWR) is one of the effective intervention strategies to reduce the occurrence of obesity in women. Population studies have shown that serum folate levels are closely related to body weight. The regulation of folic acid on lipid metabolism has been fully confirmed in both in vivo and in vitro studies. For many years, folic acid supplementation has been widely used in periconceptional women due to its role in preventing fetal neural tube defects. However, whether folic acid supplementation prior to and throughout pregnancy exerts preventive effects on PPWR remains uncertain. This study aims to investigate the preventive effect of folic acid on PPWR in rats and further explore the underlying mechanisms. METHODS In this study, pregnant rats were administered one of the dietary schedules: control diet (CON), high-fat diet (HF), control diet combined with folic acid (FA) and high-fat diet combined with folic acid (HF + FA). RESULTS We discovered that folic acid supplementation inhibited high-fat diet-induced elevations in body weight, visceral fat weight, liver weight, hepatic lipid levels and serum lipid levels at 1 week post-weaning (PW). Western blot analysis showed that folic acid supplementation inhibited the expression of endoplasmic reticulum (ER) stress-specific proteins including GRP78, PERK, eIF2α, IRE1α, XBP1 and ATF6, subsequently decreasing the expression of proteins related to lipid synthesis including SREBP-1c, ACC1 and FAS. CONCLUSIONS In conclusion, folic acid supplementation prior to and throughout pregnancy exerts preventive effects on high-fat diet-induced PPWR in rats, and the mechanism is associated with the inhibition of ER stress-mediated lipogenesis signaling pathways in the liver. Folic acid supplementation may serve as a potential strategy for preventing PPWR. In the future, the effectiveness of folic acid in PPWR prevention can be further verified by population studies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Hui Liang
- Department of Nutrition and Food Hygiene, School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao 266071, China; (H.Z.); (L.Z.); (X.Z.); (Y.M.); (D.S.); (Y.B.); (W.L.); (X.L.)
| |
Collapse
|
33
|
Kuhnert LRB, Pontes RDFC, Neto JGO, Romão JS, Pinto CEDC, Oliveira KJ. Cinnamon powder intake enhances the effect of caloric restriction on white adipose tissue in male rats. J Mol Histol 2024; 56:19. [PMID: 39627596 DOI: 10.1007/s10735-024-10288-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 10/30/2024] [Indexed: 02/07/2025]
Abstract
Caloric Restriction (CR) and cinnamon promote several benefits, including the modulation of lipid metabolism and body fat mass. We hypothesize that cinnamon may act as a mimetic of restriction or enhance the effects of caloric restriction on adipose tissue. Adult male Wistar rats were divided into Control (CT, n = 8) and Cinnamon (CIN, n = 7), with free access to standard chow; Calorie Restriction (CR, n = 8) and Calorie Restriction with Cinnamon (CIN-CR, n = 7), subjected to a 30% reduction in food intake compared to the average consumption of CT rats. Both CIN groups received 50 mg cinnamon powder (Cinnamomun verum) per kg body mass, by gavage, over 6 weeks. Cinnamon treatment did not alter food intake under either ad libitum or caloric restriction conditions. The CR and CIN-CR groups exhibit lower body mass. Basal glycemia, lipid profile, and triglyceride-glycemic index were similar between groups. The combination of both interventions induced lower visceral white adipose tissue (WAT) mass, and smaller adipocyte diameter in the visceral and subcutaneous WAT compartments, accompanied by reduced expression of genes related to lipid metabolism (Acaca, Fasn, Cd36, Srebf1c), suggesting decreased lipid synthesis. Histological analyses identified a browning phenotype in the CR, CIN, and CIN-CR groups, positive for UCP1 immunostaining. The CR and CIN-CR groups showed lower Atg7 expression, and CIN-CR animals expressed increased levels of Lamp2, suggesting modulation of autophagy. Brown adipose tissue mass and lipid content were not influenced by any intervention. These findings suggest that cinnamon may enhance the effects of caloric restriction in promoting adipocyte metabolic health.
Collapse
Affiliation(s)
- Lia Rafaella Ballard Kuhnert
- Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University, Niteroi, RJ, Brazil
- Laboratory of Experimental Pathology, Department of Immunobiology, Institute of Biology, Federal Fluminense University, Niteroi, RJ, Brazil
| | | | - Jessika Geisebel Oliveira Neto
- Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University, Niteroi, RJ, Brazil
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Juliana Santos Romão
- Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University, Niteroi, RJ, Brazil
| | - Carla Eponina de Carvalho Pinto
- Laboratory of Experimental Pathology, Department of Immunobiology, Institute of Biology, Federal Fluminense University, Niteroi, RJ, Brazil
| | - Karen Jesus Oliveira
- Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University, Niteroi, RJ, Brazil.
| |
Collapse
|
34
|
Yadav P, Quadri K, Kadian R, Waziri A, Agrawal P, Alam MS. New approaches to the treatment of metabolic dysfunction-associated steatotic liver with natural products. ILIVER 2024; 3:100131. [DOI: 10.1016/j.iliver.2024.100131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
35
|
Zumaraga MP, Desmarchelier C, Gleize B, Nowicki M, Ould-Ali D, Landrier JF, Borel P. Identification of Genetic Polymorphisms Associated with Interindividual Variability of Vitamin A Concentration in Adipose Tissue of Healthy Male Adults. J Nutr 2024; 154:3693-3703. [PMID: 39442757 DOI: 10.1016/j.tjnut.2024.10.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 09/18/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Adipose tissue vitamin A (VA), that is, mainly retinol (RET) and its esters, comes from preformed VA and proVA carotenoids present in our food. Adipose tissue VA acts as hormonal cue maintaining essential aspects of adipocyte biology, which includes fat mobilization and catabolism, energy balance, and glucose homeostasis, and it is thus of particular interest to study its determinants, including genetic ones. OBJECTIVES This study aimed to identify genetic variations associated with adipose tissue VA concentration. METHODS Forty-two healthy male adults received, in a randomized crossover design, 3 test meals. Periumbilical adipose tissue samples were collected on 6 occasions, that is, at fast and 8 h after consumption of each meal. RET concentration was measured in both plasma and the adipose tissue following saponification. Participants were genotyped using whole-genome microarrays. A total of 1305 single nucleotide polymorphism (SNPs) in or near 27 candidate genes were included for univariate analysis. Partial least squares (PLS) regression was carried out to find the best combination of SNPs associated with the interindividual variability in adipose tissue RET concentration. RESULTS Adipose tissue RET concentration was not associated with plasma RET concentrations (r = -0.184, P = 0.28). Interindividual variability of adipose tissue RET concentration was high (coefficient of variation = 62%). Twenty-nine SNPs were significantly (P < 0.05) associated with adipose tissue RET concentration and a PLS regression model identified 16 SNPs as explanatory variables of this concentration. The SNPs were in or near peroxisome proliferator activated receptor gamma, retinoid X receptor alpha, signaling receptor and transporter of retinol, cluster of differentiation 36, free fatty acid receptor 4, aldehyde dehydrogenase 1 family member A1, monoglyceride lipase, diacylglycerol O-acyltransferase 2, and polycystic kidney disease 1-like 2. CONCLUSIONS A combination of 16 SNPs has been associated with the interindividual of adipose tissue VA concentration in humans. This trial was registered at clinicaltrials.gov as NCT02100774.
Collapse
Affiliation(s)
- Mark Pretzel Zumaraga
- C2VN, Aix Marseille Univ, INRAE, INSERM, Marseille, France; Department of Science and Technology-Food and Nutrition Research Institute, Bicutan, Taguig City, Philippines
| | - Charles Desmarchelier
- C2VN, Aix Marseille Univ, INRAE, INSERM, Marseille, France; Direction générale de la recherche et de l'innovation, Paris, France
| | | | - Marion Nowicki
- C2VN, Aix Marseille Univ, INRAE, INSERM, Marseille, France
| | - Djaffar Ould-Ali
- Plastic & Anesthetic Surgery Department, Clinique Internationale du Parc Monceau, Paris, France
| | | | - Patrick Borel
- C2VN, Aix Marseille Univ, INRAE, INSERM, Marseille, France.
| |
Collapse
|
36
|
Yang Y, Shu X, Javed HU, Wu Q, Liu H, Han J, Zhou H. Dietary supplementation of poly-dihydromyricetin-fused zinc nanoparticles alleviates fatty liver hemorrhagic syndrome by improving antioxidant capacity, intestinal health and lipid metabolism of laying hens. Poult Sci 2024; 103:104301. [PMID: 39306955 PMCID: PMC11447411 DOI: 10.1016/j.psj.2024.104301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 08/28/2024] [Accepted: 09/03/2024] [Indexed: 10/06/2024] Open
Abstract
Fatty liver hemorrhagic syndrome is the main cause of noninfectious death of laying hens and results in substantial economic losses to the poultry industry. This study focused on evaluating the effects of Poly-dihydromyricetin-fused zinc nanoparticles (PDMY-Zn NPs) on antioxidant capacity, liver lipid metabolism, and intestinal health in laying hens. A total of 288 Jingfen laying hens (52 wk old) with similar body weights were randomly divided into 4 dietary groups with 6 replicates in each group for 8 wk. The control group received a basal diet, while the treatment groups were supplemented with PDMY-Zn NPs at levels of 200, 400, and 600 mg/kg, respectively. The results indicate that PDMY-Zn NPs supplementation can enhance antioxidant parameters (P < 0.05) in the blood and liver of laying hens. Simultaneously, it can mitigate vacuolar degeneration and inflammatory necrosis in hepatocytes, improve the relative expression level of related parameters associated with liver lipid metabolism and key regulatory genes (P < 0.05). Furthermore, it has been observed to reshape the composition and diversity of cecum microbes by increasing beneficial probiotics such as Lactobacillus and Prevotella, while also enhancing villi height and villi/crypt ratio in the duodenum and ileum (P < 0.05). Additionally, it elevates liver bile acid content along with the relative expression of key genes involved in liver synthesis (P < 0.05). In summary, PDMY-Zn NPs showed potential to alleviate fatty liver hemorrhagic syndrome by enhancing antioxidant capacity, regulating liver lipid metabolism, and maintaining intestinal health.
Collapse
Affiliation(s)
- Yuanting Yang
- Zhanjiang Experimental Station, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang 524013, China
| | - Xugang Shu
- College of Chemistry and Chemical Engineering, Zhongkai University of Agricultural Engineering, Guangzhou 510225, China
| | - Hafiz Umer Javed
- Guangxi College and University Key Laboratory of High-Value Utilization of Seafood and Prepared Food in Beibu Gulf, College of Food Engineering, Beibu Gulf University, Qinzhou 535011, China
| | - Qun Wu
- Zhanjiang Experimental Station, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang 524013, China
| | - Hu Liu
- Zhanjiang Experimental Station, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang 524013, China
| | - Jiancheng Han
- Zhanjiang Experimental Station, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang 524013, China
| | - Hanlin Zhou
- Zhanjiang Experimental Station, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang 524013, China.
| |
Collapse
|
37
|
Shi Y, Xu J, Yi S, Xu C, Yu F, Gu W, Zhang J, Ye L. Effects of high dietary carbohydrate intake in patients with obstructive sleep apnea. Sleep Breath 2024; 29:20. [PMID: 39607637 PMCID: PMC11604710 DOI: 10.1007/s11325-024-03188-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 08/16/2024] [Accepted: 10/14/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND This study aimed to characterize the relationship between habitual food intake and OSA. METHODS The diet of 200 volunteers was evaluated using a Food Frequency Questionnaire. The patients were further allocated to mild, moderate, and severe OSA groups. The relationships of OSA with diet were evaluated using multivariate linear regression analysis and the relationships of the energy supply ratio for three major nutrients and demand with sleep parameters were assessed using correlation analysis. RESULTS The analysis of nutrient intake showed a reduced energy intake in non-OSA patients (11953.98 ± 7578.11 kJ, 95% CI 9677.27-14230.70 kJ) as compared to severe-OSA patients (15153.43 ± 6541.89 kJ, 95% CI 13678.46-16628.40 kJ) (p < 0.05). Similarly, lower intake of carbohydrates was observed in non-OSA (249.94 ± 134.96 g/day, 95% CI 209.39-290.48 g/day) as compared to severe OSA patients (348.57 ± 112.34 g/day, 95% CI 323.24-373.90 g/day) (p < 0.05). Compared with participants without OSA, we found that those with OSA had significantly poorer diets, with higher contents of rice (β = 0.195, p = 0.006), wheat (β = 0.236, p = 0.001), stuffed food items (β = 0.278, p < 0.001), fried food (β = 0.193, p = 0.006), dairy products (β = 0.198, p = 0.005), and sweet beverages (β = 0.154, p = 0.030). After adjustment for age, sex, the presence of diabetes, and waist-hip ratio, these relationships remained. Furthermore, a higher proportion of energy intake in the form of carbohydrate was associated with more severe OSA. CONCLUSION The severity of OSA is related to the level of carbohydrate consumption. Poor dietary habits predispose toward body mass gain and a worsening of sleep-related parameters, which may accelerate the pathogenesis of OSA.
Collapse
Affiliation(s)
- Ying Shi
- Department of Respiration, Nanjing First Hospital, Nanjing Medical University, Changle Road 68, Nanjing, 210006, China
| | - Jun Xu
- Department of Respiration, Nanjing First Hospital, Nanjing Medical University, Changle Road 68, Nanjing, 210006, China
| | - Shenwen Yi
- Department of Respiration, Nanjing First Hospital, Nanjing Medical University, Changle Road 68, Nanjing, 210006, China
| | - Chenyu Xu
- Department of Respiration, Nanjing First Hospital, Nanjing Medical University, Changle Road 68, Nanjing, 210006, China
| | - Fei Yu
- Department of Respiration, Nanjing First Hospital, Nanjing Medical University, Changle Road 68, Nanjing, 210006, China
| | - Wei Gu
- Department of Respiration, Nanjing First Hospital, Nanjing Medical University, Changle Road 68, Nanjing, 210006, China
| | - Jing Zhang
- Department of Respiration, Nanjing First Hospital, Nanjing Medical University, Changle Road 68, Nanjing, 210006, China.
| | - Liang Ye
- Department of Respiration, Nanjing First Hospital, Nanjing Medical University, Changle Road 68, Nanjing, 210006, China.
| |
Collapse
|
38
|
Zhu JY, Guo L. Exercise-regulated lipolysis: Its role and mechanism in health and diseases. J Adv Res 2024:S2090-1232(24)00550-2. [PMID: 39613256 DOI: 10.1016/j.jare.2024.11.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/24/2024] [Accepted: 11/26/2024] [Indexed: 12/01/2024] Open
Abstract
Exercise has received considerable attention because of its importance not just in regulating physiological function, but also in ameliorating multiple pathological processes. Among these processes, lipolysis may play an important role in exercise-induced benefits. It is generally accepted that active lipolysis contributes to breakdown of fats, leading to the release of free fatty acids (FFAs) that serve as an energy source for muscles and other tissues during exercise. However, the significance of lipolysis in the context of exercise has not been fully understood. This review comprehensively outlines the potential regulatory mechanisms by which exercise stimulates lipolysis. The potential roles of exercise-mediated lipolysis in various physiological and pathological processes are also summarized. Additionally, we also discussed the potential non-classical effects of key lipolytic effectors induced by exercise. This will enhance our understanding of how exercise improves lipolytic function to bring about beneficial effects, offering new insights into potential therapeutic avenues for promoting health and alleviating diseases.
Collapse
Affiliation(s)
- Jie-Ying Zhu
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China 200438; Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China 200438; Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai, China 200438
| | - Liang Guo
- School of Exercise and Health and Collaborative Innovation Center for Sports and Public Health, Shanghai University of Sport, Shanghai, China 200438; Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China 200438; Key Laboratory of Exercise and Health Sciences of the Ministry of Education, Shanghai University of Sport, Shanghai, China 200438.
| |
Collapse
|
39
|
Lim W, Choi S, Kim J, Baek KS, Park M, Lee G, Lim TG. Vine Tea Extract (VTE) Inhibits High-Fat Diet-Induced Adiposity: Evidence of VTE's Anti-Obesity Effects In Vitro and In Vivo. Int J Mol Sci 2024; 25:12042. [PMID: 39596109 PMCID: PMC11593453 DOI: 10.3390/ijms252212042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 11/01/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
This study focused on evaluating the anti-obesity effects of an extract from Ampelopsis grossedentata (Hand.-Mazz.) W. T. Wang, also known as vine tea, in mature adipocytes and high-fat diet-induced obese mice. Vine tea extract (VTE) effectively decreased lipid accumulation in mature adipocytes without cytotoxicity, as confirmed by the regulation of several factors associated with adipogenesis, lipogenesis, or lipolysis. Subsequently, in a 12-week experiment with obese mice, oral VTE administration significantly reduced body weight gain induced with high-fat diet intake. Au-topsy findings showed reduced fat accumulation in various areas without liver damage. The VTE-administered group showed lower serum LDL levels, while increasing HDL, than the high-fat diet-administered group. Analysis of adipose tissue biomarkers indicated VTE's ability to inhibit adipogenesis and lipogenesis, promote lipolysis, and regulate energy metabolism, contributing to reduced adiposity induced by the consumption of a high-fat diet.
Collapse
Affiliation(s)
- Wonchul Lim
- Department of Food Science & Biotechnology, Carbohydrate Bioproduct Research Center, Sejong University, Seoul 05006, Republic of Korea;
| | - Seongmin Choi
- Department of Food Science & Biotechnology, Sejong University, Seoul 05006, Republic of Korea;
| | - Jinhak Kim
- R&D Division, Daehan Chemtech Co., Ltd., Gwacheon-si 13840, Gyeonggi-do, Republic of Korea; (J.K.); (K.-S.B.)
| | - Kwang-Soo Baek
- R&D Division, Daehan Chemtech Co., Ltd., Gwacheon-si 13840, Gyeonggi-do, Republic of Korea; (J.K.); (K.-S.B.)
| | - Minkuk Park
- Department of Integrative Biological Sciences and Industry, Sejong University, Seoul 05006, Republic of Korea; (M.P.); (G.L.)
| | - Gakyung Lee
- Department of Integrative Biological Sciences and Industry, Sejong University, Seoul 05006, Republic of Korea; (M.P.); (G.L.)
- Convergence Research Center for Natural Products, Sejong University, Seoul 05006, Republic of Korea
| | - Tae-Gyu Lim
- Department of Food Science & Biotechnology, Carbohydrate Bioproduct Research Center, Sejong University, Seoul 05006, Republic of Korea;
- Department of Food Science & Biotechnology, Sejong University, Seoul 05006, Republic of Korea;
| |
Collapse
|
40
|
Markowska J, Kasprzak-Drozd K, Niziński P, Dragan M, Kondracka A, Gondek E, Oniszczuk T, Oniszczuk A. Quercetin: A Promising Candidate for the Management of Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD). Molecules 2024; 29:5245. [PMID: 39598636 PMCID: PMC11596905 DOI: 10.3390/molecules29225245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/30/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) represents a chronic liver disease. The development of MASLD is influenced by a multitude of diseases associated with modern lifestyles, including but not limited to diabetes mellitus, hypertension, hyperlipidaemia and obesity. These conditions are often consequences of the adoption of unhealthy habits, namely a sedentary lifestyle, a lack of physical activity, poor dietary choices and excessive alcohol consumption. The treatment of MASLD is primarily based on modifying the patient's lifestyle and pharmacological intervention. Despite the absence of FDA-approved pharmacological agents for the treatment of MASLD, several potential therapeutic modalities have demonstrated efficacy in reversing the histopathological features of the disease. Among the botanical ingredients belonging to the flavonoid group is quercetin (QE). QE has been demonstrated to possess a number of beneficial physiological effects, including anti-inflammatory, anticancer and antifungal properties. Additionally, it functions as a natural antioxidant. Preclinical evidence indicates that QE may play a beneficial role in reducing liver damage and improving metabolic health. Early human studies also suggest that QE may be an effective treatment for MASLD due to its antioxidant, anti-inflammatory, and lipid-regulating properties. This review aims to summarize the available information on the therapeutic effects of QE in MASLD.
Collapse
Affiliation(s)
- Julia Markowska
- Science Circle of the Department of Inorganic Chemistry, Medical University of Lublin, Dr. Witolda Chodźki 4a, 20-093 Lublin, Poland; (J.M.); (M.D.)
| | - Kamila Kasprzak-Drozd
- Department of Inorganic Chemistry, Medical University of Lublin, Dr. Witolda Chodźki 4a, 20-093 Lublin, Poland;
| | - Przemysław Niziński
- Department of Pharmacology, Medical University of Lublin, Radziwiłłowska 11, 20-080 Lublin, Poland;
| | - Magdalena Dragan
- Science Circle of the Department of Inorganic Chemistry, Medical University of Lublin, Dr. Witolda Chodźki 4a, 20-093 Lublin, Poland; (J.M.); (M.D.)
| | - Adrianna Kondracka
- Department of Obstetrics and Pathology of Pregnancy, Medical University of Lublin, 20-081 Lublin, Poland;
| | - Ewa Gondek
- Department of Food Engineering and Process Management, Institute of Food Science, Warsaw University of Life Sciences, Nowoursynowska 159C, 02-776 Warsaw, Poland
| | - Tomasz Oniszczuk
- Department of Thermal Technology and Food Process Engineering, University of Life Sciences in Lublin, Głęboka 31, 20-612 Lublin, Poland;
| | - Anna Oniszczuk
- Department of Inorganic Chemistry, Medical University of Lublin, Dr. Witolda Chodźki 4a, 20-093 Lublin, Poland;
| |
Collapse
|
41
|
Tang B, Kang W, Dong Q, Qin Z, Duan L, Zhao X, Yuan G, Pan Y. Research progress on S-palmitoylation modification mediated by the ZDHHC family in glioblastoma. Front Cell Dev Biol 2024; 12:1413708. [PMID: 39563863 PMCID: PMC11573772 DOI: 10.3389/fcell.2024.1413708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 10/18/2024] [Indexed: 11/21/2024] Open
Abstract
S-Palmitoylation has been widely noticed and studied in a variety of diseases. Increasing evidence suggests that S-palmitoylation modification also plays a key role in Glioblastoma (GBM). The zDHHC family, as an important member of S-palmitoyltransferases, has received extensive attention for its function and mechanism in GBM which is one of the most common primary malignant tumors of the brain and has an adverse prognosis. This review focuses on the zDHHC family, essential S-palmitoyltransferases, and their involvement in GBM. By summarizing recent studies on zDHHC molecules in GBM, we highlight their significance in regulating critical processes such as cell proliferation, invasion, and apoptosis. Specifically, members of zDHHC3, zDHHC4, zDHHC5 and others affect key processes such as signal transduction and phenotypic transformation in GBM cells through different pathways, which in turn influence tumorigenesis and progression. This review systematically outlines the mechanism of zDHHC family-mediated S-palmitoylation modification in GBM, emphasizes its importance in the development of this disease, and provides potential targets and strategies for the treatment of GBM. It also offers theoretical foundations and insights for future research and clinical applications.
Collapse
Affiliation(s)
- Beiyan Tang
- The Second Medical College of Lanzhou University, Lanzhou, Gansu, China
| | - Wei Kang
- Department of Neurosurgery, Second Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Qiang Dong
- Department of Neurosurgery, Second Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Zhenwei Qin
- Department of Neurosurgery, Second Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Lei Duan
- Department of Neurosurgery, Second Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Xianjun Zhao
- Department of Neurosurgery, Second Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Guoqiang Yuan
- Key Laboratory of Neurology of Gansu Province, Lanzhou University, Lanzhou, Gansu, China
- Academician Workstation, The Second Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Yawen Pan
- Department of Neurosurgery, Second Hospital of Lanzhou University, Lanzhou, Gansu, China
- Key Laboratory of Neurology of Gansu Province, Lanzhou University, Lanzhou, Gansu, China
- Academician Workstation, The Second Hospital of Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
42
|
Smith ME, Bazinet RP. Unraveling brain palmitic acid: Origin, levels and metabolic fate. Prog Lipid Res 2024; 96:101300. [PMID: 39222711 DOI: 10.1016/j.plipres.2024.101300] [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: 03/26/2024] [Revised: 08/26/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
In the human brain, palmitic acid (16:0; PAM) comprises nearly half of total brain saturates and has been identified as the third most abundant fatty acid overall. Brain PAM supports the structure of membrane phospholipids, provides energy, and regulates protein stability. Sources underlying the origin of brain PAM are both diet and endogenous synthesis via de novo lipogenesis (DNL), primarily from glucose. However, studies investigating the origin of brain PAM are limited to tracer studies utilizing labelled (14C/11C/3H/2H) PAM, and results vary based on the model and tracer used. Nevertheless, there is evidence PAM is synthesized locally in the brain, in addition to obtained directly from the diet. Herein, we provide an overview of brain PAM origin, entry to the brain, metabolic fate, and factors influencing brain PAM kinetics and levels, the latter in the context of age, as well as neurological diseases and psychiatric disorders. Additionally, we briefly summarize the role of PAM in signaling at the level of the brain. We add to the literature a rudimentary summary on brain PAM metabolism.
Collapse
Affiliation(s)
- Mackenzie E Smith
- Department of Nutritional Sciences, University of Toronto, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| | - Richard P Bazinet
- Department of Nutritional Sciences, University of Toronto, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada.
| |
Collapse
|
43
|
Hou N, Zhou H, Li J, Xiong X, Deng H, Xiong S. Macrophage polarization and metabolic reprogramming in abdominal aortic aneurysm. Immun Inflamm Dis 2024; 12:e1268. [PMID: 39530309 PMCID: PMC11555488 DOI: 10.1002/iid3.1268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 04/03/2024] [Accepted: 04/22/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Abdominal aortic aneurysm (AAA) is a macrovascular disease with high morbidity and mortality in the elderly. The limitation of the current management is that most patients can only be followed up until the AAA diameter increases to a threshold, and surgical intervention is recommended. The development of preventive and curative drugs for AAA is urgently needed. Macrophage-mediated immune inflammation is one of the key pathological links in the occurrence and development of AAA. AIMS This review article aims to evaluate the impact of immunometabolism on macrophage biology and its role in AAA. METHODS We analyze publications focusing on the polarization and metabolic reprogramming in macrophages as well as their potential impact on AAA, and summarize the potential interventions that are currently available to regulate these processes. RESULTS The phenotypic and functional changes in macrophages are accompanied by significant alterations in metabolic pathways. The interaction between macrophage polarization and metabolic pathways significantly influences the progression of AAA. CONCLUSION Macrophage polarization is a manifestation of the gross dichotomy of macrophage function into pro-inflammatory killing and tissue repair, that is, classically activated M1 macrophages and alternatively activated M2 macrophages. Macrophage functions are closely linked to metabolic changes, and the emerging field of immunometabolism is providing unique insights into the role of macrophages in AAA. It is essential to further investigate the precise metabolic changes and their functional consequences in AAA-associated macrophages.
Collapse
Affiliation(s)
- Ningxin Hou
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Hongmin Zhou
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Jun Li
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xiaoxing Xiong
- Department of NeurosurgeryRenmin Hospital of Wuhan UniversityWuhanChina
| | - Hongping Deng
- Department of Vascular SurgeryRenmin Hospital of Wuhan UniversityWuhanChina
| | - Sizheng Xiong
- Department of Vascular SurgeryRenmin Hospital of Wuhan UniversityWuhanChina
| |
Collapse
|
44
|
Tang Y, Chen Z, Zuo Q, Kang Y. Regulation of CD8+ T cells by lipid metabolism in cancer progression. Cell Mol Immunol 2024; 21:1215-1230. [PMID: 39402302 PMCID: PMC11527989 DOI: 10.1038/s41423-024-01224-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 09/22/2024] [Indexed: 11/02/2024] Open
Abstract
Dysregulation of lipid metabolism is a key characteristic of the tumor microenvironment, where tumor cells utilize lipids for proliferation, survival, metastasis, and evasion of immune surveillance. Lipid metabolism has become a critical regulator of CD8+ T-cell-mediated antitumor immunity, with excess lipids in the tumor microenvironment impeding CD8+ T-cell activities. Considering the limited efficacy of immunotherapy in many solid tumors, targeting lipid metabolism to enhance CD8+ T-cell effector functions could significantly improve immunotherapy outcomes. In this review, we examine recent findings on how lipid metabolic processes, including lipid uptake, synthesis, and oxidation, regulate CD8+ T cells within tumors. We also assessed the impact of different lipids on CD8+ T-cell-mediated antitumor immunity, with a particular focus on how lipid metabolism affects mitochondrial function in tumor-infiltrating CD8+ T cells. Furthermore, as cancer is a systemic disease, we examined systemic factors linking lipid metabolism to CD8+ T-cell effector function. Finally, we summarize current therapeutic approaches that target lipid metabolism to increase antitumor immunity and enhance immunotherapy. Understanding the molecular and functional interplay between lipid metabolism and CD8+ T cells offers promising therapeutic opportunities for cancer treatment.
Collapse
Affiliation(s)
- Yong Tang
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA
- Ludwig Institute for Cancer Research Princeton Branch, Princeton, NJ, 08544, USA
| | - Ziqing Chen
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA
- Ludwig Institute for Cancer Research Princeton Branch, Princeton, NJ, 08544, USA
| | - Qianying Zuo
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA
- Ludwig Institute for Cancer Research Princeton Branch, Princeton, NJ, 08544, USA
| | - Yibin Kang
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA.
- Ludwig Institute for Cancer Research Princeton Branch, Princeton, NJ, 08544, USA.
- Cancer Metabolism and Growth Program, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, 08903, USA.
| |
Collapse
|
45
|
Fang X, Wang J, Ye C, Lin J, Ran J, Jia Z, Gong J, Zhang Y, Xiang J, Lu X, Xie C, Liu J. Polyphenol-mediated redox-active hydrogel with H 2S gaseous-bioelectric coupling for periodontal bone healing in diabetes. Nat Commun 2024; 15:9071. [PMID: 39433776 PMCID: PMC11494015 DOI: 10.1038/s41467-024-53290-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 10/08/2024] [Indexed: 10/23/2024] Open
Abstract
Excessive oxidative response, unbalanced immunomodulation, and impaired mesenchymal stem cell function in periodontitis in diabetes makes it a great challenge to achieve integrated periodontal tissue regeneration. Here, a polyphenol-mediated redox-active algin/gelatin hydrogel encapsulating a conductive poly(3,4-ethylenedioxythiopene)-assembled polydopamine-mediated silk microfiber network and a hydrogen sulfide sustained-release system utilizing bovine serum albumin nanoparticles is developed. This hydrogel is found to reverse the hyperglycemic inflammatory microenvironment and enhance functional tissue regeneration in diabetic periodontitis. Polydopamine confers the hydrogel with anti-oxidative and anti-inflammatory activity. The slow, sustained release of hydrogen sulfide from the bovine serum albumin nanoparticles recruits mesenchymal stem cells and promotes subsequent angiogenesis and osteogenesis. Moreover, poly(3,4-ethylenedioxythiopene)-assembled polydopamine-mediated silk microfiber confers the hydrogel with good conductivity, which enables it to transmit endogenous bioelectricity, promote cell arrangement, and increase the inflow of calcium ion. In addition, the synergistic effects of hydrogen sulfide gaseous-bioelectric coupling promotes bone formation by amplifying autophagy in periodontal ligament stem cells and modulating macrophage polarization via lipid metabolism regulation. This study provides innovative insights into the synergistic effects of conductivity, reactive oxygen species scavenging, and hydrogen sulfide on the periodontium in a hyperglycemic inflammatory microenvironment, offering a strategy for the design of gaseous-bioelectric biomaterials to promote functional tissue regeneration in immune-related diseases.
Collapse
Affiliation(s)
- Xinyi Fang
- Lab of Aging Research and Department of Geriatrics, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, PR China
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, PR China
- Hospital of Stomatology, Key Laboratory of Oral Biomedical Research of Zhejiang Province, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, 310016, PR China
| | - Jun Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Chengxinyue Ye
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Jiu Lin
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
- Hospital of Stomatology, Key Laboratory of Oral Biomedical Research of Zhejiang Province, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, 310016, PR China
| | - Jinhui Ran
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, PR China
| | - Zhanrong Jia
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, PR China
- The Tenth Affiliated Hospital of Southern Medical University, Dongguan, 523059, PR China
| | - Jinglei Gong
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Yiming Zhang
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, PR China
| | - Jie Xiang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Xiong Lu
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, PR China
| | - Chaoming Xie
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, PR China.
| | - Jin Liu
- Lab of Aging Research and Department of Geriatrics, State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, PR China.
| |
Collapse
|
46
|
Peng CC, Chen ECY, Chen CR, Chyau CC, Chen KC, Peng RY. Molecular mechanism of ectopic lipid accumulation induced by methylglyoxal via activation of the NRF2/PI3K/AKT pathway implicates renal lipotoxicity caused by diabetes mellitus. PLoS One 2024; 19:e0306575. [PMID: 39413106 PMCID: PMC11482718 DOI: 10.1371/journal.pone.0306575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 06/19/2024] [Indexed: 10/18/2024] Open
Abstract
Patients with chronic kidney disease (CKD) have a high incidence of dyslipidemia comprising high triglyceride (TG) and low high-density lipoprotein (HDL)-cholesterol levels. An abnormal increase of TGs within cells can lead to intracellular lipid accumulation. In addition to dyslipidemia, hyperglycemia in diabetes may elicit ectopic lipid deposition in non-adipose tissues. Hyperglycemia increases intracellular levels of methylglyoxal (MG) leading to cellular dysfunction. A deficit of glyoxalase I (GLO1) contributes to dicarbonyl stress. Whether dicarbonyl stress induced by MG causes renal lipotoxicity through alteration of lipid metabolism signaling is still unknown. In this study, mice with high fat diet-induced diabetes were used to investigate the renal pathology induced by MG. NRK52E cells treated with MG were further used in vitro to delineate the involvement of lipogenic signaling. After treatment with MG for 12 weeks, plasma TG levels, renal fatty changes, and tubular injuries were aggravated in diabetic mice. In NRK52E cells, MG activated the nuclear factor erythroid 2-related factor 2 (Nrf2)/phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) and sterol regulatory element-binding protein 1 (SREBP1), resulting in stimulation of fatty acid synthase. The intracellular accumulation of lipid droplets was mainly contributed by TGs, which increased the oxidative stress accompanied by high Nrf2 expression. In addition, MG time-dependently activated cyclin D, cyclin-dependent kinase 4 (CDK4), and cleaved caspase-3, evidencing that G0/G1 arrest was associated with apoptosis of NRK52E cells. In conclusion, our studies revealed the mechanism of lipotoxicity caused by MG. The target of such dicarbonyl stress may become a promising therapy for diabetic CKD.
Collapse
Affiliation(s)
- Chiung Chi Peng
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Eugene Chang Yu Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- MD Program, Medical College of Wisconsin, Milwaukee, Wisconsin, United States of America
| | - Chang-Rong Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Charng-Cherng Chyau
- Department of Biotechnology, College of Medical and Health Care, Hungkuang University, Shalu District, Taichung, Taiwan
| | - Kuan-Chou Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Urology, Taipei Medical University Shuang-Ho Hospital, Zhong-He District, New Taipei City, Taiwan
- TMU-Research Center of Urology and Kidney, Taipei Medical University, Taipei, Taiwan
| | - Robert Y. Peng
- Department of Biotechnology, College of Medical and Health Care, Hungkuang University, Shalu District, Taichung, Taiwan
| |
Collapse
|
47
|
Tauil RB, Golono PT, de Lima EP, de Alvares Goulart R, Guiguer EL, Bechara MD, Nicolau CCT, Yanaguizawa Junior JL, Fiorini AMR, Méndez-Sánchez N, Abenavoli L, Direito R, Valente VE, Laurindo LF, Barbalho SM. Metabolic-Associated Fatty Liver Disease: The Influence of Oxidative Stress, Inflammation, Mitochondrial Dysfunctions, and the Role of Polyphenols. Pharmaceuticals (Basel) 2024; 17:1354. [PMID: 39458995 PMCID: PMC11510109 DOI: 10.3390/ph17101354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/05/2024] [Accepted: 10/07/2024] [Indexed: 10/28/2024] Open
Abstract
Metabolic-Associated Fatty Liver Disease (MAFLD) is a clinical-pathological scenario that occurs due to the accumulation of triglycerides in hepatocytes which is considered a significant cause of liver conditions and contributes to an increased risk of death worldwide. Even though the possible causes of MAFLD can involve the interaction of genetics, hormones, and nutrition, lifestyle (diet and sedentary lifestyle) is the most influential factor in developing this condition. Polyphenols comprise many natural chemical compounds that can be helpful in managing metabolic diseases. Therefore, the aim of this review was to investigate the impact of oxidative stress, inflammation, mitochondrial dysfunction, and the role of polyphenols in managing MAFLD. Some polyphenols can reverse part of the liver damage related to inflammation, oxidative stress, or mitochondrial dysfunction, and among them are anthocyanin, baicalin, catechin, curcumin, chlorogenic acid, didymin, epigallocatechin-3-gallate, luteolin, mangiferin, puerarin, punicalagin, resveratrol, and silymarin. These compounds have actions in reducing plasma liver enzymes, body mass index, waist circumference, adipose visceral indices, lipids, glycated hemoglobin, insulin resistance, and the HOMA index. They also reduce nuclear factor-KB (NF-KB), interleukin (IL)-1β, IL-6, tumor necrosis factor-α (TNF-α), blood pressure, liver fat content, steatosis index, and fibrosis. On the other hand, they can improve HDL-c, adiponectin levels, and fibrogenesis markers. These results show that polyphenols are promising in the prevention and treatment of MAFLD.
Collapse
Affiliation(s)
- Raissa Bulaty Tauil
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
| | - Paula Takano Golono
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
| | - Enzo Pereira de Lima
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
| | - Ricardo de Alvares Goulart
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
| | - Elen Landgraf Guiguer
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
- Department of Biochemistry and Nutrition, School of Food and Technology of Marília (FATEC), Marília 17500-000, São Paulo, Brazil
| | - Marcelo Dib Bechara
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
| | - Claudia C. T. Nicolau
- Department of Biochemistry and Nutrition, School of Food and Technology of Marília (FATEC), Marília 17500-000, São Paulo, Brazil
| | - José Luiz Yanaguizawa Junior
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
| | - Adriana M. R. Fiorini
- Department of Biochemistry and Nutrition, School of Food and Technology of Marília (FATEC), Marília 17500-000, São Paulo, Brazil
| | - Nahum Méndez-Sánchez
- Liver Research Unit, Medica Sur Clinic & Foundation, Mexico City 14050, Mexico;
- Faculty of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico
| | - Ludovico Abenavoli
- Department of Health Sciences, University “Magna Graecia”, Viale Europa, 88100 Catanzaro, Italy;
| | - Rosa Direito
- Laboratory of Systems Integration Pharmacology, Clinical and Regulatory Science, Research Institute for Medicines, Universidade de Lisboa (iMed.ULisboa), Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal;
| | - Vitor Engrácia Valente
- Autonomic Nervous System Center, School of Philosophy and Sciences, São Paulo State University, Marília 17525-902, São Paulo, Brazil
| | - Lucas Fornari Laurindo
- Department of Biochemistry and Pharmacology, School of Medicine, Faculdade de Medicina de Marília (FAMEMA), Marília 17519-030, São Paulo, Brazil;
| | - Sandra Maria Barbalho
- Department of Biochemistry and Pharmacology, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, School of Medicine, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
- Department of Biochemistry and Nutrition, School of Food and Technology of Marília (FATEC), Marília 17500-000, São Paulo, Brazil
- Research Coordination, UNIMAR Charity Hospital, Universidade de Marília (UNIMAR), Marília 17525-902, São Paulo, Brazil
| |
Collapse
|
48
|
Chamas L, Seugnet I, Tanvé O, Enderlin V, Clerget-Froidevaux MS. The Downregulation of the Liver Lipid Metabolism Induced by Hypothyroidism in Male Mice: Metabolic Flexibility Favors Compensatory Mechanisms in White Adipose Tissue. Int J Mol Sci 2024; 25:10792. [PMID: 39409121 PMCID: PMC11477049 DOI: 10.3390/ijms251910792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 09/03/2024] [Accepted: 09/10/2024] [Indexed: 10/20/2024] Open
Abstract
In mammals, the maintenance of energy homeostasis relies on complex mechanisms requiring tight synchronization between peripheral organs and the brain. Thyroid hormones (THs), through their pleiotropic actions, play a central role in these regulations. Hypothyroidism, which is characterized by low circulating TH levels, slows down the metabolism, which leads to a reduction in energy expenditure as well as in lipid and glucose metabolism. The objective of this study was to evaluate whether the metabolic deregulations induced by hypothyroidism could be avoided through regulatory mechanisms involved in metabolic flexibility. To this end, the response to induced hypothyroidism was compared in males from two mouse strains, the wild-derived WSB/EiJ mouse strain characterized by a diet-induced obesity (DIO) resistance due to its high metabolic flexibility phenotype and C57BL/6J mice, which are prone to DIO. The results show that propylthiouracil (PTU)-induced hypothyroidism led to metabolic deregulations, particularly a reduction in hepatic lipid synthesis in both strains. Furthermore, in contrast to the C57BL/6J mice, the WSB/EiJ mice were resistant to the metabolic dysregulations induced by hypothyroidism, mainly through enhanced lipid metabolism in their adipose tissue. Indeed, WSB/EiJ mice compensated for the decrease in hepatic lipid synthesis by mobilizing lipid reserves from white adipose tissue. Gene expression analysis revealed that hypothyroidism stimulated the hypothalamic orexigenic circuit in both strains, but there was unchanged melanocortin 4 receptor (Mc4r) and leptin receptor (LepR) expression in the hypothyroid WSB/EiJ mice strain, which reflects their adaptability to maintain their body weight, in contrast to C57BL/6J mice. Thus, this study showed that WSB/EiJ male mice displayed a resistance to the metabolic dysregulations induced by hypothyroidism through compensatory mechanisms. This highlights the importance of metabolic flexibility in the ability to adapt to disturbed circulating TH levels.
Collapse
Affiliation(s)
- Lamis Chamas
- CNRS/MNHN UMR 7221 “Physiologie Moléculaire et Adaptation” Phyma, Department of “Life Adaptations” Muséum National d’Histoire Naturelle 57, Rue Cuvier CP 32, 75231 Paris, CEDEX 05, France
| | - Isabelle Seugnet
- CNRS/MNHN UMR 7221 “Physiologie Moléculaire et Adaptation” Phyma, Department of “Life Adaptations” Muséum National d’Histoire Naturelle 57, Rue Cuvier CP 32, 75231 Paris, CEDEX 05, France
| | - Odessa Tanvé
- CNRS/MNHN UMR 7221 “Physiologie Moléculaire et Adaptation” Phyma, Department of “Life Adaptations” Muséum National d’Histoire Naturelle 57, Rue Cuvier CP 32, 75231 Paris, CEDEX 05, France
| | - Valérie Enderlin
- Paris-Saclay Institute of Neuroscience (Neuro-PSI), CNRS UMR 9197, Université Paris-Saclay, 91400 Saclay, France;
| | - Marie-Stéphanie Clerget-Froidevaux
- CNRS/MNHN UMR 7221 “Physiologie Moléculaire et Adaptation” Phyma, Department of “Life Adaptations” Muséum National d’Histoire Naturelle 57, Rue Cuvier CP 32, 75231 Paris, CEDEX 05, France
| |
Collapse
|
49
|
Milani I, Codini M, Guarisco G, Chinucci M, Gaita C, Leonetti F, Capoccia D. Hepatokines and MASLD: The GLP1-Ras-FGF21-Fetuin-A Crosstalk as a Therapeutic Target. Int J Mol Sci 2024; 25:10795. [PMID: 39409124 PMCID: PMC11477334 DOI: 10.3390/ijms251910795] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/04/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024] Open
Abstract
The introduction of the term "Metabolic Steatotic Liver Disease" (MASLD) underscores the critical role of metabolic dysfunction in the development and progression of chronic liver disease and emphasizes the need for strategies that address both liver disease and its metabolic comorbidities. In recent years, a liver-focused perspective has revealed that altered endocrine function of the fatty liver is a key contributor to the metabolic dysregulation observed in MASLD. Due to its secretory capacity, the liver's increased production of proteins known as "hepatokines" has been linked to the development of insulin resistance, explaining why MASLD often precedes dysfunction in other organs and ultimately contributes to systemic metabolic disease. Among these hepatokines, fibroblast growth factor 21 (FGF21) and fetuin-A play central roles in regulating the metabolic abnormalities associated with MASLD, explaining why their dysregulated secretion in response to metabolic stress has been implicated in the metabolic abnormalities of MASLD. This review postulates why their modulation by GLP1-Ras may mediate the beneficial metabolic effects of these drugs, which have increased attention to their emerging role as pharmacotherapy for MASLD. By discussing the crosstalk between GLP1-Ras-FGF21-fetuin-A, this review hypothesizes that the possible modulation of fetuin-A by the novel GLP1-FGF21 dual agonist pharmacotherapy may contribute to the management of metabolic and liver diseases. Although research is needed to go into the details of this crosstalk, this topic may help researchers explore the mechanisms by which this type of pharmacotherapy may manage the metabolic dysfunction of MASLD.
Collapse
Affiliation(s)
- Ilaria Milani
- Department of Medico-Surgical Sciences and Biotechnologies, Faculty of Pharmacy and Medicine, University of Rome La Sapienza, 04100 Latina, Italy; (I.M.); (G.G.); (M.C.); (C.G.); (F.L.)
| | - Michela Codini
- Department of Pharmaceutical Sciences, University of Perugia, Via Fabretti 48, 06123 Perugia, Italy;
| | - Gloria Guarisco
- Department of Medico-Surgical Sciences and Biotechnologies, Faculty of Pharmacy and Medicine, University of Rome La Sapienza, 04100 Latina, Italy; (I.M.); (G.G.); (M.C.); (C.G.); (F.L.)
| | - Marianna Chinucci
- Department of Medico-Surgical Sciences and Biotechnologies, Faculty of Pharmacy and Medicine, University of Rome La Sapienza, 04100 Latina, Italy; (I.M.); (G.G.); (M.C.); (C.G.); (F.L.)
| | - Chiara Gaita
- Department of Medico-Surgical Sciences and Biotechnologies, Faculty of Pharmacy and Medicine, University of Rome La Sapienza, 04100 Latina, Italy; (I.M.); (G.G.); (M.C.); (C.G.); (F.L.)
| | - Frida Leonetti
- Department of Medico-Surgical Sciences and Biotechnologies, Faculty of Pharmacy and Medicine, University of Rome La Sapienza, 04100 Latina, Italy; (I.M.); (G.G.); (M.C.); (C.G.); (F.L.)
| | - Danila Capoccia
- Department of Medico-Surgical Sciences and Biotechnologies, Faculty of Pharmacy and Medicine, University of Rome La Sapienza, 04100 Latina, Italy; (I.M.); (G.G.); (M.C.); (C.G.); (F.L.)
| |
Collapse
|
50
|
Matthan NR, Lovato L, Petersen KS, Kris-Etherton PM, Sabate J, Rajaram S, Li Z, Reboussin DM, Lichtenstein AH. Effect of daily avocado consumption for 6 mo compared with habitual diet on red blood cell fatty acid profiles and association with cardiometabolic risk factors in individuals with abdominal obesity: a randomized trial. Am J Clin Nutr 2024; 120:794-803. [PMID: 39128497 DOI: 10.1016/j.ajcnut.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 08/02/2024] [Accepted: 08/06/2024] [Indexed: 08/13/2024] Open
Abstract
BACKGROUND Avocado intake improves dietary fat quality, but the subsequent impact on red blood cell (RBC) saturated (SFA), monounsaturated (MUFA), polyunsaturated (PUFA), and trans-fatty acid (TFA) composition and association with cardiometabolic health, has not been elucidated. OBJECTIVES To compare the effect of consuming 1 avocado/d relative to habitual diet (HAB) on RBC-FA profiles, and their association with visceral adiposity and cardiometabolic risk factors (CMRFs) in individuals with abdominal obesity. METHODS RBC-FA profiling at baseline, 3- and 6 mo was conducted in participants (n = 994) from the Habitual Diet and Avocado Trial (HAT). HAT was a multisite, free-living, parallel-arm intervention study in which participants were randomly assigned to either the avocado-supplemented group (AVO, usual diet with 1 avocado/d) or the HAB group (usual diet with limited avocado intake) for 6 mo. Changes in RBC-FA profiles, a secondary outcome measure, were determined within and between groups using linear regression and mixed effect models, adjusting for age, sex, BMI, clinical site, smoking status, and percentage of energy intake from fat at baseline. The association between changes in RBC-FAs with visceral adiposity measures and CMRFs was assessed after covariate and False Discovery Rate (FDR <0.05) adjustment. RESULTS No major differences in RBC-FA profiles were observed between groups, with the exception of MUFA cis-vaccenic [18:1n-7c], which was significantly higher in AVO (β: 0.11 [0.05, 0.17]) compared with the HAB (β: 0.03 [-0.03, 0.08]) participants. In the HAB but not AVO group, increases in MUFA cis (18:1n-7c, oleic [18;1n-9c], erucic [22:1n-9c]) and MUFA trans (palmitelaidic [16:1n-7t], vaccenic [18:1n-7t], elaidic [18:1n-9t], and petroselaidic [18;1n-10-12t), as well as PUFA γ-linolenic [18:3n-6], dihomo-γ-linolenic [20:3n-6], arachidonic [20:4n-6], and α-linolenic [18:3n-3] were associated with unfavorable changes in visceral adiposity measures, lipid profiles, glucose, insulin and high sensitivity C-reactive protein concentrations. CONCLUSIONS Daily avocado intake over 6-mo modified RBC-MUFA composition, notably 18:1n-7c, and potentially mitigated some of the unfavorable individual RBC-FA-CMRF associations observed over time in the HAB group. This trial was registered at https://clinicaltrials.gov/study as NCT03528031.
Collapse
Affiliation(s)
- Nirupa R Matthan
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, United States.
| | - Laura Lovato
- Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Kristina S Petersen
- Department of Nutritional Sciences, Pennsylvania State University, University Park, PA, United States
| | - Penny M Kris-Etherton
- Department of Nutritional Sciences, Pennsylvania State University, University Park, PA, United States
| | - Joan Sabate
- Center for Nutrition, Healthy Lifestyle and Disease Prevention, Loma Linda University School of Public Health, Loma Linda, CA, United States
| | - Sujatha Rajaram
- Center for Nutrition, Healthy Lifestyle and Disease Prevention, Loma Linda University School of Public Health, Loma Linda, CA, United States
| | - Zhaoping Li
- Center for Human Nutrition, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - David M Reboussin
- Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Alice H Lichtenstein
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, United States
| |
Collapse
|