1
|
Zhen J, Liu S, Liu L, Zheng X, Zhao G, Liang J, Xu A, Li C, Ren L, Wu J, Cheung BMY. Circulating fibroblast growth factor 21 is associated with blood pressure in the Chinese population: a community-based study. Ann Med 2025; 57:2500689. [PMID: 40356318 PMCID: PMC12077425 DOI: 10.1080/07853890.2025.2500689] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 11/19/2024] [Accepted: 12/27/2024] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Our research team previously found that fibroblast growth factor (FGF) 21, a circulating hormone, was significantly associated with atherosclerosis in human and animal models. The relationship between FGF21 and blood pressure (BP) is rarely studied in the Asian population. Therefore, we aimed to explore the relationship of FGF21 with BP in a Chinese population. METHODS We analysed data on 1051 participants from the Shenzhen-Hong Kong United Network on Cardiovascular Disease (SHUN-CVD) study. RESULTS The medians of FGF21 level were 355.1 pg/mL (IQR 234.3-574.8 pg/mL) for hypertensive patients and 253.5 (IQR 136.9-403.3 pg/mL) for non-hypertensive participants. Ln-transformed FGF21 level was associated with both systolic and diastolic BP (systolic BP: B = 4.45 [95% CI 3.41-5.49]; p < .001; diastolic BP: B = 2.72 [95% CI 2.03-3.42]; p < .001). After adjusting for sex, age, body mass index, hypercholesterolaemia, diabetes, alcohol consumption, smoking and physical activity, the association remained significant (systolic BP: B = 1.99 [95% CI 1.01-2.97]; p < .001; diastolic BP: B = 1.36 [95% CI 0.69-2.04]; p < .001). Serum FGF21 level was associated with hypertension (quartile 4 vs. quartile 1, OR = 4.19 [95% CI 2.65-6.61]; p for trend < .001). CONCLUSIONS This is the first study to elucidate the relationship of FGF21 with BP in the Asian population. FGF21 is significantly associated with BP. Besides its use as a biomarker, FGF21 may be a new drug target for hypertension treatment.
Collapse
Affiliation(s)
- Juanying Zhen
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong SAR, China
- Department of Neurology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Shuyun Liu
- Department of Neurology, Shenzhen Longhua District Central Hospital, Shenzhen, China
| | - Lin Liu
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong SAR, China
| | - Xiaodan Zheng
- Department of Neurology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Guoru Zhao
- CAS Key Laboratory of Human-Machine Intelligence-Synergy Systems, Research Center for Neural Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Jianguo Liang
- ’Precision Health Research Center Company Limited, Hong Kong SAR, China
| | - Aimin Xu
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong SAR, China
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Chao Li
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong SAR, China
- Department of Neurology, Peking University Shenzhen Hospital, Shenzhen, China
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- Department of Neurology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Lijie Ren
- Department of Neurology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Jun Wu
- Department of Neurology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Bernard Man Yung Cheung
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong SAR, China
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- Department of Neurology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
- Institute of Cardiovascular Science and Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| |
Collapse
|
2
|
Črešnovar T, Habe B, Mohorko N, Kenig S, Jenko Pražnikar Z, Petelin A. Early time-restricted eating with energy restriction has a better effect on body fat mass, diastolic blood pressure, metabolic age and fasting glucose compared to late time-restricted eating with energy restriction and/or energy restriction alone: A 3-month randomized clinical trial. Clin Nutr 2025; 49:57-68. [PMID: 40250088 DOI: 10.1016/j.clnu.2025.04.001] [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: 10/09/2024] [Revised: 03/06/2025] [Accepted: 04/01/2025] [Indexed: 04/20/2025]
Abstract
BACKGROUND & AIMS Time-restricted eating (TRE) has attracted increasing attention from researchers and the public. Recent studies suggest that the combination of TRE with energy restriction (ER) may have more favourable effects on both physical and biochemical aspects compared to ER alone. The aim of the present 3-month intervention study was to determine the effects of an 8-h early time-restricted eating and an 8-h late time-restricted eating with ER (eTRE + ER and lTRE + ER) compared to 12-h ER alone on body mass and other anthropometric and cardiometabolic risk factors in participants with overweight and obesity. METHODS Participants (n = 108) were allocated to three different groups according to their personal chronotype: eTRE + ER (37 participants), lTRE + ER (37 participants) and ER (34 participants). Ninety-three participants completed the entire 3-month intervention (34 in eTRE + ER, 28 in lTRE + ER and 31 in ER). Anthropometric and cardiometabolic risk factors were measured at baseline and after 1, 2 and 3 months of the intervention. Sleep quality and quality of life were assessed at baseline and after 3 months of the intervention. ER was determined based on the individual's resting metabolic rate. Effects were analyzed using the per-protocol approach. RESULTS Results showed a significant time main effect (p < 0.001), suggesting a decrease in body mass at the end of the 3-month intervention with a mean loss of -5.0 kg (95 % CI, -5.7, -4.3) for the eTRE + ER group, -4.4 kg (95 % CI, -5.2, -3.6) for the lTRE + ER group and -4.3 kg (95 % CI, -5.0, -3.6) for the ER group, with no significant difference between the groups (p = 0.319). eTRE + ER had greater improvements in fat mass (-1.2 % (95 % CI, -2.1, -0.2), p = 0.013) and fasting glucose (-0.35 mmol/L (95 % CI, -0.63, -0.06), p = 0.012) than participants in the lTRE + ER group and greater improvements in fat mass (-1.1 % (95 % CI, -2.0, -0.1), p = 0.022), metabolic age (-3 years (95 % CI, -5, -0), p = 0.028) and diastolic blood pressure (-4 mmHg (95 % CI, -8, -0), p = 0.033) than the participants in the ER group. No significant changes were found between the groups for the other parameters measured. CONCLUSIONS There was no difference in body mass between the eTRE + ER, lTRE + ER and ER groups after 3 months of intervention. However, eTRE + ER showed a greater benefit for fasting blood glucose, certain anthropometric parameters and diastolic blood pressure compared to lTRE + ER and/or ER alone. Other anthropometric, biochemical and health-related parameters were not affected by eating window. TRIAL REGISTRATION https://clinicaltrials.gov/study/NCT05730231.
Collapse
Affiliation(s)
- Tanja Črešnovar
- University of Primorska Faculty of Health Sciences, Polje 42, 6310 Izola, Slovenia.
| | - Bernarda Habe
- University of Primorska Faculty of Health Sciences, Polje 42, 6310 Izola, Slovenia.
| | - Nina Mohorko
- University of Primorska Faculty of Health Sciences, Polje 42, 6310 Izola, Slovenia.
| | - Saša Kenig
- University of Primorska Faculty of Health Sciences, Polje 42, 6310 Izola, Slovenia.
| | - Zala Jenko Pražnikar
- University of Primorska Faculty of Health Sciences, Polje 42, 6310 Izola, Slovenia.
| | - Ana Petelin
- University of Primorska Faculty of Health Sciences, Polje 42, 6310 Izola, Slovenia.
| |
Collapse
|
3
|
Varghese A, Gusarov I, Gamallo-Lana B, Dolgonos D, Mankan Y, Shamovsky I, Phan M, Jones R, Gomez-Jenkins M, White E, Wang R, Jones DR, Papagiannakopoulos T, Pacold ME, Mar AC, Littman DR, Nudler E. Unravelling cysteine-deficiency-associated rapid weight loss. Nature 2025:10.1038/s41586-025-08996-y. [PMID: 40399674 DOI: 10.1038/s41586-025-08996-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 04/08/2025] [Indexed: 05/23/2025]
Abstract
Around 40% of the US population and 1 in 6 individuals worldwide have obesity, with the incidence surging globally1,2. Various dietary interventions, including carbohydrate, fat and, more recently, amino acid restriction, have been explored to combat this epidemic3-6. Here we investigated the impact of removing individual amino acids on the weight profiles of mice. We show that conditional cysteine restriction resulted in the most substantial weight loss when compared to essential amino acid restriction, amounting to 30% within 1 week, which was readily reversed. We found that cysteine deficiency activated the integrated stress response and oxidative stress response, which amplify each other, leading to the induction of GDF15 and FGF21, partly explaining the phenotype7-9. Notably, we observed lower levels of tissue coenzyme A (CoA), which has been considered to be extremely stable10, resulting in reduced mitochondrial functionality and metabolic rewiring. This results in energetically inefficient anaerobic glycolysis and defective tricarboxylic acid cycle, with sustained urinary excretion of pyruvate, orotate, citrate, α-ketoglutarate, nitrogen-rich compounds and amino acids. In summary, our investigation reveals that cysteine restriction, by depleting GSH and CoA, exerts a maximal impact on weight loss, metabolism and stress signalling compared with other amino acid restrictions. These findings suggest strategies for addressing a range of metabolic diseases and the growing obesity crisis.
Collapse
Affiliation(s)
- Alan Varghese
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY, USA
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY, USA
| | - Ivan Gusarov
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY, USA
| | - Begoña Gamallo-Lana
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, USA
| | - Daria Dolgonos
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY, USA
| | - Yatin Mankan
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY, USA
| | - Ilya Shamovsky
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY, USA
| | - Mydia Phan
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY, USA
| | - Rebecca Jones
- Division of Advanced Research Technologies, NYU Grossman School of Medicine, New York, NY, USA
| | - Maria Gomez-Jenkins
- Rutgers Cancer Institute, Rutgers University, New Brunswick, NJ, USA
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ, USA
| | - Eileen White
- Rutgers Cancer Institute, Rutgers University, New Brunswick, NJ, USA
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ, USA
- Ludwig Princeton Branch, Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, USA
| | - Rui Wang
- Department of Biology, York University, Toronto, Ontario, Canada
| | - Drew R Jones
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY, USA
| | - Thales Papagiannakopoulos
- Department of Pathology, Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Michael E Pacold
- Department of Radiation Oncology and Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Adam C Mar
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, USA
| | - Dan R Littman
- Department of Cell Biology, NYU Grossman School of Medicine, New York, NY, USA.
- Howard Hughes Medical Institute, New York, NY, USA.
| | - Evgeny Nudler
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY, USA.
- Howard Hughes Medical Institute, New York, NY, USA.
| |
Collapse
|
4
|
Xie X, Song Y, Chen W, Zhao H, Chu N, Wang F. Association between circulating inflammatory proteins and gout: A Mendelian randomization study. Medicine (Baltimore) 2025; 104:e42379. [PMID: 40388724 PMCID: PMC12091660 DOI: 10.1097/md.0000000000042379] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 04/18/2025] [Indexed: 05/21/2025] Open
Abstract
Clinical studies have consistently demonstrated that inflammation is a critical factor in the pathophysiology and progression of gout. This study aims to explore the causal relationship between CIPs and gout, utilizing MR in conjunction with meta-analyses. We utilized genetic data pertaining to gout from the GWAS which involved 3576 cases and 147,221 control participants. A total of 132 CIPs were extracted from the GWAS data to identify SNPs associated with gout. The primary analytical approach was the IVW method. Sensitivity analyses indicated no pleiotropy or heterogeneity. The IVW results revealed that several CIPs were associated with gout in European populations. The analysis results indicate that FGF-21, MMP-1, G-CSF, and IFN-γ are involved in the pathogenesis of gout, and gout may influence the expression of CXCL1, IL-1Ra, and TNF-α. Consequently, targeted research focusing on specific CIPs could provide a promising strategy for the treatment and prevention of gout, offering potential therapeutic targets for the underlying inflammatory mechanisms of the disease.
Collapse
Affiliation(s)
- Xiaochao Xie
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
- Department of Endocrinology, Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Qingdao, People’s Republic of China
| | - Yanjie Song
- Department of Cardiology, Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Qingdao, People’s Republic of China
| | - Wenwen Chen
- Department of Endocrinology, Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Qingdao, People’s Republic of China
| | - Hui Zhao
- Department of Endocrinology, Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Qingdao, People’s Republic of China
| | - Nan Chu
- Department of Endocrinology, Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Qingdao, People’s Republic of China
| | - Fang Wang
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| |
Collapse
|
5
|
Dai M, Jia Z, Wang H, Zheng P, Xue Y, Gong K, Zhao R. Sodium-Glucose Cotransporter 2 Inhibitor Ameliorate Angiotensin II-Induced Hypertension and Vascular Injury by Upregulating FGF21. Inflammation 2025:10.1007/s10753-025-02309-1. [PMID: 40369373 DOI: 10.1007/s10753-025-02309-1] [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: 03/03/2025] [Revised: 04/22/2025] [Accepted: 04/25/2025] [Indexed: 05/16/2025]
Abstract
Clinical trials have demonstrated Sodium-glucose cotransporter 2 inhibitors (SGLT2i) antihypertensive effects, yet their underlying mechanisms remain to be fully elucidated. Fibroblast growth factor 21 (FGF21) circulating levels are associated with hypertension in humans. This study aims to investigate the roles of SGLT2i and FGF21 in improving hypertension and their potential mechanisms. A mouse model of Ang II-induced hypertension was established. Wild-type (WT) C57BL/6 mice and FGF21 knockout (FGF21-/-) mice were sequentially treated with Angiotensin II (Ang II) and dapagliflozin. Blood pressure was monitored. Cardiac structure was assessed using echocardiography. Serum FGF21 levels were measured, and the expression of fibroblast growth factor receptor 1 (FGFR1) in the thoracic aorta was quantified. Vascular pathology and oxidative stress responses were evaluated. Human aortic smooth muscle cells (HASMCs) were treated with Ang II or SGLT2i, and FGF21 was knocked down in HASMCs to explore its mechanism of action. SGLT2i increased the expression of FGF21 and FGFR. SGLT2i improved Ang II-induced systolic blood pressure elevation, myocardial hypertrophy, vascular wall thickening, fibrosis, and oxidative stress in WT mice. These protective effects were reduced in FGF21-/- mice. Knockdown of FGF21 in HASMCs abolished the SGLT2i-induced upregulation of antioxidant markers and the downregulation of TGF-β and fibrosis-related proteins. SGLT2i-mediated blood pressure-lowering and vascular protective effects are primarily achieved through the activation of the FGF21/FGFR1.
Collapse
Affiliation(s)
- Manyu Dai
- Department of Cardiology, National Cardiovascular Disease Regional Center for Anhui, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Zhuoran Jia
- Department of Cardiology, National Cardiovascular Disease Regional Center for Anhui, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Huimin Wang
- Department of Cardiology, National Cardiovascular Disease Regional Center for Anhui, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Peiyang Zheng
- Department of Cardiology, National Cardiovascular Disease Regional Center for Anhui, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Yangcheng Xue
- Department of Cardiology, National Cardiovascular Disease Regional Center for Anhui, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Ke Gong
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei, 230031, China
| | - Ren Zhao
- Department of Cardiology, National Cardiovascular Disease Regional Center for Anhui, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China.
| |
Collapse
|
6
|
Huang ZL, Zhang SB, Xu SF, Gu XN, Wu ZQ, Zhang Y, Li J, Ji LL. TSG attenuated NAFLD and facilitated weight loss in HFD-fed mice via activating the RUNX1/FGF21 signaling axis. Acta Pharmacol Sin 2025:10.1038/s41401-025-01568-w. [PMID: 40307458 DOI: 10.1038/s41401-025-01568-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 04/14/2025] [Indexed: 05/02/2025]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a chronic liver disease characterized by steatosis in hepatocytes and is now becoming the major cause of liver-related mortality. Fibroblast growth factor 21 (FGF21) is an endocrine hormone mainly secreted by the liver, which can bind to its receptor (FGFR) and co-receptor beta klotho (KLB) to form a receptor complex, exerting its lipid-lowering function. 2,3,5,4'-Tetrahydroxy-stilbene-2-O-β-D-glucoside (TSG), a natural compound isolated from Polygonum multiflorum Thunb, has shown excellent activity in lowering lipid content and efficacy in improving NAFLD. In this study we investigated whether FGF21 was implicated in the therapeutic effect of TSG in NAFLD mice. NAFLD was induced in mice by feeding with a high-fat diet (HFD) for 12 weeks, and treated with TSG (20, 40 mg·kg-1·d-1, i.g.) during the last 4 weeks. We showed that TSG treatment significantly alleviated NAFLD in HFD-fed mice evidenced by reduced hepatic triglyceride (TG) and non-esterified fatty acids (NEFA), diminished lipid droplets and decreased NAFLD activity score (NAS) in liver tissues. We demonstrated that TSG treatment significantly increased the mRNA and protein levels of FGF21 in vitro and in vivo, and reduced lipid accumulation in both the liver and adipose tissues. Transcriptomics analysis revealed that TSG treatment significantly increased the nuclear translocation of a transcription factor RUNX1. Knockdown of Runx1 in HFD-fed mice eliminated the efficacy of TSG in alleviating NAFLD, reducing hepatic lipid accumulation and regulating FGF21 signaling pathway in liver and adipose tissues. In conclusion, TSG alleviates NAFLD by enhancing the FGF21-mediated lipid metabolism in a RUNX1-dependent manner.
Collapse
Affiliation(s)
- Zhen-Lin Huang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Shao-Bo Zhang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Shang-Fu Xu
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China
| | - Xin-Nan Gu
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ze-Qi Wu
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yue Zhang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jian Li
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, China
- Technology Center of Jinling Pharmaceutical Co., Ltd, Nanjing, 210009, China
| | - Li-Li Ji
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
7
|
Huijbers A, Korzilius JW, Morsche RT, van der Graaf M, Wanten GJA. Intestinal failure-associated steatosis and fibroblast growth factor 21 plasma levels among adult chronic intestinal failure patients. Clin Nutr ESPEN 2025; 68:1-7. [PMID: 40294745 DOI: 10.1016/j.clnesp.2025.04.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 04/14/2025] [Accepted: 04/23/2025] [Indexed: 04/30/2025]
Abstract
BACKGROUND & AIMS Adult patients with chronic intestinal failure (CIF) may develop intestinal failure-associated steatosis. Asymptomatic steatosis can lead to steatohepatitis and its downstream complications. Monitoring steatosis in daily practice in adult CIF patients is hampered by limited, reliable, accessible, non-invasive methods to measure liver fat content (LFC). Fibroblast growth factor 21 (FGF21) is a hormone that is mainly produced by hepatocytes, and higher plasma levels are associated with the presence and the degree of liver steatosis in several clinical conditions. Furthermore, FGF21 analogues have been shown to reduce fatty liver. FGF21 has previously been suggested as a biomarker for liver steatosis. The aim of this study was to assess the diagnostic performance of FGF21 plasma levels to detect steatosis and steatosis severity in adult CIF patients. METHODS FGF21 plasma levels were quantified using enzyme-linked immunosorbent assay (ELISA) in 48 adult CIF patients who had been receiving home parenteral nutrition (HPN) or intravenous fluids for ≥3 months for ≥2 times per week. Liver fat content (LFC, %) was assessed with proton magnetic resonance spectroscopy (1H-MRS). Patient characteristics of patients with steatosis (LFC >5.5 %) and without steatosis (LFC ≤5.5 %) were compared using the Mann-Whitney U test or Fisher's exact test. The diagnostic value of FGF21 levels to diagnose the presence of steatosis (LFC >5.5 %) was performed by determining the area under the receiver operating characteristics curve (AUC), and the optimal cut-off value was determined. Furthermore, Spearman's rho correlation coefficient was calculated to evaluate the association between FGF21 levels and LFC. RESULTS FGF21 plasma levels were measured in 48 patients (median age of 56 years, 71 % female) with a median duration of HPN use of 57 months. Steatosis was diagnosed in 8/48 (17 %) patients, with a median LFC of 8.4 % (range 5.7-39.9 %). CIF patients with steatosis had higher median FGF21 plasma levels (658 pg/mL) than patients without steatosis (299 pg/mL). The area under the curve (AUC) of FGF21 to predict steatosis (LFC >5.5 %) was 0.80 [95 % CI 0.63, 0.96]. With the optimal FGF21 cut-off point at 453 pg/mL, the sensitivity as well as the specificity was 75 %. The calculated Spearman rho correlation found a significant positive correlation (ρ = 0.65, p < 0.001) between FGF21 plasma levels and LFC (%). CONCLUSION Adult CIF patients with steatosis had higher FGF21 plasma levels than CIF patients without steatosis. FGF21 is a good predictor for diagnosing steatosis and has a good correlation with LFC. FGF21 should be considered as a biomarker for steatosis in adult patients with CIF.
Collapse
Affiliation(s)
- Angelique Huijbers
- Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Julia W Korzilius
- Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Rene Te Morsche
- Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | | | - Geert J A Wanten
- Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, the Netherlands.
| |
Collapse
|
8
|
Chen J, Li W, Yu L, Zhang B, Li Z, Zou P, Ding B, Dai X, Wang Q. Combined Effects of Ketogenic Diet and Aerobic Exercise on Skeletal Muscle Fiber Remodeling and Metabolic Adaptation in Simulated Microgravity Mice. Metabolites 2025; 15:270. [PMID: 40278399 PMCID: PMC12029359 DOI: 10.3390/metabo15040270] [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: 03/13/2025] [Revised: 04/11/2025] [Accepted: 04/11/2025] [Indexed: 04/26/2025] Open
Abstract
Objective: Prolonged microgravity environments impair skeletal muscle homeostasis by triggering fiber-type transitions and metabolic dysregulation. Although exercise and nutritional interventions may alleviate disuse atrophy, their synergistic effects under microgravity conditions remain poorly characterized. This study investigated the effects of an 8-week ketogenic diet combined with aerobic exercise in hindlimb-unloaded mice on muscle fiber remodeling and metabolic adaptation. Methods: Seven-week-old male C57BL/6J mice were randomly divided into six groups: normal diet control (NC), normal diet with hindlimb unloading (NH), normal diet with hindlimb unloading and exercise (NHE), ketogenic diet control (KC), ketogenic diet with hindlimb unloading (KH), and ketogenic diet with hindlimb unloading and exercise (KHE). During the last two weeks of intervention, hindlimb unloading was applied to simulate microgravity. Aerobic exercise groups performed moderate-intensity treadmill running (12 m/min, 60 min/day, and 6 days/week) for 8 weeks. Body weight, blood ketone, and glucose levels were measured weekly. Post-intervention assessments included the respiratory exchange ratio (RER), exhaustive exercise performance tests, and biochemical analyses of blood metabolic parameters. The skeletal muscle fiber-type composition was evaluated via immunofluorescence staining, lipid deposition was assessed using Oil Red O staining, glycogen content was analyzed by Periodic Acid-Schiff (PAS) staining, and gene expression was quantified using quantitative real-time PCR (RT-qPCR). Results: Hindlimb unloading significantly decreased body weight, induced muscle atrophy, and reduced exercise endurance in mice. However, the combination of KD and aerobic exercise significantly attenuated these adverse effects, as evidenced by increased proportions of oxidative muscle fibers (MyHC-I) and decreased proportions of glycolytic fibers (MyHC-IIb). Additionally, this combined intervention upregulated the expression of lipid metabolism-associated genes, including CPT-1b, HADH, PGC-1α, and FGF21, enhancing lipid metabolism and ketone utilization. These metabolic adaptations corresponded with improved exercise performance, demonstrated by the increased time to exhaustion in the KHE group compared to other hindlimb unloading groups. Conclusions: The combination of a ketogenic diet and aerobic exercise effectively ameliorates simulated microgravity-induced skeletal muscle atrophy and endurance impairment, primarily by promoting a fiber-type transition from MyHC-IIb to MyHC-I and enhancing lipid metabolism gene expression (CPT-1b, HADH, and PGC-1α). These findings underscore the potential therapeutic value of combined dietary and exercise interventions for mitigating muscle atrophy under simulated microgravity conditions.
Collapse
Affiliation(s)
- Jun Chen
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China;
- Sports Nutrition Center, National Institute of Sports Medicine, Beijing 100029, China
| | - Wenjiong Li
- National Key Laboratory of Space Medicine, Beijing 100094, China; (W.L.); (Z.L.); (P.Z.); (B.D.)
| | - Liang Yu
- Department of Exercise Physiology, Beijing Sport University, Beijing 100084, China; (L.Y.); (B.Z.)
| | - Bowei Zhang
- Department of Exercise Physiology, Beijing Sport University, Beijing 100084, China; (L.Y.); (B.Z.)
| | - Zhili Li
- National Key Laboratory of Space Medicine, Beijing 100094, China; (W.L.); (Z.L.); (P.Z.); (B.D.)
| | - Peng Zou
- National Key Laboratory of Space Medicine, Beijing 100094, China; (W.L.); (Z.L.); (P.Z.); (B.D.)
| | - Bai Ding
- National Key Laboratory of Space Medicine, Beijing 100094, China; (W.L.); (Z.L.); (P.Z.); (B.D.)
| | - Xiaoqian Dai
- National Key Laboratory of Space Medicine, Beijing 100094, China; (W.L.); (Z.L.); (P.Z.); (B.D.)
| | - Qirong Wang
- Sports Nutrition Center, National Institute of Sports Medicine, Beijing 100029, China
| |
Collapse
|
9
|
Shouman WA, Najmeddine S, Sinno L, Dib Nehme R, Ghawi A, Ziade JA, Altara R, Amin G, Booz GW, Zouein FA. Hepatokines and their role in cardiohepatic interactions in heart failure. Eur J Pharmacol 2025; 992:177356. [PMID: 39922419 PMCID: PMC11862882 DOI: 10.1016/j.ejphar.2025.177356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 01/29/2025] [Accepted: 02/05/2025] [Indexed: 02/10/2025]
Abstract
Heart failure is one of the leading causes of death and disease worldwide. It is a condition that affects multiple systems within the body. There is a large body of evidence supporting that the liver is a major organ involved in the pathogenesis of heart failure. Cardiac hepatopathy and cirrhotic cardiomyopathy are two conditions that are associated with poor clinical outcomes in patients with heart failure. Despite the extensive proposed explanations of the mechanisms entailing heart failure, there remains a gap in the role of proteins and metabolic regulators produced by hepatocytes and their effect on the development, progression, and prognosis of heart failure, including adverse cardiac remodeling, fibrosis, cardiac cachexia, and renal dysfunction associated with heart failure. The aim of this review is to identify the major hepatokines being studied (adropin, fetuin-A, fetuin-B, FGF-21, selenoprotein P and α1-microglobulin) as modulators of metabolic homeostasis and cardiac dysfunction in heart failure. Research suggests that these factors play a role in modulating oxidative stress, fibrosis, apoptosis, inflammatory responses, immune cell activation, mitochondrial dysfunction, and cellular migration. The exact role of each of these hepatokines is under on-going research and requires more investigations for future clinical use.
Collapse
Affiliation(s)
- Wael A Shouman
- Department of Pharmacology and Toxicology, American University of Beirut Faculty of Medicine, Beirut, Lebanon
| | - Sarah Najmeddine
- Department of Pharmacology and Toxicology, American University of Beirut Faculty of Medicine, Beirut, Lebanon
| | - Lilas Sinno
- Department of Pharmacology and Toxicology, American University of Beirut Faculty of Medicine, Beirut, Lebanon
| | - Ryan Dib Nehme
- Department of Pharmacology and Toxicology, American University of Beirut Faculty of Medicine, Beirut, Lebanon
| | - Alaa Ghawi
- Department of Pharmacology and Toxicology, American University of Beirut Faculty of Medicine, Beirut, Lebanon
| | - Joanna A Ziade
- Department of Pharmacology and Toxicology, American University of Beirut Faculty of Medicine, Beirut, Lebanon
| | - Raffaele Altara
- Department of Pathology, School of Medicine, University of Mississippi Medical Center, 14, Jackson, MS, USA; Department of Anatomy & Embryology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | - Ghadir Amin
- Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - George W Booz
- Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Fouad A Zouein
- Department of Pharmacology and Toxicology, American University of Beirut Faculty of Medicine, Beirut, Lebanon; The Cardiovascular, Renal, and Metabolic Diseases Research Center of Excellence, American University of Beirut Medical Center, Riad El-Solh, Beirut, Lebanon; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, MS, USA.
| |
Collapse
|
10
|
Takahama Y. FGF21 keeps the thymus young. NATURE AGING 2025; 5:534-535. [PMID: 39972171 DOI: 10.1038/s43587-025-00814-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Affiliation(s)
- Yousuke Takahama
- Thymus Biology Section, Experimental Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
11
|
Barros DR, Hegele RA. Fibroblast growth factor 21: update on genetics and molecular biology. Curr Opin Lipidol 2025; 36:88-95. [PMID: 39450972 PMCID: PMC11888835 DOI: 10.1097/mol.0000000000000960] [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] [Indexed: 10/26/2024]
Abstract
PURPOSE OF REVIEW Since its discovery, most research on fibroblast growth factor 21 (FGF21) has focused on its antihyperglycemia properties. However, attention has recently shifted towards elucidating the ability of FGF21 to lower circulating lipid levels and ameliorate liver inflammation and steatosis. We here discuss the physiology of FGF21 and its role in lipid metabolism, with a focus on genetics, which has up until now not been fully appreciated. RECENT FINDINGS New developments have uncovered associations of common small-effect variants of the FGF21 gene, such as the single nucleotide polymorphisms rs2548957 and rs838133, with numerous physiological, biochemical and behavioural phenotypes linked to energy metabolism and liver function. In addition, rare loss-of-function variants of the cellular receptors for FGF21 have been recently associated with severe endocrine and metabolic phenotypes. These associations corroborate the findings from basic studies and preliminary clinical investigations into the therapeutic potential of FGF21 for the treatment of metabolic dysfunction-associated steatotic liver disease (MASLD) and hypertriglyceridemia. Furthermore, recent breakthrough research has begun to dissect mechanisms of a potential FGF21 brain-adipose axis. Such inter-organ communication would be comparable to that seen with other potent metabolic hormones. A deeper understanding of FGF21 could prove to be further beneficial for drug development. SUMMARY FGF21 is a potent regulator of lipid and energy homeostasis and its physiology is currently at the centre of investigative efforts to develop agents targeting hypertriglyceridemia and MASLD.
Collapse
Affiliation(s)
- Daniel R Barros
- Departments of Medicine and Biochemistry, and Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | | |
Collapse
|
12
|
Xiao J, Li J, Wu X, Hao Y, Zhao X, Zhang W, Xu B, Ma T, Zhang L, Xiang R, Cui H, Yang C, Yan P, Tang M, Wang Y, Qu Y, Chen L, Liu Y, Zou Y, Zhang L, Liu Z, Yao Y, Yang C, Zhang B, Jiang X. Adult Height, Cardiovascular Disease, and the Underlying Mechanism: A Comprehensive Epidemiological and Genetic Analysis. Can J Cardiol 2025:S0828-282X(25)00237-5. [PMID: 40174860 DOI: 10.1016/j.cjca.2025.03.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 02/13/2025] [Accepted: 03/06/2025] [Indexed: 04/04/2025] Open
Abstract
BACKGROUND Adult height measures the complete growth of an individual and influences the development of cardiovascular disease (CVD). Despite recent within-sibling studies that have suggested minimal effects from environmental confounders, biological mechanisms underlying the height-CVD relationship remain elusive. METHODS Leveraging the large-scale UK Biobank data set and summary statistics from the latest genome-wide association studies, we reevaluated the effect of height on 8 major CVD subtypes. Phenotypic associations were determined using Cox proportional hazard analysis. Putative causal relationships were assessed using univariable Mendelian randomization. Mediation analysis and 2-step Mendelian randomization were further performed to investigate the mediation effect of 15 common cardiometabolic or pulmonary risk factors. RESULTS Height was consistently associated with a decreased risk of coronary artery disease (CAD), confirmed in epidemiological (hazard ratio, 0.90; 95% confidence interval [CI], 0.88-0.91) and genetic (odds ratio, 0.89, 95% CI, 0.86-0.92) analysis. Forced vital capacity was identified as the most significant mediator for the height-CAD relationship in epidemiological (proportion-mediated, 65.6%; 95% CI, 53.1%-78.0%) and genetic (proportion-mediated, 46.2%; 95% CI, 5.0%-87.5%) analysis. Notably, obesity, and blood pressure, lipid, and C-reactive protein levels also exhibited significant mediatory effects. Despite a consistent risk effect of height on atrial fibrillation and venous thromboembolism, no promising mediator was identified. CONCLUSIONS Our study confirms the health effects of height on CAD, atrial fibrillation, and venous thromboembolism and emphasizes forced vital capacity as the primary pathway that links height to CAD. Importantly, it indicates that the CAD risk associated with nonmodifiable height could be mitigated through enhanced lung function and cardiometabolic conditions.
Collapse
Affiliation(s)
- Jinyu Xiao
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Jiayuan Li
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Xueyao Wu
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yu Hao
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Xunying Zhao
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Wenqiang Zhang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Bin Xu
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Tianpei Ma
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Li Zhang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Rong Xiang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Huijie Cui
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Chao Yang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Peijing Yan
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Mingshuang Tang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yutong Wang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yang Qu
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Lin Chen
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yunjie Liu
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yanqiu Zou
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Ling Zhang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China; Department of Iatrical Polymer Material and Artificial Apparatus, School of Polymer Science and Engineering, Sichuan University, Chengdu, China
| | - Zhenmi Liu
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China; Department of Maternal, Child and Adolescent Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yuqin Yao
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China; Department of Occupational and Environmental Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Chunxia Yang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Ben Zhang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China; Department of Cardiology, Department of Neurology, and Department of Oncology, Hainan General Hospital and Hainan Affiliated Hospital, Hainan Medical University, Haikou, China.
| | - Xia Jiang
- Department of Epidemiology and Biostatistics, Institute of Systems Epidemiology, West China-PUMC C. C. Chen Institute of Health, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China; Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China; Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
13
|
Qiu S, Ding X, Ma X, Zhang L, Chen J, Wei W. Muscle cells affect the promoting effect of FGF21 on lipid accumulation in porcine adipocytes through AhR/FGFR1 signaling pathway. Biochem Biophys Res Commun 2025; 754:151520. [PMID: 40015071 DOI: 10.1016/j.bbrc.2025.151520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 02/19/2025] [Accepted: 02/20/2025] [Indexed: 03/01/2025]
Abstract
The intramuscular fat (IMF) content, as an important meat quality trait, can directly affect the tenderness, juiciness, and flavor of pork. Reasonably increasing the IMF content can improve the palatability of pork. Therefore, identification of important factors for the lipid accumulation among muscles is the breakthrough point for improving meat quality. FGF21, identified as a novel metabolic regulator, has been found to regulate glucose and lipid metabolism in 3T3-L1 adipocytes, but its function in porcine adipocytes remains unclear. In this study, we discovered that the administration of recombinant FGF21 protein promotes adipogenic differentiation and increases triglyceride accumulation in porcine adipocytes. While the expression of FGFR1 in adipocytes under muscle conditions is inhibited, affecting the signal transduction of FGF21. This inhibitory effect is accompanied by activation of the AhR signaling pathway. When treated with the AhR antagonist CH223191, there was a partial restoration of FGFR1 expression levels. This indicates that muscle cells suppress the expression of FGFR1 in adipocytes by activating the AhR signaling pathway, thereby affecting the signal transduction of FGF21. Our results reveal the regulatory role of FGF21 in pig adipocyte differentiation and the regulatory mechanism of muscle environment on FGFR1 expression, providing new theoretical basis for IMF content improvement from the perspective of FGF21-FGFR1 signaling transduction.
Collapse
Affiliation(s)
- Shengda Qiu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Xiaolei Ding
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Xiangfei Ma
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Lifan Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Jie Chen
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Wei Wei
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
14
|
McLeod K, Datta V, Fuller S. Adipokines as Cardioprotective Factors: BAT Steps Up to the Plate. Biomedicines 2025; 13:710. [PMID: 40149686 PMCID: PMC11940801 DOI: 10.3390/biomedicines13030710] [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: 01/10/2025] [Revised: 03/07/2025] [Accepted: 03/11/2025] [Indexed: 03/29/2025] Open
Abstract
Cardiovascular disease is the leading cause of death throughout most of the industrialized world. Metabolic syndrome (MetS) and its associated pathologies are underlying factors in the etiology of cardiovascular disease, as well as a plethora of other maladies which cause excess morbidity and mortality. Adipose tissue (AT) has come to be regarded as a bona fide endocrine organ which secretes specific molecular entities constituting part of a complex web of inter-organ crosstalk that functions as a key determinant of whole-body metabolic phenotype. Brown adipose tissue (BAT) has classically been regarded as a thermogenic tissue exerting its metabolic effects primarily through its capacity to oxidize substrates decoupled from ATP resynthesis, thereby resulting in increased energy expenditure (EE) and heat production. However, in recent years, BAT has begun to receive attention as a secretory organ in its own right. The molecules secreted specifically by BAT have been termed "batokines", and currently available evidence supports the notion that batokines exert favorable metabolic effects on multiple organ systems. While maintenance of healthy body composition by conferring resistance to excessive adiposity is a rather obvious mechanism by which BAT operates via increased EE, effects on critical organs such as the heart remain unclear. This narrative review focuses on four types of batokines (FGF21, neuregulin 4, 12,13-diHOME, and BAT-derived microRNAs) for which evidence of modulation of cardiovascular function exists in the context of pathological states such as hypertension, atherosclerosis, and ischemia/reperfusion injury. Given the overwhelming burden of cardiometabolic disease, further study of the functions of BAT and its secretome is warranted and will intensify in the future.
Collapse
Affiliation(s)
- Keely McLeod
- School of Kinesiology, University of Louisiana at Lafayette, Lafayette, LA 70506, USA; (K.M.); (V.D.)
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, USA
| | - Victoria Datta
- School of Kinesiology, University of Louisiana at Lafayette, Lafayette, LA 70506, USA; (K.M.); (V.D.)
| | - Scott Fuller
- School of Kinesiology, University of Louisiana at Lafayette, Lafayette, LA 70506, USA; (K.M.); (V.D.)
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, USA
| |
Collapse
|
15
|
Phan P, Hoang J, Kumar TKS. Overexpression and biophysical and functional characterization of a recombinant FGF21. BIOPHYSICAL REPORTS 2025; 5:100198. [PMID: 39884432 PMCID: PMC11869967 DOI: 10.1016/j.bpr.2025.100198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 01/17/2025] [Accepted: 01/24/2025] [Indexed: 02/01/2025]
Abstract
Fibroblast growth factor 21 (FGF21) is an endocrine FGF that plays a vital role in regulating essential metabolic pathways. FGF21 increases glucose uptake by cells, promotes fatty acid oxidation, reduces blood glucose levels, and alleviates metabolic diseases. However, detailed studies on its stability and biophysical characteristics have not been reported. Herein, we present the overexpression, biophysical characterization, and metabolic activity of a soluble recombinant FGF21 (rFGF21). The far-UV circular dichroism spectra of rFGF21 show a negative trough at 215 nm, indicating that the protein's backbone predominantly adopts a β sheet conformation. rFGF21 shows intrinsic tyrosine fluorescence at 305 nm. Thermal denaturation using differential scanning calorimetry reveals that rFGF21 is relatively thermally unstable, with a melting temperature of 46.8°C (±0.1°C). The urea-induced unfolding of rFGF21 is rapid, with a chemical transition midpoint of 0.4 M. rFGF21 is readily cleaved by trypsin in limited trypsin digestion assays. Isothermal titration calorimetry experiments show that rFGF21 does not bind to heparin. Interestingly, rFGF21 demonstrates proliferative activity in NIH/3T3 fibroblasts and enhances mitochondrial oxidative phosphorylation and fatty acid oxidation in 3T3-L1 adipocytes. These findings provide a crucial framework for the engineering of novel structure-based variants of FGF21 with improved stability and biological activity to treat metabolic disorders.
Collapse
Affiliation(s)
- Phuc Phan
- Department of Chemistry and Biochemistry, Fulbright College of Art and Sciences, University of Arkansas, Fayetteville, Arkansas
| | - Jason Hoang
- Department of Chemistry and Biochemistry, Fulbright College of Art and Sciences, University of Arkansas, Fayetteville, Arkansas
| | | |
Collapse
|
16
|
Li J, Liu T, Xian M, Zhou K, Wei J. The Power of Exercise: Unlocking the Biological Mysteries of Peripheral-Central Crosstalk in Parkinson's Disease. J Adv Res 2025:S2090-1232(25)00143-2. [PMID: 40049515 DOI: 10.1016/j.jare.2025.03.001] [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: 07/23/2024] [Revised: 01/06/2025] [Accepted: 03/01/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Exercise is a widely recognized non-pharmacological treatment for Parkinson's Disease (PD). The bidirectional regulation between the brain and peripheral organs has emerged as a promising area of research, with the mechanisms by which exercise impacts PD closely linked to the interplay between peripheral signals and the central nervous system. AIM OF REVIEW This review aims to summarize the mechanisms by which exercise influences peripheral-central crosstalk to improve PD, discuss the molecular processes mediating these interactions, elucidate the pathways through which exercise may modulate PD pathophysiology, and identify directions for future research. KEY SCIENTIFIC CONCEPTS OF REVIEW This review examines how exercise-induced cytokine release promotes neuroprotection in PD. It discusses how exercise can stimulate cytokine secretion through various pathways, including the gut-brain, muscle-brain, liver-brain, adipose-brain, and bone-brain axes, thereby alleviating PD symptoms. Additionally, the potential contributions of the heart-brain, lung-brain, and spleen-brain axes, as well as multi-axis crosstalk-such as the brain-gut-muscle and brain-gut-bone axes-are explored in the context of exercise therapy. The study highlights the need for further research into peripheral-central crosstalk and outlines future directions to address challenges in clinical PD therapy.
Collapse
Affiliation(s)
- Jingwen Li
- Institute for Sports and Brain Health, School of Physical Education, Henan University, Kaifeng, Henan, 475004, China
| | - Tingting Liu
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Meiyan Xian
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Ke Zhou
- Institute for Sports and Brain Health, School of Physical Education, Henan University, Kaifeng, Henan, 475004, China.
| | - Jianshe Wei
- Institute for Sports and Brain Health, School of Physical Education, Henan University, Kaifeng, Henan, 475004, China; Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China.
| |
Collapse
|
17
|
Theofilis P, Oikonomou E, Karakasis P, Pamporis K, Dimitriadis K, Kokkou E, Lambadiari V, Siasos G, Tsioufis K, Tousoulis D. FGF21 Analogues in Patients With Metabolic Diseases: Systematic Review and Meta-Analysis of Randomised Controlled Trials. Liver Int 2025; 45:e70016. [PMID: 39898512 DOI: 10.1111/liv.70016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 12/26/2024] [Accepted: 01/24/2025] [Indexed: 02/04/2025]
Abstract
BACKGROUND AND AIMS Liver-related complications are frequent in patients with metabolic diseases, with limited treatment options currently available. This systematic review and meta-analysis aimed to assess the effect of fibroblast growth factor-21 (FGF21) analogues on hepatic steatosis, inflammation and fibrosis in patients with metabolic diseases. METHODS We conducted a systematic literature search in Pubmed, Scopus and Web of Science for randomised controlled trials (RCTs) assessing the effect of FGF21 analogues on hepatic steatosis evaluated by hepatic fat fraction (HFF), inflammation and fibrosis compared to placebo. Adverse events (AEs) were also recorded. RESULTS Treatment with FGF21 analogues was associated with metabolic-associated steatohepatitis (MASH) resolution without fibrosis worsening (5 studies, risk ratio [RR] 4.40, 95% confidence interval [CI]: 2.41, 8.03, p < 0.001) and fibrosis improvement by 1 grade without MASH worsening (6 studies, RR 1.79, 95% CI: 1.24, 2.59, p = 0.002). FGF21 analogues significantly lowered HFF compared to placebo (6 studies, SMD -1.08, 95% CI: -1.28, -0.88, p < 0.001), while patients receiving FGF21 analogues were more likely to exhibit a reduction in HFF by 30% (10 studies, RR 4.08, 95% CI: 3.08, 5.40, p < 0.001) or 50% (6 studies, RR 10.43, 95% CI: 5.47, 19.87, p < 0.001). HFF normalisation (≤ 5%) was more frequently achieved with FGF21 analogues (6 studies, RR 14.58, 95% CI: 4.70, 45.18, p < 0.001). The results remained robust after sensitivity analyses. Serious AE and AE leading to drug discontinuation were similar in patients receiving FGF21 analogues or placebo. CONCLUSIONS FGF21 analogues can reduce hepatic steatosis, inflammation and fibrosis in patients with metabolic diseases, representing a possible treatment option for steatotic liver disease.
Collapse
Affiliation(s)
- Panagiotis Theofilis
- 1st Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Evangelos Oikonomou
- 3rd Department of Cardiology, Thoracic Diseases General Hospital "Sotiria", National and Kapodistrian University of Athens, Athens, Greece
| | - Paschalis Karakasis
- 2nd Department of Cardiology, Hippokration General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Konstantinos Pamporis
- 1st Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Kyriakos Dimitriadis
- 1st Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Eleni Kokkou
- 3rd Department of Cardiology, Thoracic Diseases General Hospital "Sotiria", National and Kapodistrian University of Athens, Athens, Greece
| | - Vaia Lambadiari
- Second Department of Internal Medicine, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Gerasimos Siasos
- 3rd Department of Cardiology, Thoracic Diseases General Hospital "Sotiria", National and Kapodistrian University of Athens, Athens, Greece
- Cardiovascular Division, Harvard Medical School, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Konstantinos Tsioufis
- 1st Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitris Tousoulis
- 1st Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
18
|
Zhang Z, Cui M, Wang H, Yuan W, Liu Z, Gao H, Guan X, Chen X, Xie L, Chen S, He Y, Wang Q. Co-exposure to F-53B and nanoplastics induced hepatic glucolipid metabolism disorders by the PI3K-AKT signaling pathway. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 368:125771. [PMID: 39894156 DOI: 10.1016/j.envpol.2025.125771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 01/28/2025] [Accepted: 01/28/2025] [Indexed: 02/04/2025]
Abstract
Recent investigations suggest that the chemical compound F-53B (6:2 chlorinated polyfluorinated ether sulfonate) may pose risks of liver toxicity. Within environmental settings, F-53B attaches to microplastics and nanoplastics, which are capable of being consumed by diverse species. To investigate the synergistic effects on hepatotoxicity, adult male mice were subjected to F-53B at daily doses of 1, 10, and 100 μg/kg, NPs at 100 mg/kg per day, or a combination of both treatments for a duration of 2 months. The results indicated that NPs moderately increased the buildup of F-53B within both the liver and plasma. Co-exposure to F-53B (100 μg/kg/day) and NPs induced hepatocellular edema and elevated plasma ALT levels, which were rarely observed in groups exposed to F-53B or NPs alone. Additionally, we found that co-exposure decreased the concentrations of total cholesterol (TC) and triglycerides (TG) in both plasma and liver tissues, while increasing fasting plasma glucose and insulin levels. Transcriptomic analysis revealed that the PI3K-AKT signaling pathway is potentially involved in mediating hepatic metabolic disorders. Further experiments demonstrated that the combined treatment significantly suppressed the expression of FGF21, an upstream regulator of the PI3K-AKT pathway. This alteration resulted in the suppression of PI3K-regulated gene expression associated with glucose and lipid metabolism. The findings suggest that F-53B impairs hepatic glucolipid metabolism in mice by suppressing of the PI3K-AKT signaling cascade, with NPs amplifying its toxicity.
Collapse
Affiliation(s)
- Zhihan Zhang
- Department of Toxicology, School of Public Health, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Mengxing Cui
- Department of Toxicology, School of Public Health, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Han Wang
- Department of Toxicology, School of Public Health, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Wenke Yuan
- Department of Toxicology, School of Public Health, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Ziqi Liu
- Department of Toxicology, School of Public Health, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Huan Gao
- Department of Toxicology, School of Public Health, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xinchao Guan
- Department of Toxicology, School of Public Health, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xiaoyu Chen
- Department of Toxicology, School of Public Health, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Lijie Xie
- Department of Toxicology, School of Public Health, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Shilin Chen
- Department of Toxicology, School of Public Health, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Yujie He
- Department of Toxicology, School of Public Health, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Qing Wang
- Department of Toxicology, School of Public Health, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
19
|
Song L, Ye H, Lv Z, Liu Y, Lu Z, Chen J, Pan H, Cai L, Chen Y, Huang S, Zan X, Huang X, Yu C. Hexahistidine-metal assembly encapsulated fibroblast growth factor 21 for lipopolysaccharide-induced acute lung injury. Eur J Pharm Biopharm 2025; 208:114650. [PMID: 39870250 DOI: 10.1016/j.ejpb.2025.114650] [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: 08/01/2024] [Revised: 01/16/2025] [Accepted: 01/24/2025] [Indexed: 01/29/2025]
Abstract
Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) represents a spectrum of potentially fatal conditions that currently lack effective drug treatment. Recent researches suggest that Fibroblast Growth Factor 21 (FGF21) may protect against ALI/ARDS. However, the clinical use of FGF21 is limited by its rapid degradation, restricted targeting capabilities, and numerous adverse effects. Addressing this challenge, the study employs a pH-responsive nanoparticle delivery system known as Hexahistidine-metal Assembly (HmA) for administering FGF21. The entrapment efficiency (EE%) and loading capacity (LCwt%) of HmA exceed 90 % and 35 %, respectively, while the HmA@FGF21 nanoparticles exhibit an average size of 130 nm, a PDI value of approximately 0.28, and a zeta potential of 24 mV. In animal experiments, HmA@FGF21 administered in lipopolysaccharide (LPS)-induced lung injury significantly exceed those of standalone FGF21, including mitigating the pathological manifestations and reducing the wet/dry ratio, total protein concentration, and overall cell count in BALF of ALI, whether administered via the airway or intravenously. This therapeutic approach therefore shows promise for precise delivery of FGF21 to the lungs to treat ALI, and may offer a novel, and efficient method for delivery of potential pharmacological agents to address other lung diseases.
Collapse
Affiliation(s)
- Lanlan Song
- Division of Pulmonary Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325000, China
| | - Huihui Ye
- Division of Pulmonary Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325000, China
| | - Zhanghang Lv
- Division of Pulmonary Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325000, China
| | - Yichen Liu
- Division of Pulmonary Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325000, China
| | - Ziyi Lu
- Division of Pulmonary Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325000, China
| | - Jun Chen
- Division of Pulmonary Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325000, China
| | - Haofeng Pan
- Division of Pulmonary Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325000, China
| | - Luqiong Cai
- Division of Pulmonary Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325000, China
| | - Yuxin Chen
- Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Shiqing Huang
- Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Xingjie Zan
- Joint Centre of Translational Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China.
| | - Xiaoying Huang
- Division of Pulmonary Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou Key Laboratory of Interdiscipline and Translational Medicine, Wenzhou Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325000, China.
| | - Chang Yu
- Intervention Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| |
Collapse
|
20
|
Ghanem M, Archer G, Justet A, Jaillet M, Vasarmidi E, Mordant P, Castier Y, Mal H, Cazes A, Poté N, Crestani B, Mailleux A. FGF21 Signaling Exerts Antifibrotic Properties during Pulmonary Fibrosis. Am J Respir Crit Care Med 2025; 211:486-498. [PMID: 39637324 DOI: 10.1164/rccm.202311-2021oc] [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: 11/03/2023] [Accepted: 12/05/2024] [Indexed: 12/07/2024] Open
Abstract
Rationale: Idiopathic pulmonary fibrosis (IPF) is a lethal disease with limited therapeutic options. FGF21 (fibroblast growth factor 21), an endocrine fibroblast growth factor that acts through the FGFR1 (fibroblast growth factor receptor 1)/KLB (β-Klotho) pathway, mitigates liver fibrosis. Objectives: We hypothesized that FGF21 could exert antifibrotic properties in the lung. Methods: The concentrations of FGF21 and KLB in the plasma of patients with IPF and control subjects were assessed. Pulmonary fibrosis development was assessed in Fgf21-deficient mice compared with wild-type littermates, at Day 14 (D14) after the intratracheal injection of bleomycin. We determined the effect of repeated subcutaneous injections of a PEGylated FGF21 analog at D7, D10, D14, and D17 after bleomycin on the development of pulmonary fibrosis. Mice were killed at D21. The effects of FGF21, alone or with KLB, on apoptosis in murine lung epithelial 15 cells and on the phenotype of human lung fibroblasts were assessed in vitro. Measurements and Main Results: In the plasma of patients with IPF, FGF21 concentrations were increased, while KLB concentrations were decreased. Fgf21-deficient mice showed increased sensitivity to bleomycin in comparison with their wild-type littermates. Treatment with PEGylated FGF21 mitigated lung fibrogenesis, as evidenced by a lower injury score and decreased fibrosis markers and profibrotic mediator expression compared with the control group receiving the diluent. In murine lung epithelial 15 cells, stimulation with FGF21 and KLB inhibited apoptosis, through the decrease of BAX and BIM. Fibroblastic phenotype remained unaltered. Conclusions: Our data indicate a possible antifibrotic effect of FGF21 in the lung achieved through the inhibition of alveolar type 2 cell apoptosis.
Collapse
Affiliation(s)
- Mada Ghanem
- Faculté de Médecine Xavier Bichat, Université Paris Cité, Institut National de la Santé et de la Recherche Médicale, UMR1152, FHU APOLLO, Labex INFLAMEX, Paris, France; and
| | - Gabrielle Archer
- Faculté de Médecine Xavier Bichat, Université Paris Cité, Institut National de la Santé et de la Recherche Médicale, UMR1152, FHU APOLLO, Labex INFLAMEX, Paris, France; and
| | - Aurélien Justet
- Faculté de Médecine Xavier Bichat, Université Paris Cité, Institut National de la Santé et de la Recherche Médicale, UMR1152, FHU APOLLO, Labex INFLAMEX, Paris, France; and
| | - Madeleine Jaillet
- Faculté de Médecine Xavier Bichat, Université Paris Cité, Institut National de la Santé et de la Recherche Médicale, UMR1152, FHU APOLLO, Labex INFLAMEX, Paris, France; and
| | - Eirini Vasarmidi
- Faculté de Médecine Xavier Bichat, Université Paris Cité, Institut National de la Santé et de la Recherche Médicale, UMR1152, FHU APOLLO, Labex INFLAMEX, Paris, France; and
| | | | | | - Hervé Mal
- Service de Pneumologie et Transplantation
| | - Aurélie Cazes
- Faculté de Médecine Xavier Bichat, Université Paris Cité, Institut National de la Santé et de la Recherche Médicale, UMR1152, FHU APOLLO, Labex INFLAMEX, Paris, France; and
- Département d'Anatomopathologie, and
| | - Nicolas Poté
- Faculté de Médecine Xavier Bichat, Université Paris Cité, Institut National de la Santé et de la Recherche Médicale, UMR1152, FHU APOLLO, Labex INFLAMEX, Paris, France; and
- Département d'Anatomopathologie, and
| | - Bruno Crestani
- Faculté de Médecine Xavier Bichat, Université Paris Cité, Institut National de la Santé et de la Recherche Médicale, UMR1152, FHU APOLLO, Labex INFLAMEX, Paris, France; and
- Service de Pneumologie A, Centre de Référence des Maladies Pulmonaires Rares, Assistance Publique-Hôpitaux de Paris, Hôpital Bichat, Paris, France
| | - Arnaud Mailleux
- Faculté de Médecine Xavier Bichat, Université Paris Cité, Institut National de la Santé et de la Recherche Médicale, UMR1152, FHU APOLLO, Labex INFLAMEX, Paris, France; and
| |
Collapse
|
21
|
Ru Q, Li Y, Zhang X, Chen L, Wu Y, Min J, Wang F. Iron homeostasis and ferroptosis in muscle diseases and disorders: mechanisms and therapeutic prospects. Bone Res 2025; 13:27. [PMID: 40000618 PMCID: PMC11861620 DOI: 10.1038/s41413-024-00398-6] [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: 09/05/2024] [Revised: 11/23/2024] [Accepted: 12/16/2024] [Indexed: 02/27/2025] Open
Abstract
The muscular system plays a critical role in the human body by governing skeletal movement, cardiovascular function, and the activities of digestive organs. Additionally, muscle tissues serve an endocrine function by secreting myogenic cytokines, thereby regulating metabolism throughout the entire body. Maintaining muscle function requires iron homeostasis. Recent studies suggest that disruptions in iron metabolism and ferroptosis, a form of iron-dependent cell death, are essential contributors to the progression of a wide range of muscle diseases and disorders, including sarcopenia, cardiomyopathy, and amyotrophic lateral sclerosis. Thus, a comprehensive overview of the mechanisms regulating iron metabolism and ferroptosis in these conditions is crucial for identifying potential therapeutic targets and developing new strategies for disease treatment and/or prevention. This review aims to summarize recent advances in understanding the molecular mechanisms underlying ferroptosis in the context of muscle injury, as well as associated muscle diseases and disorders. Moreover, we discuss potential targets within the ferroptosis pathway and possible strategies for managing muscle disorders. Finally, we shed new light on current limitations and future prospects for therapeutic interventions targeting ferroptosis.
Collapse
Affiliation(s)
- Qin Ru
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xi Zhang
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Lin Chen
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Yuxiang Wu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China.
| | - Junxia Min
- The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China.
| | - Fudi Wang
- The Second Affiliated Hospital, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
22
|
Li J, Jiang H, Bai W, Yang Y, Zhou G, Chen W, Shao J. Fibroblast Growth Factor 21 Protects Against Cerebral Ischemia/Reperfusion Injury by Inhibiting Oxidative Stress and Ferroptosis. Neuropsychiatr Dis Treat 2025; 21:355-371. [PMID: 40027603 PMCID: PMC11871945 DOI: 10.2147/ndt.s504180] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 02/14/2025] [Indexed: 03/05/2025] Open
Abstract
Purpose Cerebral ischemia/reperfusion injury (CIRI) severely impacts patient outcomes and quality of life, with limited treatment options. Although fibroblast growth factor21 (FGF21) is known for its metabolic and anti-inflammatory effects, its role and mechanisms in CIRI are not well explored. Methods After developing an MCAO/R injury model, mice received intraperitoneal injections of FGF21 (1.5 mg/kg) 15 min pre-reperfusion, as well as 8 and 16 h post-reperfusion. The TTC, TUNEL, H&E, and Nissl stainings were used 24 h post-reperfusion to determine the infarct volume, apoptotic cells, brain pathological damage, and nerve cell survival, respectively. ELISA and Western blotting were employed to detect oxidative stress (OxS) products and ferroptosis-related markers. RNA-seq of the ischemic penumbra tissues was conducted, followed by bioinformatics analysis to screen and identify differentially expressed genes (DEGs). Then, we used qPCR to validate relevant molecule mRNA expression while using immunofluorescence staining to assess CYBB protein localization and expression. Results The FGF21 reduced the infarct volume in MCAO/R-injured mice, diminished apoptotic cell numbers, and alleviated pathological damage to ischemic brain tissue. Furthermore, FGF21 inhibited OxS and ferroptosis post-CIRI. RNA-seq revealed a significant differential expression of numerous genes, extensively involving diverse biological processes post- ischemia/reperfusion injury (IRI). Bioinformatics analysis and validation results indicated that CYBB was the most significantly differentially expressed ferroptosis-related molecule, and it may be a novel key regulatory molecule mediating anti-IRI of FGF21. Conclusion FGF21 protects CIRI by inhibiting OxS and ferroptosis. The CYBB, a new key regulator, may mediate its anti-ferroptotic effects, offering new insights into CIRI therapies.
Collapse
Affiliation(s)
- Junjie Li
- Department of Anesthesiology, the First Affiliated Hospital of Kunming Medical University, Kunming City, Yunnan Province, People’s Republic of China
| | - Haiyan Jiang
- Department of Anesthesiology, the First Affiliated Hospital of Kunming Medical University, Kunming City, Yunnan Province, People’s Republic of China
| | - Wenya Bai
- Department of Anesthesiology, the First Affiliated Hospital of Kunming Medical University, Kunming City, Yunnan Province, People’s Republic of China
| | - Yuan Yang
- Department of Anesthesiology, the First Affiliated Hospital of Kunming Medical University, Kunming City, Yunnan Province, People’s Republic of China
| | - Guilin Zhou
- Department of Anesthesiology, the First Affiliated Hospital of Kunming Medical University, Kunming City, Yunnan Province, People’s Republic of China
| | - Wendong Chen
- Department of Anesthesiology, the First Affiliated Hospital of Kunming Medical University, Kunming City, Yunnan Province, People’s Republic of China
| | - Jianlin Shao
- Department of Anesthesiology, the First Affiliated Hospital of Kunming Medical University, Kunming City, Yunnan Province, People’s Republic of China
| |
Collapse
|
23
|
Rao Z, Tang Y, Zhu J, Lu Z, Chen Z, Wang J, Bao Y, Mukondiwa AV, Wang C, Wang X, Luo Y, Li X. Enhanced FGF21 Delivery via Neutrophil-Membrane-Coated Nanoparticles Improves Therapeutic Efficacy for Myocardial Ischemia-Reperfusion Injury. NANOMATERIALS (BASEL, SWITZERLAND) 2025; 15:346. [PMID: 40072149 PMCID: PMC11901824 DOI: 10.3390/nano15050346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 02/15/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025]
Abstract
Acute myocardial infarction, a leading cause of death globally, is often associated with cardiometabolic disorders such as atherosclerosis and metabolic syndrome. Metabolic treatment of these disorders can improve cardiac outcomes, as exemplified by the GLP-1 agonist semaglutide. Fibroblast growth factor 21 (FGF21), a novel metabolic regulator, plays pivotal roles in lipid mobilization and energy conversion, reducing lipotoxicity, inflammation, mitochondrial health, and subsequent tissue damage in organs such as the liver, pancreas, and heart. Here, we test the therapeutic efficacy of FGF21 in mice with ischemia-reperfusion (I/R) injury, a model of acute myocardial infarction. We employed the strategic method of coating the FGF21-encapsulating liposomal nanoparticles with a neutrophil membrane designed to camouflage FGF21 from macrophage-mediated efferocytotic clearance and promote its targeted accumulation at I/R foci due to the inherent neutrophilic attraction to the inflammatory site. Our findings revealed that the coated FGF21 nanoparticles markedly accumulated within the lesions with a prolonged half-life, in additional to the liver, leading to substantial improvements in cardiac performance by enhancing mitochondrial energetic function and reducing oxidative stress, inflammation, and cell death. Therefore, our research highlights a viable strategy for the enhanced delivery of therapeutical FGF21 analogs to lesions beyond the liver following myocardial infarction.
Collapse
Affiliation(s)
- Zhiheng Rao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China; (Z.R.); (Y.T.); (J.Z.); (Z.L.); (Z.C.); (J.W.); (Y.B.); (A.V.M.); (C.W.); (X.W.)
| | - Yuli Tang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China; (Z.R.); (Y.T.); (J.Z.); (Z.L.); (Z.C.); (J.W.); (Y.B.); (A.V.M.); (C.W.); (X.W.)
| | - Jiamei Zhu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China; (Z.R.); (Y.T.); (J.Z.); (Z.L.); (Z.C.); (J.W.); (Y.B.); (A.V.M.); (C.W.); (X.W.)
| | - Zhenzhen Lu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China; (Z.R.); (Y.T.); (J.Z.); (Z.L.); (Z.C.); (J.W.); (Y.B.); (A.V.M.); (C.W.); (X.W.)
| | - Zhichao Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China; (Z.R.); (Y.T.); (J.Z.); (Z.L.); (Z.C.); (J.W.); (Y.B.); (A.V.M.); (C.W.); (X.W.)
| | - Jiaojiao Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China; (Z.R.); (Y.T.); (J.Z.); (Z.L.); (Z.C.); (J.W.); (Y.B.); (A.V.M.); (C.W.); (X.W.)
| | - Yuxuan Bao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China; (Z.R.); (Y.T.); (J.Z.); (Z.L.); (Z.C.); (J.W.); (Y.B.); (A.V.M.); (C.W.); (X.W.)
| | - Alan Vengai Mukondiwa
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China; (Z.R.); (Y.T.); (J.Z.); (Z.L.); (Z.C.); (J.W.); (Y.B.); (A.V.M.); (C.W.); (X.W.)
| | - Cong Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China; (Z.R.); (Y.T.); (J.Z.); (Z.L.); (Z.C.); (J.W.); (Y.B.); (A.V.M.); (C.W.); (X.W.)
| | - Xiaojie Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China; (Z.R.); (Y.T.); (J.Z.); (Z.L.); (Z.C.); (J.W.); (Y.B.); (A.V.M.); (C.W.); (X.W.)
| | - Yongde Luo
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China; (Z.R.); (Y.T.); (J.Z.); (Z.L.); (Z.C.); (J.W.); (Y.B.); (A.V.M.); (C.W.); (X.W.)
- Oujiang Laboratory, Wenzhou 325000, China
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Xiaokun Li
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China; (Z.R.); (Y.T.); (J.Z.); (Z.L.); (Z.C.); (J.W.); (Y.B.); (A.V.M.); (C.W.); (X.W.)
- Oujiang Laboratory, Wenzhou 325000, China
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| |
Collapse
|
24
|
Zhao L, Li J, Dang Y, Fisher D, Hien NTT, Musabaev E, Pronyuk K, Zhao L. Protective role of sulforaphane in lipid metabolism-related diseases. Mol Biol Rep 2025; 52:241. [PMID: 39961997 DOI: 10.1007/s11033-025-10358-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: 12/05/2024] [Accepted: 02/11/2025] [Indexed: 05/09/2025]
Abstract
Sulforaphane (SFN) is a phytochemical bioactive substance commonly found in cruciferous plants, such as broccoli and mustard. It has been reported to possess antibacterial, anti-inflammatory, anti-oxidant, anti-cancer and autophagy regulating properties. Recent studies have revealed that SFN regulates fat metabolism both in vivo and in vitro through various mechanisms, including alleviating endoplasmic reticulum stress, inhibiting inflammatory response and improving mitochondrial dysfunction, involving Nrf2/ARE, NF-κB, NLRP3 inflammasome, HDAC8-PGC1α axis and other signaling pathways. By curbing complications associated with abnormal fat metabolic diseases, SFN exhibits therapeutic effects on conditions like obesity, fatty liver disease, atherosclerosis, type 2 diabetes, etc., with minimal side effects. Therefore, it holds promise as a potential alternative treatment for lipid metabolism-related diseases. Although its extraction method has been matured, the thermal instability and preservation difficulties of SFN limit its clinical promotion. More effective and low-cost methods to improve the stability and production of SFN remain to be further studied. This paper reviews the physiological and biological activities of SFN, and summarizes the protective effects and molecular mechanisms of SFN in diseases related to abnormal lipid metabolism. Additionally, it proposes potential challenges, possible solutions and future research directions in the clinical application of SFN.
Collapse
Affiliation(s)
- Lingfeng Zhao
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jiahuan Li
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yiping Dang
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - David Fisher
- Department of Medical Biosciences, Faculty of Natural Sciences, University of the Western Cape, Cape Town, 7100, South Africa
| | | | - Erkin Musabaev
- The Research Institute of Virology, Ministry of Health, 100122, Tashkent, Uzbekistan
| | - Khrystyna Pronyuk
- Infectious Diseases Department, O.Bogomolets National Medical University, Kiev, 02132, Ukraine
| | - Lei Zhao
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
25
|
Zhang K, Gan J, Wang B, Lei W, Zhen D, Yang J, Wang N, Wen C, Gao X, Li X, Xu A, Liu X, Li Y, Wu F, Lin Z. FGF21 protects against HFpEF by improving cardiac mitochondrial bioenergetics in mice. Nat Commun 2025; 16:1661. [PMID: 39955281 PMCID: PMC11829982 DOI: 10.1038/s41467-025-56885-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 02/05/2025] [Indexed: 02/17/2025] Open
Abstract
Fibroblast growth factor 21 (FGF21), a metabolic hormone with pleiotropic effects, is beneficial for various cardiac disorders. However, FGF21's role in heart failure with preserved ejection fraction (HFpEF) remains unclear. Here, we show that elevated circulating FGF21 levels are negatively associated with cardiac diastolic function in patients with HFpEF. Global or adipose FGF21 deficiency exacerbates cardiac diastolic dysfunction and damage in high-fat diet (HFD) plus N[w]-nitro-L-arginine methyl ester (L-NAME)-induced HFpEF mice, whereas these effects are notably reversed by FGF21 replenishment. Mechanistically, FGF21 enhances the production of adiponectin (APN), which in turn indirectly acts on cardiomyocytes, or FGF21 directly targets cardiomyocytes, to negatively regulate pyruvate dehydrogenase kinase 4 (PDK4) production by activating PI3K/AKT signals, then promoting mitochondrial bioenergetics. Additionally, APN deletion strikingly abrogates FGF21's protective effects against HFpEF, while genetic PDK4 inactivation markedly mitigates HFpEF in mice. Thus, FGF21 protects against HFpEF via fine-tuning the multiorgan crosstalk among the adipose, liver, and heart.
Collapse
Affiliation(s)
- Ke Zhang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jing Gan
- The Affiliated Dongguan Songshan Lake Center Hospital, The Innovative Center of Cardiometabolic Disease, Guangdong Medical University, Dongguan, China
| | - Baile Wang
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
| | - Wei Lei
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
- Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Dong Zhen
- The Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jie Yang
- Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Anzhen Hospital of Capital Medical University, Beijing, China
| | - Ningrui Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Congcong Wen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Xiaotang Gao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Xiaokun Li
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
| | - Xinguang Liu
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, Guangdong Medical University, Dongguan, China.
| | - Yulin Li
- Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Anzhen Hospital of Capital Medical University, Beijing, China.
| | - Fan Wu
- The Affiliated Dongguan Songshan Lake Center Hospital, The Innovative Center of Cardiometabolic Disease, Guangdong Medical University, Dongguan, China.
| | - Zhuofeng Lin
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China.
- The Affiliated Dongguan Songshan Lake Center Hospital, The Innovative Center of Cardiometabolic Disease, Guangdong Medical University, Dongguan, China.
| |
Collapse
|
26
|
Geng W, Liao W, Cao X, Yang Y. Therapeutic Targets and Approaches to Manage Inflammation of NAFLD. Biomedicines 2025; 13:393. [PMID: 40002806 PMCID: PMC11853636 DOI: 10.3390/biomedicines13020393] [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: 12/23/2024] [Revised: 01/18/2025] [Accepted: 01/21/2025] [Indexed: 02/27/2025] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) and its advanced form, non-alcoholic steatohepatitis (NASH), are the leading causes of chronic liver disease globally. They are driven by complex mechanisms where inflammation plays a pivotal role in disease progression. Current therapies, including lifestyle changes and pharmacological agents, are limited in efficacy, particularly in addressing the advanced stages of the disease. Emerging approaches targeting inflammation, metabolic dysfunction, and fibrosis offer promising new directions, though challenges such as treatment complexity and heterogeneity persist. This review concludes the main therapeutic targets and approaches to manage inflammation currently and emphasizes the critical need for future drug development and combination therapy for NAFLD/NASH management.
Collapse
Affiliation(s)
- Wanying Geng
- 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China;
- Department of Gastroenterology, Department of Internal Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China; (W.L.); (X.C.)
| | - Wanying Liao
- Department of Gastroenterology, Department of Internal Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China; (W.L.); (X.C.)
| | - Xinyuan Cao
- Department of Gastroenterology, Department of Internal Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China; (W.L.); (X.C.)
| | - Yingyun Yang
- Department of Gastroenterology, Department of Internal Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China; (W.L.); (X.C.)
| |
Collapse
|
27
|
Ashiqueali SA, Hayslip N, Chaudhari DS, Schneider A, Zhu X, Rubis B, Seavey CE, Alam MT, Hussein R, Noureddine SA, Golusinska-Kardach E, Pazdrowski P, Yadav H, Masternak MM. Fecal microbiota transplant from long-living Ames dwarf mice alters the microbial composition and biomarkers of liver health in normal mice. GeroScience 2025:10.1007/s11357-025-01539-3. [PMID: 39904968 DOI: 10.1007/s11357-025-01539-3] [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: 12/09/2024] [Accepted: 01/20/2025] [Indexed: 02/06/2025] Open
Abstract
Aging is associated with intestinal dysbiosis, a condition characterized by diminished microbial biodiversity and inflammation. This leads to increased vulnerability to extraintestinal manifestations such as autoimmune, metabolic, and neurodegenerative conditions thereby accelerating mortality. As such, modulation of the gut microbiome is a promising way to extend healthspan. In this study, we explore the effects of fecal microbiota transplant (FMT) from long-living Ames dwarf donors to their normal littermates, and vice versa, on the recipient gut microbiota and liver transcriptome. Importantly, our previous studies highlight differences between the microbiome of Ames dwarf mice relative to their normal siblings, potentially contributing to their extended lifespan and remarkable healthspan. Our findings demonstrate that FMT from Ames dwarf mice to normal mice significantly alters the recipient's gut microbiota, potentially reprogramming bacterial functions related to healthy aging, and changes the liver transcriptome, indicating improved metabolic health. Particularly, the microbiome of Ames dwarf mice, characterized by a higher abundance of beneficial bacterial families such as Peptococcaceae, Oscillospiraceae, and Lachnospiraceae, appears to play a crucial role in modulating these effects. Alongside, our mRNA sequencing and RT-PCR validation reveals that FMT may contribute to the significant downregulation of p21, Elovl3, and Insig2, genes involved with cellular senescence and liver metabolic pathways. Our data suggest a regulatory axis exists between the gut and liver, highlighting the potential of microbiome-targeted therapies in promoting healthy aging. Future research should focus on functional validation of altered microbial communities and explore the underlying biomolecular pathways that confer geroprotection.
Collapse
Affiliation(s)
- Sarah A Ashiqueali
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA
- Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, Orlando, FL, USA
| | - Natalie Hayslip
- Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, Orlando, FL, USA
- University of South Florida (USF) Morsani College of Medicine, Tampa, FL, USA
| | - Diptaraj S Chaudhari
- Research and Development Service, John D. Dingell VA Medical Center, Detroit, MI, USA
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI, USA
- USF Center for Microbiome Research, Microbiomes Institute, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Augusto Schneider
- Faculdade de Nutrição, Universidade Federal de Pelotas, Pelotas, Brazil
| | - Xiang Zhu
- Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, Orlando, FL, USA
| | - Blazej Rubis
- Department of Clinical Chemistry and Molecular Diagnostics, Poznań University of Medical Sciences, Poznań, Poland
| | - Corey E Seavey
- Enteric Neuroscience Program (ENSP), Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, MN, USA
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Department of Internal Medicine, University of Central Florida College of Medicine, Orlando, FL, USA
| | - Md Tanjim Alam
- Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, Orlando, FL, USA
| | - Ridwan Hussein
- Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, Orlando, FL, USA
- Sidney Kimmel Medical College, Philadelphia, PA, USA
| | - Sarah A Noureddine
- Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, Orlando, FL, USA
| | - Ewelina Golusinska-Kardach
- Department of Dental Surgery, Periodontology and Oral Mucosa Diseases, Poznan University of Medical Sciences, Poznan, Poland
| | - Pawel Pazdrowski
- Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, Orlando, FL, USA
- Poznan University of Medical Sciences, Student Scientific Association, Poznan, Poland
| | - Hariom Yadav
- USF Center for Microbiome Research, Microbiomes Institute, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Michal M Masternak
- Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, Orlando, FL, USA.
- Department of Head and Neck Surgery, Poznan University of Medical Sciences, Poznan, Poland.
| |
Collapse
|
28
|
Lu SL, Pan ZH, Cui Z, Wang JL, Yang JL, Lv YF, Cao CY, Huang XF. AAV2-mediated ABD-FGF21 gene delivery produces a sustained anti-hyperglycemic effect in type 2 diabetic mouse. Life Sci 2025; 362:123344. [PMID: 39736351 DOI: 10.1016/j.lfs.2024.123344] [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: 11/03/2024] [Revised: 12/16/2024] [Accepted: 12/26/2024] [Indexed: 01/01/2025]
Abstract
BACKGROUND Fibroblast Growth Factor 21 (FGF21) is a naturally occurring peptide hormone involved in the regulation of glycolipid metabolism, and it shows promise as a potential treatment for type 2 diabetes mellitus (T2DM). However, the short half-life and poor pharmacokinetics of native FGF21 limit its efficacy in reducing hyperglycemia in vivo. Therefore, maintaining stable and sustained blood concentrations of FGF21 is crucial for its role as an effective regulator of glycolipid metabolism in vivo. In this study, we developed an AAV2-mediated gene delivery system incorporating an Albumin-binding domain (ABD) fused to FGF21, and we evaluated its effects in a type 2 diabetic mouse model. METHODS The plasmids pAAV-FGF21-Luciferase, pHelper, and the capsid plasmid were transfected into HEK293T cells to generate recombinant AAV (rAAV) virus. A type 2 diabetes mellitus (T2DM) mouse model was established for evaluation. The rAAV was administered via tail vein injection into the mice. The effects of rAAV injection on various parameters were assessed using commercial kits. Histological changes in the liver and adipose tissue of T2DM mice were examined using hematoxylin and eosin (H&E) staining. RESULTS The data showed that the inclusion of ABD significantly prolonged the half-life of FGF21 in the serum of mice. Additionally, AAV2-mediated delivery of ABD-FGF21 to the liver resulted in sustained gene expression and a significant increase in circulating FGF21 levels in mice. Treatment with AAV2-ABD-FGF21 led to several benefits, including reduced fasting glucose, improved insulin sensitivity, decreased triglyceride and total cholesterol levels, and improved body weight in T2DM mice. Furthermore, serum analysis and histological examination showed no significant liver damage at the study endpoint after seven weeks. CONCLUSION In conclusion, we have developed a novel strategy for producing long-acting FGF21 using the AAV vector, and AAV2-ABD-FGF21 shows promise as a therapeutic approach for type 2 diabetes mellitus and other glycolipid metabolic disorders.
Collapse
Affiliation(s)
- Sen-Lin Lu
- College of Medicine and Health Sciences, China Three Gorges University, Yichang 443002, China; Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, China
| | - Zhi-Hao Pan
- College of Medicine and Health Sciences, China Three Gorges University, Yichang 443002, China; Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, China
| | - Zhi Cui
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, China; College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, China
| | - Ji-Li Wang
- College of Medicine and Health Sciences, China Three Gorges University, Yichang 443002, China; Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, China
| | - Jian-Lin Yang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, China; College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, China
| | - Ya-Feng Lv
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, China; College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, China.
| | - Chun-Yu Cao
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, China; College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, China.
| | - Xiao-Fei Huang
- College of Medicine and Health Sciences, China Three Gorges University, Yichang 443002, China; Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, China.
| |
Collapse
|
29
|
Carter MM, Zeng X, Ward CP, Landry M, Perelman D, Hennings T, Meng X, Weakley AM, Cabrera AV, Robinson JL, Nguyen T, Higginbottom S, Maecker HT, Sonnenburg ED, Fischbach MA, Gardner CD, Sonnenburg JL. A gut pathobiont regulates circulating glycine and host metabolism in a twin study comparing vegan and omnivorous diets. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.01.08.25320192. [PMID: 39830242 PMCID: PMC11741504 DOI: 10.1101/2025.01.08.25320192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Metabolic diseases including type 2 diabetes and obesity pose a significant global health burden. Plant-based diets, including vegan diets, are linked to favorable metabolic outcomes, yet the underlying mechanisms remain unclear. In a randomized trial involving 21 pairs of identical twins, we investigated the effects of vegan and omnivorous diets on the host metabolome, immune system, and gut microbiome. Vegan diets induced significant shifts in serum and stool metabolomes, cytokine profiles, and gut microbial composition. Despite lower dietary glycine intake, vegan diet subjects exhibited elevated serum glycine levels linked to reduced abundance of the gut pathobiont Bilophila wadsworthia. Functional studies demonstrated that B. wadsworthia metabolizes glycine via the glycine reductase pathway and modulates host glycine availability. Removing B. wadsworthia from a complex microbiota in mice elevated glycine levels and improved metabolic markers. These findings reveal a previously underappreciated mechanism by which diet regulates host metabolic status via the gut microbiota.
Collapse
Affiliation(s)
- Matthew M. Carter
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Xianfeng Zeng
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Catherine P. Ward
- Stanford Prevention Research Center, Department of Medicine, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Matthew Landry
- Department of Population Health and Disease Prevention, Joe C. Wen School of Population & Public Health, University of California, Irvine, Irvine, CA, USA
| | - Dalia Perelman
- Stanford Prevention Research Center, Department of Medicine, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Tayler Hennings
- Stanford Prevention Research Center, Department of Medicine, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Xiandong Meng
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Allison M. Weakley
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Ashley V. Cabrera
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Jennifer L. Robinson
- Stanford Prevention Research Center, Department of Medicine, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Tran Nguyen
- Human Immune Monitoring Center, Institute for Immunity, Transplantation, and Infection, Stanford University, School of Medicine, Stanford, CA, USA
| | - Steven Higginbottom
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Holden T. Maecker
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Human Immune Monitoring Center, Institute for Immunity, Transplantation, and Infection, Stanford University, School of Medicine, Stanford, CA, USA
| | - Erica D. Sonnenburg
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael A. Fischbach
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Christopher D. Gardner
- Stanford Prevention Research Center, Department of Medicine, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Justin L. Sonnenburg
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
- Center for Human Microbiome Studies, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
30
|
Zeng J, Cheong LYT, Lo CH. Therapeutic targeting of obesity-induced neuroinflammation and neurodegeneration. Front Endocrinol (Lausanne) 2025; 15:1456948. [PMID: 39897964 PMCID: PMC11781992 DOI: 10.3389/fendo.2024.1456948] [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: 06/29/2024] [Accepted: 12/30/2024] [Indexed: 02/04/2025] Open
Abstract
Obesity is a major modifiable risk factor leading to neuroinflammation and neurodegeneration. Excessive fat storage in obesity promotes the progressive infiltration of immune cells into adipose tissue, resulting in the release of pro-inflammatory factors such as cytokines and adipokines. These inflammatory mediators circulate through the bloodstream, propagating inflammation both in the periphery and in the central nervous system. Gut dysbiosis, which results in a leaky intestinal barrier, exacerbates inflammation and plays a significant role in linking obesity to the pathogenesis of neuroinflammation and neurodegeneration through the gut-brain/gut-brain-liver axis. Inflammatory states within the brain can lead to insulin resistance, mitochondrial dysfunction, autolysosomal dysfunction, and increased oxidative stress. These disruptions impair normal neuronal function and subsequently lead to cognitive decline and motor deficits, similar to the pathologies observed in major neurodegenerative diseases, including Alzheimer's disease, multiple sclerosis, and Parkinson's disease. Understanding the underlying disease mechanisms is crucial for developing therapeutic strategies to address defects in these inflammatory and metabolic pathways. In this review, we summarize and provide insights into different therapeutic strategies, including methods to alter gut dysbiosis, lifestyle changes, dietary supplementation, as well as pharmacological agents derived from natural sources, that target obesity-induced neuroinflammation and neurodegeneration.
Collapse
Affiliation(s)
- Jialiu Zeng
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, NY, United States
- Interdisciplinary Neuroscience Program, Syracuse University, Syracuse, NY, United States
| | - Lenny Yi Tong Cheong
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Chih Hung Lo
- Interdisciplinary Neuroscience Program, Syracuse University, Syracuse, NY, United States
- Department of Biology, Syracuse University, Syracuse, NY, United States
| |
Collapse
|
31
|
Heinken A, Asara JM, Gnanaguru G, Singh C. Systemic regulation of retinal medium-chain fatty acid oxidation repletes TCA cycle flux in oxygen-induced retinopathy. Commun Biol 2025; 8:25. [PMID: 39789310 PMCID: PMC11718186 DOI: 10.1038/s42003-024-07394-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: 06/17/2024] [Accepted: 12/10/2024] [Indexed: 01/12/2025] Open
Abstract
Activation of anaplerosis takes away glutamine from the biosynthetic pathways to the energy-producing TCA cycle. Especially, induction of hyperoxia driven anaplerosis in neurovascular tissues such as the retina during early stages of development could deplete biosynthetic precursors from newly proliferating endothelial cells impeding physiological angiogenesis and leading to vasoobliteration. Using an oxygen-induced retinopathy (OIR) mouse model, we investigated the metabolic differences between OIR-resistant BALB/cByJ and OIR susceptible C57BL/6J strains at system levels to understand the molecular underpinnings that potentially contribute to hyperoxia-induced vascular abnormalities in the neural retina. Our systems level in vivo RNA-seq, proteomics, and lipidomic profiling and ex-vivo retinal explant studies show that the medium-chain fatty acids serves as an alternative source to feed the TCA cycle. Our findings strongly implicate that medium-chain fatty acids could suppress glutamine-fueled anaplerosis and ameliorate hyperoxia-induced vascular abnormalities in conditions such as retinopathy of prematurity.
Collapse
Affiliation(s)
- Almut Heinken
- Inserm UMRS 1256 NGERE, University of Lorraine, Nancy, France
| | - John M Asara
- Division of Signal Transduction/Mass Spectrometry Core, Beth Israel Deaconess Medical Center, Boston, MA, 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Gopalan Gnanaguru
- Department of Ophthalmology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, 02111, USA
| | - Charandeep Singh
- Department of Ophthalmology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, 02111, USA.
- Division of Biochemical and Molecular Nutrition, Gerald J. and Dorothy R. Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, 02111, USA.
| |
Collapse
|
32
|
Moharamzadeh S, Kashef M, Salehpour M, Torabi M, Vesali S, Samsonchi Z, Hajizadeh-Saffar E. Effects of exercise intensity and diet on cardiac tissue structure and FGF21/β-Klotho signaling in type 2 diabetic mice: a comparative study of HFD and HFD + STZ induced type 2 diabetes models in mice. Diabetol Metab Syndr 2025; 17:4. [PMID: 39757236 DOI: 10.1186/s13098-024-01541-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 11/27/2024] [Indexed: 01/07/2025] Open
Abstract
BACKGROUND Structural heart disease is one of the leading causes of death in people with type 2 diabetes (T2D), which is not known to have an effect on exercise training. The aim of this study was to compare the effects of high-intensity interval training (HIIT) and moderate-intensity continuous training (MICT) on heart tissue structure, the serum level of FGF21 and the heart tissue level of β-Klotho, an FGF21 coreceptor, in HFD and HFD + STZ-induced diabetic mice. METHODS Thirty-six male C57BL/6J mice were divided into high-fat diet (HFD) and normal chow diet (ND) groups. After 20 weeks of diet, the HFD mice were divided into HFD and HFD + STZ groups, and the latter group was injected with STZ. Then, the mice in the ND, HFD and HFD + STZ groups were divided into three subgroups of control (C), HIIT and MICT, and mice were placed in one of nine groups ND-C, ND-HIIT, ND-MICT, HFD-C, HFD-HIIT, HFD-MICT, HFD + STZ-C, HFD + STZ-HIIT, and HFD + STZ-MICT. The mice in the exercise training (ET) groups were run on a treadmill for eight weeks. Finally, the tissue and serum samples were collected and analyzed by two-way ANOVA. RESULTS Statistical analyses showed that the main effect of diabetes inducing model (DIM) was significant for all variables (p < 0.05), except vascular density (p = 0.055); the main effect of ET type on fasting blood glucose and FGF21 was significant (p < 0.001); and the interaction was significant for fasting blood glucose, heart weight and FGF21 (p < 0.001). Post hoc and subgroup analysis showed a superior effect of MICT over HIIT in decreasing fasting blood glucose and serum level of FGF21 (p < 0.001). Additionally, the results of the myocardial tissue qualitative analyses differed between the diabetic mouse models and the ET groups. CONCLUSIONS In a mouse model, type 2 diabetes can negatively affect heart tissue structure and FGF21 signaling in cardiac tissue, and both HIIT and MICT can prevent this effect. However, MICT likely more effective that HIIT in reducing circulating FGF21.
Collapse
Affiliation(s)
- Sevda Moharamzadeh
- Department of Exercise Physiology, Faculty of Sport Science, Shahid Rajaei Teacher Training University, Tehran, Iran
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Basic and Population-Based Studies in NCD, Reproductive Epidemiology Research Center, Royan Institute, ACECR, Tehran, Iran
| | - Majid Kashef
- Department of Exercise Physiology, Faculty of Sport Science, Shahid Rajaei Teacher Training University, Tehran, Iran.
| | - Mojtaba Salehpour
- Department of Exercise Physiology, Faculty of Sport Science, Shahid Rajaei Teacher Training University, Tehran, Iran
| | - Meysam Torabi
- Department of Exercise Physiology, Faculty of Sport Science, Guilan University, Rasht, Iran
- Department of Basic and Population-Based Studies in NCD, Reproductive Epidemiology Research Center, Royan Institute, ACECR, Tehran, Iran
| | - Samira Vesali
- Reproductive Epidemiology Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Zakieh Samsonchi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Advanced Therapy Medicinal Product Technology Development Center, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Ensiyeh Hajizadeh-Saffar
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
- Advanced Therapy Medicinal Product Technology Development Center, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
33
|
Jain U, Srivastava P, Sharma A, Sinha S, Johari S. Impaired Fibroblast Growth Factor 21 (FGF21) Associated with Visceral Adiposity Leads to Insulin Resistance: The Core Defect in Diabetes Mellitus. Curr Diabetes Rev 2025; 21:e260424229342. [PMID: 38676505 DOI: 10.2174/0115733998265915231116043813] [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: 06/23/2023] [Revised: 09/06/2023] [Accepted: 09/20/2023] [Indexed: 04/29/2024]
Abstract
The Central nervous system (CNS) is the prime regulator of signaling pathways whose function includes regulation of food intake (consumption), energy expenditure, and other metabolic responses like glycolysis, gluconeogenesis, fatty acid oxidation, and thermogenesis that have been implicated in chronic inflammatory disorders. Type 2 diabetes mellitus (T2DM) and obesity are two metabolic disorders that are linked together and have become an epidemic worldwide, thus raising significant public health concerns. Fibroblast growth factor 21 (FGF21) is an endocrine hormone with pleiotropic metabolic effects that increase insulin sensitivity and energy expenditure by elevating thermogenesis in brown or beige adipocytes, thus reducing body weight and sugar intake. In contrast, during starvation conditions, FGF21 induces its expression in the liver to initiate glucose homeostasis. Insulin resistance is one of the main anomalies caused by impaired FGF21 signaling, which also causes abnormal regulation of other signaling pathways. Tumor necrosis factor alpha (TNF-α), the cytokine released by adipocytes and inflammatory cells in response to chronic inflammation, is regarded major factor that reduces the expression of FGF21 and modulates underlying insulin resistance that causes imbalanced glucose homeostasis. This review aims to shed light on the mechanisms underlying the development of insulin resistance in obese individuals as well as the fundamental flaw in type 2 diabetes, which is malfunctioning obese adipose tissue.
Collapse
Affiliation(s)
- Unnati Jain
- Department of Biosciences, Institute of Management Studies Ghaziabad (University Courses Campus), NH09, Adhyatmik Nagar, Ghaziabad, Uttar Pradesh, India
| | - Priyanka Srivastava
- Department of Biosciences, Institute of Management Studies Ghaziabad (University Courses Campus), NH09, Adhyatmik Nagar, Ghaziabad, Uttar Pradesh, India
| | - Ashwani Sharma
- Insight BioSolutions, Rue Joseph Colin, 35000 Rennes, France
| | - Subrata Sinha
- Centre of Biotechnology and Bioinformatics, Dibrugarh University, Dibrugarh, Assam 786004, India
| | - Surabhi Johari
- Department of Biosciences, Institute of Management Studies Ghaziabad (University Courses Campus), NH09, Adhyatmik Nagar, Ghaziabad, Uttar Pradesh, India
| |
Collapse
|
34
|
Xu W, Gao X, Luo H, Chen Y. FGF21 attenuates salt-sensitive hypertension via regulating HNF4α/ACE2 axis in the hypothalamic paraventricular nucleus of mice. Clin Exp Hypertens 2024; 46:2361671. [PMID: 38841901 DOI: 10.1080/10641963.2024.2361671] [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: 05/02/2024] [Accepted: 05/24/2024] [Indexed: 06/07/2024]
Abstract
BACKGROUND Fibroblast growth factor 21 (FGF21) has a protective effect against cardiovascular disease. However, the role of FGF21 in hypertension remains elusive. METHODS Ten-week-old male C57BL/6 mice were randomly divided into normal-salt (NS) group, NS+FGF21 group, deoxycorticosterone acetate-salt (DOCA) group and DOCA+FGF21 group. The mice in NS group underwent uninephrectomy without receiving DOCA and 1% NaCl and the mice in DOCA group were subjected to uninephrectomy and DOCA-salt (DOCA and 1% NaCl) treatment for 6 weeks. At the same time, the mice were infused with vehicle (artificial cerebrospinal fluid, aCSF) or FGF21 (1 mg/kg) into the bilateral paraventricular nucleus (PVN) of mice. RESULTS Here, we showed that FGF21 treatment lowered DOCA salt-induced inflammation and oxidative stress in the PVN, which reduced sympathetic nerve activity and hypertension. Mechanistically, FGF21 treatment decreased the expression of HNF4α and inhibited the binding activity of HNF4α to the promoter region of ACE2 in the PVN of DOCA salt-treated mice, which further up-regulated ACE2/Ang (1-7) signals in the PVN. In addition, ACE2 deficiency abolished the protective effect of FGF21 in DOCA salt-treated mice, suggesting that FGF21-mediated antihypertensive effect was dependent on ACE2. CONCLUSIONS The results demonstrate that FGF21 protects against salt-sensitive hypertension via regulating HNF4α/ACE2/Ang (1-7) axis in the PVN of DOCA salt-treated mice via multi-organ crosstalk between liver, brain and blood vessels.
Collapse
Affiliation(s)
- Wei Xu
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xia Gao
- Department of Geriatrics, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Hao Luo
- Department of Cardiology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yingmei Chen
- Department of Cardiology, General Hospital of Western Theater Command, Chengdu, Sichuan, China
| |
Collapse
|
35
|
Liu S, Hua L, Mo X, Lei B, Zhang R, Zhou S, Jiang X, Fang Z, Feng B, Che L, Xu S, Lin Y, Wu D, Zhuo Y, Jin C. Comparative Impact of Alternate-Day Fasting and Time-Restricted Feeding on Placental Function and Fetal Development in Maternal Obesity. Nutrients 2024; 17:25. [PMID: 39796458 PMCID: PMC11723168 DOI: 10.3390/nu17010025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 12/13/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND Maternal obesity detrimentally affects placental function and fetal development. Both alternate-day fasting (ADF) and time-restricted feeding (TRF) are dietary interventions that can improve metabolic health, yet their comparative effects on placental function and fetal development remain unexplored. OBJECTIVES This study aims to investigate the effects of ADF and TRF on placental function and fetal development during maternal consumption of a high-fat diet (HFD). METHODS One hundred 8-week-old female mice were assigned to one of four dietary regimens: (1) normal diet with ad libitum feeding (NA); (2) HFD with ad libitum feeding (HA); (3) HFD with ADF (HI); and (4) HFD with TRF (HT), administered both before and during pregnancy. On gestational day 18.5, serum and placental samples were collected from both mothers and fetuses to examine placental function and fetal development. RESULTS During gestation, both ADF and TRF substantially alleviated the metabolic impairments caused by an HFD in obese maternal mice. TRF mice demonstrated enhanced placental nutrient transport and fetal development, associated with reduced endoplasmic reticulum (ER) stress and inflammatory responses. In contrast, ADF markedly intensified placental stress and inflammatory responses, diminished placental nutrient transport efficiency, and consequently induced fetal growth restriction. CONCLUSIONS Both ADF and TRF during pregnancy significantly mitigated metabolic impairments in obese dams on an HFD. TRF mice demonstrated enhanced placental nutrient transport and fetal development, associated with reduced endoplasmic reticulum (ER) stress and inflammatory responses. In contrast, ADF markedly intensified placental stress and inflammatory responses, diminished placental nutrient transport efficiency, and consequently induced fetal growth restriction.
Collapse
Affiliation(s)
- Siyuan Liu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (S.L.); (L.H.); (X.M.); (B.L.); (R.Z.); (S.Z.); (X.J.); (Z.F.); (B.F.); (L.C.); (S.X.); (Y.L.); (D.W.); (Y.Z.)
- Key Laboratory for Animal Disease-Resistant Nutrition of Sichuan Province, The Ministry of Education of China, Chengdu 611130, China
| | - Lun Hua
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (S.L.); (L.H.); (X.M.); (B.L.); (R.Z.); (S.Z.); (X.J.); (Z.F.); (B.F.); (L.C.); (S.X.); (Y.L.); (D.W.); (Y.Z.)
- Key Laboratory for Animal Disease-Resistant Nutrition of Sichuan Province, The Ministry of Education of China, Chengdu 611130, China
| | - Xi Mo
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (S.L.); (L.H.); (X.M.); (B.L.); (R.Z.); (S.Z.); (X.J.); (Z.F.); (B.F.); (L.C.); (S.X.); (Y.L.); (D.W.); (Y.Z.)
- Key Laboratory for Animal Disease-Resistant Nutrition of Sichuan Province, The Ministry of Education of China, Chengdu 611130, China
| | - Bing Lei
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (S.L.); (L.H.); (X.M.); (B.L.); (R.Z.); (S.Z.); (X.J.); (Z.F.); (B.F.); (L.C.); (S.X.); (Y.L.); (D.W.); (Y.Z.)
- Key Laboratory for Animal Disease-Resistant Nutrition of Sichuan Province, The Ministry of Education of China, Chengdu 611130, China
| | - Ruihao Zhang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (S.L.); (L.H.); (X.M.); (B.L.); (R.Z.); (S.Z.); (X.J.); (Z.F.); (B.F.); (L.C.); (S.X.); (Y.L.); (D.W.); (Y.Z.)
- Key Laboratory for Animal Disease-Resistant Nutrition of Sichuan Province, The Ministry of Education of China, Chengdu 611130, China
| | - Shihao Zhou
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (S.L.); (L.H.); (X.M.); (B.L.); (R.Z.); (S.Z.); (X.J.); (Z.F.); (B.F.); (L.C.); (S.X.); (Y.L.); (D.W.); (Y.Z.)
- Key Laboratory for Animal Disease-Resistant Nutrition of Sichuan Province, The Ministry of Education of China, Chengdu 611130, China
| | - Xuemei Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (S.L.); (L.H.); (X.M.); (B.L.); (R.Z.); (S.Z.); (X.J.); (Z.F.); (B.F.); (L.C.); (S.X.); (Y.L.); (D.W.); (Y.Z.)
- Key Laboratory for Animal Disease-Resistant Nutrition of Sichuan Province, The Ministry of Education of China, Chengdu 611130, China
| | - Zhengfeng Fang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (S.L.); (L.H.); (X.M.); (B.L.); (R.Z.); (S.Z.); (X.J.); (Z.F.); (B.F.); (L.C.); (S.X.); (Y.L.); (D.W.); (Y.Z.)
- Key Laboratory for Animal Disease-Resistant Nutrition of Sichuan Province, The Ministry of Education of China, Chengdu 611130, China
| | - Bin Feng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (S.L.); (L.H.); (X.M.); (B.L.); (R.Z.); (S.Z.); (X.J.); (Z.F.); (B.F.); (L.C.); (S.X.); (Y.L.); (D.W.); (Y.Z.)
- Key Laboratory for Animal Disease-Resistant Nutrition of Sichuan Province, The Ministry of Education of China, Chengdu 611130, China
| | - Lianqiang Che
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (S.L.); (L.H.); (X.M.); (B.L.); (R.Z.); (S.Z.); (X.J.); (Z.F.); (B.F.); (L.C.); (S.X.); (Y.L.); (D.W.); (Y.Z.)
- Key Laboratory for Animal Disease-Resistant Nutrition of Sichuan Province, The Ministry of Education of China, Chengdu 611130, China
| | - Shengyu Xu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (S.L.); (L.H.); (X.M.); (B.L.); (R.Z.); (S.Z.); (X.J.); (Z.F.); (B.F.); (L.C.); (S.X.); (Y.L.); (D.W.); (Y.Z.)
- Key Laboratory for Animal Disease-Resistant Nutrition of Sichuan Province, The Ministry of Education of China, Chengdu 611130, China
| | - Yan Lin
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (S.L.); (L.H.); (X.M.); (B.L.); (R.Z.); (S.Z.); (X.J.); (Z.F.); (B.F.); (L.C.); (S.X.); (Y.L.); (D.W.); (Y.Z.)
- Key Laboratory for Animal Disease-Resistant Nutrition of Sichuan Province, The Ministry of Education of China, Chengdu 611130, China
| | - De Wu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (S.L.); (L.H.); (X.M.); (B.L.); (R.Z.); (S.Z.); (X.J.); (Z.F.); (B.F.); (L.C.); (S.X.); (Y.L.); (D.W.); (Y.Z.)
- Key Laboratory for Animal Disease-Resistant Nutrition of Sichuan Province, The Ministry of Education of China, Chengdu 611130, China
| | - Yong Zhuo
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (S.L.); (L.H.); (X.M.); (B.L.); (R.Z.); (S.Z.); (X.J.); (Z.F.); (B.F.); (L.C.); (S.X.); (Y.L.); (D.W.); (Y.Z.)
- Key Laboratory for Animal Disease-Resistant Nutrition of Sichuan Province, The Ministry of Education of China, Chengdu 611130, China
| | - Chao Jin
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (S.L.); (L.H.); (X.M.); (B.L.); (R.Z.); (S.Z.); (X.J.); (Z.F.); (B.F.); (L.C.); (S.X.); (Y.L.); (D.W.); (Y.Z.)
- Key Laboratory for Animal Disease-Resistant Nutrition of Sichuan Province, The Ministry of Education of China, Chengdu 611130, China
| |
Collapse
|
36
|
Ye X, Pei F, Li W, Xue J, Huang X, Huang J, Zhang L. Fibroblast growth factor 21 attenuates pulmonary ischemia/reperfusion injury via inhibiting endoplasmic reticulum stress-induced ferroptosis though FGFR1/PPARδ signaling pathway. Int Immunopharmacol 2024; 143:113307. [PMID: 39366074 DOI: 10.1016/j.intimp.2024.113307] [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: 06/06/2024] [Revised: 09/28/2024] [Accepted: 09/29/2024] [Indexed: 10/06/2024]
Abstract
BACKGROUND Acute lung injury is a critical life-threatening complication of pulmonary and cardiac surgery with a high rate of morbidity and mortality. Fibroblast growth factor 21 (FGF21) has been reported to play an important role in protecting vital organs from damage. This study aims to investigate the potential protective role and mechanism of FGF21 in pulmonary ischemia/reperfusion (I/R)-induced acute lung injury. METHODS A pulmonary epithelial cell line was treated with hypoxia/regeneration (H/R) in vitro and a mouse model of acute lung injury was induced with pulmonary I/R in vivo. Lung injury after pulmonary I/R was compared between FGF21-konckout (KO) mice and wild-type (WT) mice. Recombinant FGF21 was administrated in vivo and in vitro to determine its therapeutic effect. RESULTS Circulating levels of FGF21 in mice with pulmonary I/R injury were significantly higher than in those without pulmonary I/R injury. Lung injury was aggravated in FGF21-KO mice compared with WT mice and the administration of FGF21 alleviated lung injury in mouse treated with I/R and pulmonary epithelial cell injury treated with H/R. FGF21 treatment decreased endoplasmic reticulum (ER) stress, Fe2+ and lipid reactive oxygen species (ROS) contents and GPX4 expression and increased PTGS2 levels. Mechanistically, FGF21 upregulated the expression of FGFR1 and PPARδ, ameliorated ER stress and ER stress induced-ferroptosis. Furthermore, FGF21 increased the expression level of PPARδ in pulmonary epithelial cell exposed to H/R, which was inhibited by FGFR1 inhibitor (PD173074). The protective effects of FGF21 were abolished by co-treatment with PPARδ inhibitor (GSK0660), indicating FGF21 attenuated ER stress-induced ferroptosis by dependent on FGFR1/PPARδ signaling pathway. CONCLUSION Our study reveals that FGF21 protects against pulmonary I/R injury via inhibiting ER stress-induced ferroptosis though FGFR1/PPARδ signaling pathway. Boosting endogenous FGF21 or the administration of recombinant FGF21 could be promising therapeutic strategies for pulmonary IRI.
Collapse
Affiliation(s)
- Xinqiao Ye
- Department of Thoracic Surgery, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou 341009, Jiangxi, China
| | - Fang Pei
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou 341009, Jiangxi, China.
| | - Wei Li
- People's Hospital of Ningdu County, Ganzhou 342899, Jiangxi, China
| | - Jinping Xue
- Xinfeng Hospital of Traditional Chinese Midicine, Ganzhou 341699, Jiangxi, China
| | - Xiuyun Huang
- Dingnan Second Hospital, Ganzhou 341999, Jiangxi, China
| | - Jianming Huang
- Department of Thoracic Surgery, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou 341009, Jiangxi, China
| | - Liyan Zhang
- Department of Clinical Laboratory, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 519041, Guangdong, China
| |
Collapse
|
37
|
Basir H, Nugrahani ASD, Aman AM, Bakri S, Rasyid H, Umar H, H. P. F, Ichsan AM, Zainuddin AA. The association between fibroblast growth factor 21 with diabetes retinopathy among type 2 diabetes mellitus patients: a systematic review, meta-analysis, and meta-regression. PeerJ 2024; 12:e18308. [PMID: 39687000 PMCID: PMC11648683 DOI: 10.7717/peerj.18308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 09/24/2024] [Indexed: 12/18/2024] Open
Abstract
Background Diabetic retinopathy (DR), a leading cause of vision loss worldwide, is a common complication of type 2 diabetes mellitus (T2DM) driven by chronic hyperglycemia and microvascular damage. Fibroblast growth factor 21 (FGF21) is crucial in blood sugar regulation and has been linked to DR incidence and severity. While some studies suggest that FGF21 levels may contribute to the DR incidence, others propose a protective role. This discrepancy necessitates further analysis, prompting this study to evaluate the association between FGF21 levels and DR incidence and severity in T2DM patients. Methods A systematic search was conducted through MEDLINE, Web of Science, Scopus, and Embase up to May 2024 for studies evaluating the association between FGF21 and DR incidence and severity. A random-effect model meta-analysis was performed to calculate the pooled standardized mean difference (SMD) and 95% confidence intervals (CI). A univariate meta-regression was performed to analyze factors influencing pooled size estimates. All statistical analyses were performed using STATA 17 software. Result This systematic review and meta-analysis of 5,852 participants revealed that FGF21 was positively correlated with DR (SMD 3.11; 95% CI [0.92-5.30], p = 0.005) and sight-threatening DR (STDR) incidence (SMD 3.61; 95% CI [0.82-6.41], p = 0.01). There was no significant difference in FGF21 levels in DR vs STDR (p = 0.79). Subgroup analysis revealed a significant difference in DR incidence between LDL groups, with higher DR incidence in the group with low-density lipoprotein (LDL) levels >100 (P < 0.00001). Meta-regression revealed no variables significantly influenced the pooled size estimates. Conclusion A higher level of FGF21 was associated with higher DR and STDR incidence among T2DM patients, highlighting its potential utilization as a biomarker for DR detection and enabling the exploration of FGF21-based treatment strategies. However, variables independently predicting DR among patients with elevated FGF21 levels shall be explored further. PROSPERO ID CRD42024559142.
Collapse
Affiliation(s)
- Herni Basir
- Endocrinology and Metabolism Division, Department of Internal Medicine, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | | | - Andi Makbul Aman
- Endocrinology and Metabolism Division, Department of Internal Medicine, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Syakib Bakri
- Department of Internal Medicine, Faculty of Medicine Hasanuddin University, Makassar, Indonesia
| | - Haerani Rasyid
- Department of Internal Medicine, Faculty of Medicine Hasanuddin University, Makassar, Indonesia
| | - Husaini Umar
- Endocrinology and Metabolism Division, Department of Internal Medicine, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Faridin H. P.
- Department of Internal Medicine, Faculty of Medicine Hasanuddin University, Makassar, Indonesia
| | - Andi Muhammad Ichsan
- Department of Ophthalmology, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Andi Alfian Zainuddin
- Department of Public Health, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| |
Collapse
|
38
|
Mishra A, Sobha D, Patel D, Suresh PS. Intermittent fasting in health and disease. Arch Physiol Biochem 2024; 130:755-767. [PMID: 37828854 DOI: 10.1080/13813455.2023.2268301] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 09/29/2023] [Indexed: 10/14/2023]
Abstract
CONTEXT Intermittent fasting, a new-age dietary concept derived from an age-old tradition, involves repetitive cycles of fasting/calorie restriction and eating. OBJECTIVE We aim to take a deep dive into the biological responses to intermittent fasting, delineate the disease-modifying and cognitive effects of intermittent fasting, and also shed light on the possible side effects. METHODS Numerous in vitro and in vivo studies were reviewed, followed by an in-depth analysis, and compilation of their implications in health and disease. RESULTS Intermittent fasting improves the body's stress tolerance, which is further amplified with exercise. It impacts various pathological conditions like cancer, obesity, diabetes, cardiovascular disease, and neurodegenerative diseases. CONCLUSION During dietary restriction, the human body experiences a metabolic switch due to the depletion of liver glycogen, which promotes a shift towards utilising fatty acids and ketones in the system, thereby significantly impacting adiposity, ageing and the immune response to various diseases.
Collapse
Affiliation(s)
- Anubhav Mishra
- School of Biotechnology, National Institute of Technology, Calicut, Calicut, India
| | - Devika Sobha
- School of Biotechnology, National Institute of Technology, Calicut, Calicut, India
| | - Dimple Patel
- School of Biotechnology, National Institute of Technology, Calicut, Calicut, India
| | - Padmanaban S Suresh
- School of Biotechnology, National Institute of Technology, Calicut, Calicut, India
| |
Collapse
|
39
|
Nguyen HDT, Le TM, Jung DR, Jo Y, Choi Y, Lee D, Lee OE, Cho J, Park NJY, Seo I, Chong GO, Shin JH, Han HS. Transcriptomic analysis reveals Streptococcus agalactiae activation of oncogenic pathways in cervical adenocarcinoma. Oncol Lett 2024; 28:588. [PMID: 39411203 PMCID: PMC11474141 DOI: 10.3892/ol.2024.14720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 09/06/2024] [Indexed: 10/19/2024] Open
Abstract
Cervical adenocarcinoma (AC), a subtype of uterine cervical cancer (CC), poses a challenge due to its resistance to therapy and poor prognosis compared with squamous cervical carcinoma. Streptococcus agalactiae [group B Streptococcus (GBS)], a Gram-positive coccus, has been associated with cervical intraepithelial neoplasia in CC. However, the underlying mechanism interaction between GBS and CC, particularly AC, remains elusive. Leveraging The Cancer Genome Atlas public data and time-series transcriptomic data, the present study investigated the interaction between GBS and AC, revealing activation of two pivotal pathways: 'MAPK signaling pathway' and 'mTORC1 signaling'. Western blotting, reverse transcription-quantitative PCR and cell viability assays were performed to validate the activation of these pathways and their role in promoting cancer cell proliferation. Subsequently, the present study evaluated the efficacy of two anticancer drugs targeting these pathways (binimetinib and ridaforolimus) in AC cell treatment. Binimetinib demonstrated a cytostatic effect, while ridaforolimus had a modest impact on HeLa cells after 48 h of treatment, as observed in both cell viability and cytotoxicity assays. The combination of binimetinib and ridaforolimus resulted in a significantly greater cytotoxic effect compared to binimetinib or ridaforolimus monotherapy, although the synergy score indicated an additive effect. In general, the MAPK and mTORC1 signaling pathways were identified as the main pathways associated with GBS and AC cells. The combination of binimetinib and ridaforolimus could be a potential AC treatment.
Collapse
Affiliation(s)
- Hong Duc Thi Nguyen
- Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Tan Minh Le
- Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Da-Ryung Jung
- Department of Applied Biosciences, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Youngjae Jo
- Department of Applied Biosciences, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Yeseul Choi
- Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Donghyeon Lee
- Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Olive Em Lee
- Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Junghwan Cho
- Clinical Omics Institute, Kyungpook National University, Daegu 41405, Republic of Korea
| | - Nora Jee-Young Park
- Clinical Omics Institute, Kyungpook National University, Daegu 41405, Republic of Korea
- Department of Pathology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
- Department of Pathology, Kyungpook National University Chilgok Hospital, Daegu 41404, Republic of Korea
| | - Incheol Seo
- Department of Immunology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Gun Oh Chong
- Clinical Omics Institute, Kyungpook National University, Daegu 41405, Republic of Korea
- Department of Obstetrics and Gynecology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
- Department of Obstetrics and Gynecology, Kyungpook National University Chilgok Hospital, Daegu 41404, Republic of Korea
| | - Jae-Ho Shin
- Department of Applied Biosciences, Kyungpook National University, Daegu 41566, Republic of Korea
- Department of Integrative Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea
- Next Generation Sequencing Core Facility, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Hyung Soo Han
- Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu 41944, Republic of Korea
- Clinical Omics Institute, Kyungpook National University, Daegu 41405, Republic of Korea
- Department of Physiology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| |
Collapse
|
40
|
Petrov ME, Zuraikat FM, Cheng B, Aggarwal B, Jelic S, Laferrère B, St-Onge MP. Impact of sleep restriction on biomarkers of thyroid function: Two pooled randomized trials. Sleep Med 2024; 124:606-612. [PMID: 39488926 DOI: 10.1016/j.sleep.2024.10.035] [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: 08/13/2024] [Accepted: 10/27/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND Chronic, mildly insufficient sleep is associated with increased cardiometabolic risk, but whether the regulation of thyroid hormones and related growth factors are mechanisms of this association is unclear. We investigated whether 6 wk of mild sleep restriction (SR) alters levels of free thyroxine (FT4), thyroid stimulating hormone (TSH), and fibroblast growth factor-21 (FGF-21), a modulator of FT4, in adults with adequate habitual sleep (AS; 7-9 h/night). METHODS Healthy adults participated in one of two randomized, crossover studies with identical 6-wk intervention phases: AS and SR (1.5 h/night < AS). Fasted blood samples were collected at baseline and endpoint of each phase. Outcomes were concentrations of FT4, TSH, and FGF-21 (women only). Linear mixed models tested the effects of SR vs AS on the outcomes, adjusting for baseline levels, week, sex, and sex-by-condition interaction. RESULTS Thirty participants (20 women; 73% racial/ethnic minority; age 21-64 y [M±SD = 36.2 ± 12.8 y]) were included. In the full sample, no effects of SR on FT4 (β±SE = 0.02 ± 0.04, p = 0.654) or TSH (β±SE = -0.02 ± 0.04, p = 0.650) were observed; however, there were sex-by-condition interactions for both FT4 (p-interaction = 0.056) and TSH (p-interaction = 0.049). In sex-stratified analyses, TSH was reduced in SR vs. AS in women (β±SE = -0.11 ± 0.04, p = 0.011, Cohen's f2 = 0.55) but not men (β±SE = 0.09 ± 0.08, p = 0.261). Among women (n = 17), FGF-21 was not significantly different between conditions (β±SE = 8.51 ± 17.70, p = 0.638). CONCLUSION Prolonged mild SR reduces TSH in women, whereas FT4 and FGF-21 remain unaffected compared with AS. If sustained, disruptions to the thyrotropic axis in women may contribute to their more pronounced cardiometabolic risk in response to SR compared with men.
Collapse
Affiliation(s)
- Megan E Petrov
- Edson College of Nursing and Health Innovation, Arizona State University, Phoenix, AZ, USA.
| | - Faris M Zuraikat
- Center of Excellence for Sleep & Circadian Research, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA; Division of General Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA; Institute of Human Nutrition, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA.
| | - Bin Cheng
- Department of Biostatistics, Mailman School of Public Health, Columbia University Irving Medical Center, New York, NY, USA.
| | - Brooke Aggarwal
- Center of Excellence for Sleep & Circadian Research, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA; Institute of Human Nutrition, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA; Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA.
| | - Sanja Jelic
- Center of Excellence for Sleep & Circadian Research, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA; Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Columbia University Irving Medical Center, New York City, NY, USA.
| | - Blandine Laferrère
- Center of Excellence for Sleep & Circadian Research, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA; Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Columbia University Irving Medical Center, New York City, NY, USA; Division of Endocrinology, Department of Medicine, Columbia University Irving Medical Center, New York City, NY, USA.
| | - Marie-Pierre St-Onge
- Center of Excellence for Sleep & Circadian Research, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA; Division of General Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
41
|
Lu FQ, Feng HM, Wang JG, Song KL. Causal relationships between immune cells, inflammatory cytokines, and pertussis: Bidirectional 2-sample Mendelian randomization study and mediation analysis. Medicine (Baltimore) 2024; 103:e40712. [PMID: 39612418 PMCID: PMC11608674 DOI: 10.1097/md.0000000000040712] [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: 09/13/2024] [Accepted: 11/08/2024] [Indexed: 12/01/2024] Open
Abstract
Studies have shown that immune cells play an important role in the occurrence and development of pertussis, but the specific causal relationships are yet to be determined. Additionally, inflammatory cytokines, as regulators of immune responses, may mediate the relationship between immune cells and pertussis, and the specific mechanisms involved require further exploration. This study utilizes data from multiple large-scale genome-wide association studies, covering 731 types of immune cells and 91 types of inflammatory cytokines. The bidirectional 2-sample Mendelian randomization (MR) method is employed, with inverse-variance weighted as the main statistical approach, to assess the causal relationships between immune cells, inflammatory cytokines, and pertussis. Furthermore, a 2-step MR method is used to investigate the mediating role of inflammatory cytokines in the effect of immune cells on pertussis. Our study results indicate that 11 types of immune cells have a protective effect against pertussis, with the strongest protection observed from CD25 on CD28+ CD4+ cells (OR = 0.3533, CI = 0.1636-0.7627, P = .008). Conversely, 19 types of immune cells are positively associated with the risk of pertussis, with the strongest correlation found in CD3- lymphocyte %lymphocyte (OR = 3.6613, CI = 1.5012-8.299, P = .0043). Additionally, 3 inflammatory cytokines - IL-4, IL-18R1, and FGF-21 - show a causal relationship with pertussis. Our mediation MR results indicate that inflammatory cytokines do not act as mediators in the relationship between immune cells and pertussis. This study suggests a causal relationship between immune cells and pertussis, while inflammatory cytokines do not appear to be mediating factors in the pathway from immune cells to pertussis.
Collapse
Affiliation(s)
- Fu-Qing Lu
- Department of Pediatrics, The People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Hui-Mei Feng
- Department of Pediatrics, The People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Ji-Gan Wang
- Department of Pediatrics, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Guangxi Clinical Research Center for Pediatric Diseases, Nanning, China
| | - Kun-Ling Song
- Department of Pediatrics, The People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| |
Collapse
|
42
|
Wei X, Lu Y, Hong S. Gut Microbiota Modulates Fgf21 Expression and Metabolic Phenotypes Induced by Ketogenic Diet. Nutrients 2024; 16:4028. [PMID: 39683422 DOI: 10.3390/nu16234028] [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: 10/22/2024] [Revised: 11/15/2024] [Accepted: 11/18/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND The ketogenic diet (KD) is a widely used intervention for obesity and diabetes, effectively reducing body weight and blood glucose levels. However, the molecular mechanisms by which the KD influences body weight and glucose metabolism are not fully understood. While previous research has shown that the KD affects the gut microbiota, the exact role of microbiota in mediating its metabolic effects remains unclear. METHODS In this study, we used antibiotics to eliminate the gut microbiota, confirming its necessity for the KD's impact on weight loss and glucose metabolism. We also demonstrated the significant role of FGF21 in these processes, through antibiotics intervention in Fgf21-deficient mice. RESULTS Furthermore, we revealed that the KD alters serum valine levels via the gut microbiota, which in turn regulates hepatic Fgf21 expression and circulating FGF21 levels through the GCN2-eIF2α-ATF5 signaling pathway. Additionally, we demonstrated that valine supplementation inhibits the elevated expression of FGF21, leading to the reduced body weight and improved glucose metabolism of the KD-fed mice. Overall, we found that the gut microbiota from the KD regulates Fgf21 transcription via the GCN2-eIF2α-ATF5 signaling pathway. ultimately affecting body weight and glucose metabolism. CONCLUSION Our findings highlight a complex regulatory network linking the KD, Fgf21 expression, and gut microbiota, offering a theoretical foundation for targeted therapies to enhance the metabolic benefits of the KD.
Collapse
Affiliation(s)
- Xinyi Wei
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Yunxu Lu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Shangyu Hong
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, China
| |
Collapse
|
43
|
Bazhan N, Kazantseva A, Dubinina A, Balybina N, Jakovleva T, Makarova E. Age of Cafeteria Diet Onset Influences Obesity Phenotype in Mice in a Sex-Specific Manner. Int J Mol Sci 2024; 25:12436. [PMID: 39596499 PMCID: PMC11595127 DOI: 10.3390/ijms252212436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 11/15/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
We investigated the influence of sex and the age of obesogenic diet initiation on the obesity phenotypes at a later age. C57Bl mice started the Cafeteria Diet (CafD, with increased fat and carbohydrates, ad libitum, from 7 weeks of age (7CafD, pre-puberty) or 17 weeks of age (7CafD, post-puberty) while control C57Bl mice were fed regular chow. At 27 weeks of age, 7CafD males (n = 9) compared to 17CafD males (n = 7) had lower body weight, white adipose tissue (WAT) relative weight, and plasma cholesterol levels, and a higher expression of thermogenic genes in WAT and brown adipose tissue (BAT), and fatty acid oxidation (FAO) and insulin signalling genes in muscles. The 7CafD females (n = 8), compared to 17CafD females (n = 6), had higher plasma triglyceride levels and hepatic glycogen content, but lower insulin sensitivity and hepatic expression of FAO and insulin signalling genes. The 7CafD females, compared to 7CafD males, had more WAT, and a reduced expression of FAO genes in muscles and thermogenic genes in WAT. The 17CafD females, compared to 17CafD males, had lower plasma leptin and insulin levels, and higher insulin sensitivity and expression of insulin signalling genes in the liver and muscles. Thus, the initiation of the obesogenic diet before puberty led to a more adaptive metabolic phenotypes in males, and after puberty, in females.
Collapse
Affiliation(s)
- Nadezhda Bazhan
- The Laboratory of Physiological Genetics, The Institute of Cytology and Genetics, 630090 Novosibirsk, Russia; (A.K.); (A.D.); (N.B.); (T.J.); (E.M.)
| | | | | | | | | | | |
Collapse
|
44
|
Chankamngoen W, Thammayon N, Suntornsaratoon P, Nammultriputtar K, Kitiyanant N, Donpromma N, Chaichanan J, Supcharoen P, Teerapo K, Teerapornpuntakit J, Rodrat M, Panupinthu N, Svasti S, Wongdee K, Charoenphandhu N. Fibroblast growth factor-21 potentiates the stimulatory effects of 1,25-dihydroxyvitamin D 3 on transepithelial calcium transport and TRPV6 Ca 2+ channel expression. Biochem Biophys Res Commun 2024; 733:150429. [PMID: 39053106 DOI: 10.1016/j.bbrc.2024.150429] [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: 05/28/2024] [Revised: 07/15/2024] [Accepted: 07/18/2024] [Indexed: 07/27/2024]
Abstract
Fibroblast growth factor (FGF)-21 is a salient liver-derived endocrine regulator for metabolism of glucose and triglyceride as well as bone remodeling. Previously, certain peptides in the FGF family have been shown to modulate calcium absorption across the intestinal epithelia. Since FGF21 receptor, i.e., FGF receptor-1, is abundantly expressed in the enterocytes, there was a possibility that FGF21 might exert direct actions on the intestine. Herein, a large-scale production of recombinant FGF21 at the multi-gram level was developed in order to minimize variations among various batches. In the oral glucose tolerance test, recombinant FGF21 was found to reduce plasma glucose levels in mice fed high-fat diet. A series of experiments applying radioactive tracer 45Ca in Ussing chamber showed that FGF21 potentiated the stimulatory effect of low-dose 1,25-dihydroxyvitamin D3 [10 nM 1,25(OH)2D3] on the transepithelial calcium transport across intestinal epithelium-like Caco-2 monolayer. FGF21 + 1,25(OH)2D3 also decreased transepithelial resistance, but had no effect on epithelial potential difference or short-circuit current. Furthermore, 1,25(OH)2D3 alone upregulated the Caco-2 mRNA expression of the major apical calcium channels, i.e., transient receptor potential vanilloid subfamily member 6 (TRPV6), which was further elevated by a combination of FGF21 and 1,25(OH)2D3, consistent with the upregulated TRPV6 protein expression in enterocytes of FGF21-treated mice. However, FGF21 was without effects on the mRNA expression of voltage-gated calcium channel 1.3, calbindin-D9k, plasma membrane Ca2+-ATPase 1b, claudin-12 or claudin-15. In conclusion, FGF21 did exert a direct action on the intestinal epithelial cells by potentiating the 1,25(OH)2D3-enhanced calcium transport, presumably through the upregulation of TRPV6 expression.
Collapse
Affiliation(s)
- Wasutorn Chankamngoen
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Nithipak Thammayon
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Panan Suntornsaratoon
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, Thailand; Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Ketsaraporn Nammultriputtar
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, Thailand; Graduate Program in Molecular Medicine, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Narisorn Kitiyanant
- Center for Advanced Therapeutics (CAT), Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
| | - Natthida Donpromma
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, Thailand; Center for Advanced Therapeutics (CAT), Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
| | - Jirapan Chaichanan
- Establishment of Mahidol University Bio-industrial Development Center, Mahidol University, Nakhon Pathom, Thailand
| | - Promsup Supcharoen
- Mahidol University Frontier Research Facility (MU-FRF), Mahidol University, Nakhon Pathom, Thailand
| | - Kittitat Teerapo
- Mahidol University Frontier Research Facility (MU-FRF), Mahidol University, Nakhon Pathom, Thailand
| | | | - Mayuree Rodrat
- Center for Advanced Therapeutics (CAT), Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
| | - Nattapon Panupinthu
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, Thailand; Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Saovaros Svasti
- Thalassemia Research Center, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
| | - Kannikar Wongdee
- Faculty of Allied Health Sciences, Burapha University, Chonburi, Thailand.
| | - Narattaphol Charoenphandhu
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, Thailand; Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand; Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand; The Academy of Science, The Royal Society of Thailand, Bangkok, Thailand.
| |
Collapse
|
45
|
Chen Q, Cheng W, Zhang J, Chi C, Lin M, He C, Liao Z, Gong F. Fibroblast growth factor 21 improves insulin sensitivity by modulating the bile acid-gut microbiota axis in type Ⅱ diabetic mice. Free Radic Biol Med 2024; 224:600-617. [PMID: 39288846 DOI: 10.1016/j.freeradbiomed.2024.09.017] [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: 06/12/2024] [Revised: 08/23/2024] [Accepted: 09/12/2024] [Indexed: 09/19/2024]
Abstract
BACKGROUND Fibroblast growth factor 21 (FGF21) is an important regulator of glycolipid metabolism. However, whether the gut microbiota is related to the anti-diabetic and obesity effects of FGF21 remains unclear. METHODS Our research used KO/KO db/db male mice and streptozotocin (STZ)-induced to simulate the construction of two type II diabetic mellitus (T2DM) models, and detected impaired glucose tolerance in the model by using the ipGTT and ITT assays, and collected feces from the model mice for sequencing of the intestinal flora and the content of short-chain fatty acids. H&E staining was used to detect changes in intestinal tissue, the serum levels of LPS and GLP-1 were detected by ELISA. RESULTS In this study, we found that FGF21 significantly improved insulin sensitivity, attenuated intestinal lesions, and decreased serum lipopolysaccharide (LPS) concentrations in T2DM mice. Moreover, FGF21 reshaped the gut microbiota and altered their metabolic pathways in T2DM mice, promoting the production of short-chain fatty acids (SCFAs) and the secretion of glucagon-like peptide 1 (GLP-1). Fecal transplantation experiments further confirmed that feces from FGF21-treated diabetic mice demonstrated similar effects as FGF21 in terms of anti-diabetic activity and regulation of gut microbiota dysbiosis. Additionally, the antibiotic depletion of gut microbiota abolished the beneficial effects of FGF21, including increased GLP-1 secretion and fecal SCFA concentration. Additionally, the FGF21 effects of ameliorating intestinal damage and suppressing plasma LPS secretion were suppressed. All these findings suggest that FGF21 prevents intestinal lesions by modifying the gut microbiota composition. Furthermore, FGF21 affected bile acid synthesis by inhibiting CYP7A1, the key enzyme of bile acid synthesis. CONCLUSSION Therefore, FGF21 enriched beneficial bacteria by preventing bile acid synthesis and stimulating the secretion of the intestinal hormone GLP-1 via the increased production of gut microbiota metabolites, thereby exerting its anti-diabetic effects.
Collapse
Affiliation(s)
- Qiongzhen Chen
- College of Life and Environmental Science, Wenzhou University, Wenzhou, 325000, China
| | - Wenwen Cheng
- College of Life and Environmental Science, Wenzhou University, Wenzhou, 325000, China
| | - Jiangnan Zhang
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325015, China
| | - Changxing Chi
- Department of Endocrinology, Yanbian University Hospital, Yanji, 136200, China
| | - Mengyi Lin
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325015, China
| | - Chenbei He
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325015, China
| | - Zhiyong Liao
- College of Life and Environmental Science, Wenzhou University, Wenzhou, 325000, China.
| | - Fanghua Gong
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325015, China.
| |
Collapse
|
46
|
Morita H, Hoshiga M. Fibroblast Growth Factors in Cardiovascular Disease. J Atheroscler Thromb 2024; 31:1496-1511. [PMID: 39168622 PMCID: PMC11537794 DOI: 10.5551/jat.rv22025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 08/23/2024] Open
Abstract
Despite advancements in managing traditional cardiovascular risk factors, many cardiovascular diseases (CVDs) persist. Fibroblast growth factors (FGFs) have emerged as potential diagnostic markers and therapeutic targets for CVDs. FGF1, FGF2, and FGF4 are primarily used for therapeutic angiogenesis. Clinical applications are being explored based on animal studies using approaches such as recombinant protein administration and adenovirus-mediated gene delivery, targeting patients with coronary artery disease and lower extremity arterial disease. Although promising results have been observed in animal models and early-stage clinical trials, further studies are required to assess their therapeutic potential. The FGF19 subfamily, consisting of FGF19, FGF21, and FGF23, act via endocrine signaling in various organs. FGF19, primarily expressed in the small intestine, plays important roles in glucose, lipid, and bile acid metabolism and has therapeutic potential for metabolic disorders. FGF21, found in various tissues, improves glucose metabolism and insulin sensitivity, suggesting potential for treating obesity and diabetes. FGF23, primarily secreted by osteocytes, regulates vitamin D and phosphate metabolism and serves as an important biomarker for chronic kidney disease and CVDs. Thus, FGFs holds promise for both therapeutic and diagnostic applications in metabolic and cardiovascular diseases. Understanding the mechanisms of FGF may pave the way for novel strategies to prevent and manage CVDs, potentially addressing the limitations of current treatments. This review explores the roles of FGF1, FGF2, FGF4, and the FGF19 subfamily in maintaining cardiovascular health. Further research and clinical trials are crucial to fully understand the therapeutic potential of FGFs in managing cardiovascular health.
Collapse
Affiliation(s)
- Hideaki Morita
- Department of Cardiology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Masaaki Hoshiga
- Department of Cardiology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| |
Collapse
|
47
|
Wang J, Li L, Li L, Shen Y, Qiu F. Lycopene alleviates age-related cognitive deficit via activating liver-brain fibroblast growth factor-21 signalling. Redox Biol 2024; 77:103363. [PMID: 39307046 PMCID: PMC11447408 DOI: 10.1016/j.redox.2024.103363] [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: 08/21/2024] [Revised: 09/17/2024] [Accepted: 09/18/2024] [Indexed: 10/06/2024] Open
Abstract
Brain function is linked with many peripheral tissues, including the liver, where hepatic fibroblast growth factor 21 (FGF21) mediates communication between the liver and brain. Lycopene (LYC), a naturally occurring carotenoid, posses multiple health-promoting properties, including neuroprotective function. Here, we investigated the effects of LYC on age-related memory impairment and the relative contribution of liver-brain FGF21 signaling in these process. The results showed that after treatment with LYC for 3 months, brain aging and age-related cognitive deficits were effectively managed. In addition, LYC ameliorated neuronal degeneration, mitochondrial dysfunction and synaptic damage, and promoted synaptic vesicle fusion in 18-month-old mice. Notably, LYC activated liver-brain FGF21 signalling in aging mice. Whereas all these central effects of LYC were negated by blocking FGF21 via i. v. injection of adeno-associated virus in aging mice. Furthermore, recombinant FGF21 elevated mitochondrial ATP levels and enhanced synaptic vesicle fusion in mouse hippocampal HT-22 cells, which promoted neurotransmitter release. Additionally, we co-cultured hepatocytes and neurons in Transwell and found that LYC enhanced hepatocytes' support for neurons. This support included improved cell senescence, enhanced mitochondrial function, and increased axon length in co-cultured neurons. In conclusion, LYC protects against age-related cognitive deficit, partly explained by activating liver-brain FGF21 signalling, hence promoting neurotransmitters release via increasing mitochondrial ATP levels and enhancing synaptic vesicle fusion. These findings revealed that FGF21 could be a potential therapeutical target in nutritional intervention strategies to improve cognitive damage caused by aging and age-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Jia Wang
- Nutritional and Food Sciences Research Institute, Department of Nutrition and Food Hygiene, School of Public Health, Shanxi Medical University, Taiyuan, 030001, China; MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, School of Public Health, Shanxi Medical University, Taiyuan, 030001, China.
| | - Lu Li
- Nutritional and Food Sciences Research Institute, Department of Nutrition and Food Hygiene, School of Public Health, Shanxi Medical University, Taiyuan, 030001, China
| | - Li Li
- Nutritional and Food Sciences Research Institute, Department of Nutrition and Food Hygiene, School of Public Health, Shanxi Medical University, Taiyuan, 030001, China
| | - Yuqi Shen
- Nutritional and Food Sciences Research Institute, Department of Nutrition and Food Hygiene, School of Public Health, Shanxi Medical University, Taiyuan, 030001, China
| | - Fubin Qiu
- Nutritional and Food Sciences Research Institute, Department of Nutrition and Food Hygiene, School of Public Health, Shanxi Medical University, Taiyuan, 030001, China; MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, School of Public Health, Shanxi Medical University, Taiyuan, 030001, China.
| |
Collapse
|
48
|
Jian H, Li R, Huang X, Li J, Li Y, Ma J, Zhu M, Dong X, Yang H, Zou X. Branched-chain amino acids alleviate NAFLD via inhibiting de novo lipogenesis and activating fatty acid β-oxidation in laying hens. Redox Biol 2024; 77:103385. [PMID: 39426289 PMCID: PMC11536022 DOI: 10.1016/j.redox.2024.103385] [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: 09/05/2024] [Revised: 10/02/2024] [Accepted: 10/02/2024] [Indexed: 10/21/2024] Open
Abstract
The adverse metabolic impacts of branched-chain amino acids (BCAA) have been elucidated are mediated by isoleucine and valine. Dietary restriction of isoleucine promotes metabolic health and increases lifespan. However, a high protein diet enriched in BCAA is presently the most useful therapeutic strategy for nonalcoholic fatty liver disease (NAFLD), yet, its underlying mechanism remains largely unknown. Fatty liver hemorrhagic syndrome (FLHS), a specialized laying hen NAFLD model, can spontaneously develop fatty liver and hepatic steatosis under a high-energy and high-protein dietary background that the pathogenesis of FLHS is similar to human NAFLD. The mechanism underlying dietary BCAA control of NAFLD development in laying hens remains unclear. Herein, we demonstrate that dietary supplementation with 67 % High BCAA has unique mitigative impacts on NAFLD in laying hens. A High BCAA diet alleviates NAFLD, by inhibiting the tryptophan-ILA-AHR axis and MAPK9-mediated de novo lipogenesis (DNL), promoting ketogenesis and energy metabolism, and activating PPAR-RXR and pexophagy to promote fatty acid β-oxidation. Furthermore, we uncover that High BCAA strongly activates ubiquitin-proteasome autophagy via downregulating UFMylation to trigger MAPK9-mediated DNL, fatty acid elongation and lipid droplet formation-related proteins ubiquitination degradation, activating PPAR-RXR and pexophagy mediated fatty acid β-oxidation and lipolysis. Together, our data highlight moderating intake of high BCAA by inhibiting the AHR/MAPK9 are promising new strategies in NAFLD and FLHS treatment.
Collapse
Affiliation(s)
- Huafeng Jian
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Hangzhou, 310058, China; Xianghu Laboratory, Hangzhou, 311231, China
| | - Ru Li
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Hangzhou, 310058, China
| | - Xuan Huang
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Hangzhou, 310058, China
| | - Jiankui Li
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Hangzhou, 310058, China
| | - Yan Li
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Hangzhou, 310058, China
| | | | - Mingkun Zhu
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, 212100, China; Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, 212100, China
| | - Xinyang Dong
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Hangzhou, 310058, China
| | - Hua Yang
- Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China.
| | - Xiaoting Zou
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Key Laboratory of Molecular Animal Nutrition (Zhejiang University), Ministry of Education, Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products, Hangzhou, 310058, China.
| |
Collapse
|
49
|
Enyew Belay K, Jemal RH, Tuyizere A. Innovative Glucagon-based Therapies for Obesity. J Endocr Soc 2024; 8:bvae197. [PMID: 39574787 PMCID: PMC11579655 DOI: 10.1210/jendso/bvae197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Indexed: 11/24/2024] Open
Abstract
Obesity poses a significant global health challenge, with an alarming rise in prevalence rates. Traditional interventions, including lifestyle modifications, often fall short of achieving sustainable weight loss, ultimately leading to surgical interventions, which carry a significant burden and side effects. This necessitates the exploration of effective and relatively tolerable pharmacological alternatives. Among emerging therapeutic avenues, glucagon-based treatments have garnered attention for their potential to modulate metabolic pathways and regulate appetite. This paper discusses current research on the physiological mechanisms underlying obesity and the role of glucagon in energy homeostasis. Glucagon, traditionally recognized for its glycemic control functions, has emerged as a promising target for obesity management due to its multifaceted effects on metabolism, appetite regulation, and energy expenditure. This review focuses on the pharmacological landscape, encompassing single and dual agonist therapies targeting glucagon receptors (GcgRs), glucagon-like peptide-1 receptors (GLP-1Rs), glucose-dependent insulinotropic polypeptide receptors (GIPRs), amylin, triiodothyronine, fibroblast growth factor 21, and peptide tyrosine tyrosine. Moreover, novel triple-agonist therapies that simultaneously target GLP-1R, GIPR, and GcgR show promise in augmenting further metabolic benefits. This review paper tries to summarize key findings from preclinical and clinical studies, elucidating the mechanisms of action, safety profiles, and therapeutic potential of glucagon-based therapies in combating obesity and its comorbidities. Additionally, it explores ongoing research endeavors, including phase III trials, aimed at further validating the efficacy and safety of these innovative treatment modalities.
Collapse
Affiliation(s)
- Kibret Enyew Belay
- Department of Internal Medicine, Endocrinology and Metabolism Unit, Bahir Dar University, Bahir Dar 6000, Ethiopia
| | - Rebil Heiru Jemal
- Department of Internal Medicine, Adama Hospital Medical College, Adama 1000, Ethiopia
| | - Aloys Tuyizere
- Department of Internal Medicine, Endocrinology, Diabetes and Metabolism Unit, University of Rwanda, Kigali 00200, Rwanda
| |
Collapse
|
50
|
Shi X, Zheng Q, Wang X, Guo W, Lin Z, Gao Y, Shore E, Martin RC, Lv G, Li Y. Compromised macrophages contribute to progression of MASH to hepatocellular carcinoma in FGF21KO mice. SCIENCE ADVANCES 2024; 10:eado9311. [PMID: 39441934 PMCID: PMC11498219 DOI: 10.1126/sciadv.ado9311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 09/17/2024] [Indexed: 10/25/2024]
Abstract
Metabolic dysfunction-associated steatohepatitis is well accepted as a potential precursor of hepatocellular carcinoma. Previously, we reported that fibroblast growth factor 21 (FGF21) revealed a novel anti-inflammatory activity via inhibiting the TLR4-IL-17A signaling, which could be a potential anticarcinogenetic mechanism to prevent to MASH-HCC transition. Here, we set out to determine whether FGF21 has a major impact on Kupffer cells' (KCs) ability during MASH-HCC transition. We found aberrant hepatic FGF21 and KC pool in human MASH-HCC. Lack of FGF21 up-regulated ALOX15, which converted the oxidized fatty acids to induce excessive KC death and mobilization of monocyte-derived macrophages (MoMFs) for KC replacement. Lack of FGF21 oversupplied free fatty acids for sphingosine-1-phosphate (S1P) cascade synthesis to mediate MASH-HCC transition via S1P-YAP signaling and cross-talk between tumor cells and macrophages. In conclusion, lack of FGF21 accelerated MASH-HCC transition via the S1P-AP signaling. Compromised MoMFs could present as tumor-associated macrophage phenotype rendering tumor immune microenvironment for MASH-HCC transition.
Collapse
Affiliation(s)
- Xiaoju Shi
- Department of Surgery, School of Medicine, University of Louisville, Louisville, KY 40202, USA
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun 130021, China
| | - Qianqian Zheng
- Department of Surgery, School of Medicine, University of Louisville, Louisville, KY 40202, USA
- Department of Pathophysiology, Basic Medicine College, China Medical University, Shenyang 110122, China
| | - Xingtong Wang
- Department of Surgery, School of Medicine, University of Louisville, Louisville, KY 40202, USA
- Department of Hematology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Wei Guo
- Department of Surgery, School of Medicine, University of Louisville, Louisville, KY 40202, USA
- Department of Hematology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Ziqi Lin
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110022, China
| | - Yonglin Gao
- Department of Surgery, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Emily Shore
- Department of Surgery, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Robert C. Martin
- Department of Surgery, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | - Guoyue Lv
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun 130021, China
| | - Yan Li
- Department of Surgery, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| |
Collapse
|