1
|
Yang R, Jiang Q, Liu W, Wang F, Cao S. Serum polychlorinated biphenyls as a risk factor for MASLD: Exploring the association and underlying mechanisms. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 981:179617. [PMID: 40354702 DOI: 10.1016/j.scitotenv.2025.179617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 04/30/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025]
Abstract
BACKGROUND Fatty liver disease, a growing global health issue, is closely tied to metabolic disorders. The 2023 definition of metabolic-associated steatotic liver disease (MASLD) incorporates cardiometabolic risk factors, but the potential role of persistent organic pollutants (POPs) like polychlorinated biphenyls (PCBs), remains underexplored. Investigating the impact of environmental toxins on liver health is crucial for understanding emerging public health risks. METHODS 1080 participants were included from the 1999-2018 National Health and Nutrition Examination Survey (NHANES). We employed weighted generalized linear models, weighted quantile sum regression, and Bayesian kernel machine regression to assess the relationship between serum PCB levels and MASLD, with NAFLD included for comparison. Protein interaction and enrichment analyses were also conducted to explore the underlying mechanisms. RESULTS PCB146, PCB156, PCB187, PCB174, and PCB180 were significantly associated with an increased MASLD risk in the GLM. Significant positive associations were found between serum PCB mixtures and MASLD in the WQS model (β: 0.411, p: 0.0056) and BKMR model (p < 0.05), with PCB180 contributing the most (β: 0.644, PIP: 0.903). NAFLD did not show significant associations. Network pharmacological analysis demonstrated enrichment in the regulation of lipolysis of adipocytes and the cAMP signaling pathway, and PPAR-γ and MAOA show significant importance in the protein-protein interaction networks. CONCLUSION This study underscores the epidemiological and mechanical link between MASLD and PCB exposure, highlighting the superiority of MASLD in identifying the impact of POPs on liver disease risk and particularly identifying PCB180 as a sentinel marker for PCB surveillance.
Collapse
Affiliation(s)
- Ruichen Yang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Qingqing Jiang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China; School of Nursing, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Wentao Liu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Furong Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China; School of Nursing, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Shiyi Cao
- School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| |
Collapse
|
2
|
Taheri E, Yilmaz Y, Ghorat F, Moslem A, Zali MR. Association of diet quality scores with risk of metabolic-associated fatty liver disease in Iranian population: a nested case-control study. J Diabetes Metab Disord 2025; 24:46. [PMID: 39816985 PMCID: PMC11729581 DOI: 10.1007/s40200-024-01544-x] [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: 08/21/2024] [Accepted: 12/05/2024] [Indexed: 01/18/2025]
Abstract
Background and aim A healthy diet has been recommended for non-alcoholic fatty liver disease (NAFLD). We aim to investigate the associations of diet quality indices with the risk of developingmetabolic-associated fatty liver disease (MAFLD). Methods We conducted this nested case-control study by recruiting 968 cases with MAFLD and 964 controls from the participants of the baseline phase of the Sabzevar Persian Cohort Study (SPCS). MAFLD was defined as having a fatty liver index ≥ 60 plus at least one of the following: overweight or obese, Type II diabetes mellitus, or evidence of metabolic dysregulation. Healthy Eating Index-2015 (HEI-2015) and Alternative Healthy Eating Index-2010 (AHEI-2010) were calculated from a validated food frequency questionnaire. We estimated the associations of HEI-2015 and AHEI-2010 with MAFLD risk using multivariable logistic regression. Results Among those in the highest relative to the lowest quintile of HEI-2015 and AHEI-2010, the multivariable-adjusted odds ratios (OR) were 0.45 (95% CI [confidence interval] 0.29-0.69; P trend = 0.002) and 0.55 (95% CI 0.35-0.85; P trend = 0.04), respectively. Conclusion The results of our study suggest that there is a significant associationbetween adherence to a healthy diet, indicated by a higher score of HEI or AHEI, and a reduced likelihood of developingMAFLD. Supplementary Information The online version contains supplementary material available at 10.1007/s40200-024-01544-x.
Collapse
Affiliation(s)
- Ehsaneh Taheri
- Liver and Pancreatobiliary Diseases Research Center, Digestive Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Yusuf Yilmaz
- Department of Gastroenterology, School of Medicine, Recep Tayyip Erdoğan University, Rize, Türkiye
| | - Fereshteh Ghorat
- Non-communicable Diseases Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Alireza Moslem
- Department of Anesthesiology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Reza Zali
- Gastroenterology and Liver Disease Research Center, Research Institute for Gastroenterology and Liver Disease, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
Zhang X, Nguyen MH. Metabolic dysfunction-associated steatotic liver disease: A sexually dimorphic disease and breast and gynecological cancer. Metabolism 2025; 167:156190. [PMID: 40081614 DOI: 10.1016/j.metabol.2025.156190] [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: 01/02/2025] [Revised: 02/26/2025] [Accepted: 03/09/2025] [Indexed: 03/16/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) has become a global public health and economic burden worldwide in the past few decades. Epidemiological studies have shown that MASLD is a multisystem disease that is associated not only with liver-related complications but also with an increased risk of developing extrahepatic cancers. MASLD is a sexually dimorphic disease with sex hormones playing an important role in the development and progression of MASLD, especially by the levels and ratios of circulating estrogens and androgens. MASLD is associated with hormone-sensitive cancers including breast and gynecological cancer. The risk of breast and gynecological cancer is elevated in individuals with MASLD driven by shared metabolic risk factors including obesity and insulin resistance. Multiple potential mechanisms underline these associations including metabolic dysfunction, gut dysbiosis, chronic inflammation and dysregulated release of hepatokines. However, the effect of hormone therapy including hormone replacement therapy and anti-estrogen treatment on MASLD and female-specific cancers remains debatable at this time. This synopsis will review the associations between MASLD and breast and gynecological cancer, their underlying mechanisms, implications of hormonal therapies, and their future directions.
Collapse
Affiliation(s)
- Xinrong Zhang
- Division of Gastroenterology and Hepatology, School of Medicine, Stanford University Medical Center, Palo Alto, CA, United States
| | - Mindie H Nguyen
- Division of Gastroenterology and Hepatology, School of Medicine, Stanford University Medical Center, Palo Alto, CA, United States; Department of Epidemiology and Population Health, Stanford University Medical Center, Palo Alto, CA, United States; Stanford Cancer Institute, Stanford University Medical Center, Palo Alto, CA, United States.
| |
Collapse
|
4
|
Zhang S, Yang Y, Yang L, Meng F, Mu Y, Yuan Y, Zhang Y. The role of PPARs family members in acne. J Dermatol Sci 2025:S0923-1811(25)00043-X. [PMID: 40413060 DOI: 10.1016/j.jdermsci.2025.04.003] [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: 01/11/2025] [Revised: 03/03/2025] [Accepted: 04/01/2025] [Indexed: 05/27/2025]
Abstract
Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors that play a crucial role in the genetic regulation of lipid metabolism and energy homeostasis. There is increasing evidence to suggest that PPARs may have a significant impact on the development and progression of acne, including its role in regulating lipid production, inhibiting keratinocyte proliferation, and reducing acne-related inflammation. As such, PPARs present themselves as promising therapeutic targets for both the prevention and treatment of acne. This article provides an overview of the role of PPARs in the pathological processes underlying acne, with particular emphasis on inflammation and lipid regulation.
Collapse
Affiliation(s)
- Shaoyang Zhang
- Yunnan Key Laboratory of Breast Cancer Precision Medicine, Academy of Biomedical Engineering, Kunming Medical University, Kunming, China
| | - Yating Yang
- Yunnan Key Laboratory of Breast Cancer Precision Medicine, Academy of Biomedical Engineering, Kunming Medical University, Kunming, China
| | - Li Yang
- Yunnan Key Laboratory of Breast Cancer Precision Medicine, Academy of Biomedical Engineering, Kunming Medical University, Kunming, China
| | - Fangyu Meng
- Yunnan Key Laboratory of Breast Cancer Precision Medicine, Academy of Biomedical Engineering, Kunming Medical University, Kunming, China
| | - Yizhe Mu
- Yunnan Key Laboratory of Breast Cancer Precision Medicine, Academy of Biomedical Engineering, Kunming Medical University, Kunming, China
| | - Yuhang Yuan
- Yunnan Key Laboratory of Breast Cancer Precision Medicine, Academy of Biomedical Engineering, Kunming Medical University, Kunming, China
| | - Yuan Zhang
- Yunnan Key Laboratory of Breast Cancer Precision Medicine, Academy of Biomedical Engineering, Kunming Medical University, Kunming, China.
| |
Collapse
|
5
|
Jaskulak M, Zimowska M, Rolbiecka M, Zorena K. Understanding the role of endocrine disrupting chemicals as environmental obesogens in the obesity epidemic: A comprehensive overview of epidemiological studies between 2014 and 2024. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 299:118401. [PMID: 40412253 DOI: 10.1016/j.ecoenv.2025.118401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 05/19/2025] [Accepted: 05/20/2025] [Indexed: 05/27/2025]
Abstract
The prevalence of obesity has reached epidemic proportions worldwide, posing a significant public health concern due to its association with various chronic diseases and healthcare costs. In addition to traditional risk factors such as diet and physical activity, emerging evidence suggests that environmental pollutants, termed obesogens, may contribute to the obesity epidemic. Obesogens are endocrine-disrupting chemicals (EDCs) that can alter lipid homeostasis, promote adipogenesis, and disrupt metabolic regulation, leading to increased adiposity and obesity risk. This review explores available data from human studies published in the last decade, along with the mechanisms underlying obesogenic action, including their effects on adipocyte differentiation, adipose tissue development, and metabolic regulation. Overall, 75 studies were analyzed. Early-life exposure during critical developmental windows has been shown to increase obesity risk later in life, potentially through epigenetic modifications and transgenerational effects. Epidemiological studies provide evidence of associations between prenatal or early-life exposure and increased obesity risk in offspring. Additionally, study found more consistent associations between exposure to some EDCs (including phthalates, parabens, and bisphenols) and obesity or metabolic outcomes in children and women, while results for other chemicals (i.e. PFAS and organochlorine pesticides) were more heterogeneous, especially in adolescents and adults. Key findings indicate consistent associations between phthalate exposure and obesity in children, with mixed results for adults. Future research should focus on elucidating the full spectrum of obesogens, their mechanisms of action, and their implications for obesity risk across generations. This knowledge will inform preventive strategies and public health interventions aimed at addressing the obesity epidemic and its associated health burden.
Collapse
Affiliation(s)
- Marta Jaskulak
- Department of Immunobiology and Environmental Microbiology, Department of Health Sciences, Medical University of Gdansk, Poland.
| | - Malwina Zimowska
- Department of Immunobiology and Environmental Microbiology, Department of Health Sciences, Medical University of Gdansk, Poland
| | - Marta Rolbiecka
- Department of Immunobiology and Environmental Microbiology, Department of Health Sciences, Medical University of Gdansk, Poland
| | - Katarzyna Zorena
- Department of Immunobiology and Environmental Microbiology, Department of Health Sciences, Medical University of Gdansk, Poland
| |
Collapse
|
6
|
Okada M, Hanayama M, Yamamoto Y, Miyake T, Yoshida O, Takeshita E, Ikeda Y, Hiasa Y. Effect of pemafibrate in reducing intestinal long-chain fatty acid absorption and hepatic fibrosis in metabolic dysfunction-associated steatohepatitis rats. BMC Gastroenterol 2025; 25:385. [PMID: 40389836 PMCID: PMC12090548 DOI: 10.1186/s12876-025-03967-z] [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: 12/06/2024] [Accepted: 05/02/2025] [Indexed: 05/21/2025] Open
Abstract
BACKGROUND Pemafibrate helps regulate fatty acid dynamics in the liver, potentially preventing metabolic dysfunction-associated steatohepatitis (MASH). However, its effect on intestinal long-chain fatty acid (LCFA) metabolism in MASH remains unclear. Thus, we aimed to examine the influence of pemafibrate on intestinal LCFA metabolism and hepatic fibrosis in a MASH rat model. METHODS Sprague-Dawley rats were fed a high-fat and high-cholesterol diet to induce MASH and then divided into pemafibrate-treated (pemafibrate (+)) and untreated (pemafibrate (-)) groups. Triglyceride deposition in the small intestine and fibrosis, along with α-smooth muscle actin level in the liver, were evaluated. Furthermore, the mRNA expression levels of genes associated with lipid metabolism in the small intestine and markers of fibrosis and hepatic stellate cells activation in the liver were measured. RESULTS The pemafibrate-treated group had markedly lower triglyceride deposition and lipid absorption in the intestine, and significantly lower levels of molecules involved in intestinal lipid regulation than the pemafibrate-untreated group. Moreover, hepatic fibrosis significantly improved, and the mRNA levels of fibrosis-related molecules and hepatic stellate cell activation factors significantly decreased in the pemafibrate-treated compared with those in the pemafibrate-untreated group. CONCLUSIONS Pemafibrate reduced lipid droplet formation and LCFA absorption in the intestinal tract and alleviated hepatic fibrosis in MASH model rats.
Collapse
Affiliation(s)
- Masaya Okada
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Masakazu Hanayama
- Department of Gastroenterology, Matsuyama Shimin Hospital, Matsuyama, Ehime, Japan
| | - Yasunori Yamamoto
- Endoscopy Center, Ehime University Hospital, 454 Shitsukawa, Toon, Ehime, 791-0295, Japan.
| | - Teruki Miyake
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Osamu Yoshida
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Eiji Takeshita
- Department of Inflammatory Bowel Diseases and Therapeutics, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Yoshio Ikeda
- Endoscopy Center, Ehime University Hospital, 454 Shitsukawa, Toon, Ehime, 791-0295, Japan
| | - Yoichi Hiasa
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| |
Collapse
|
7
|
Supplee JG, Marmorstein R, Wellen KE. Molecular targets of bempedoic acid and related decoy fatty acids. Trends Endocrinol Metab 2025:S1043-2760(25)00077-3. [PMID: 40345862 DOI: 10.1016/j.tem.2025.04.002] [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: 02/03/2025] [Revised: 03/28/2025] [Accepted: 04/07/2025] [Indexed: 05/11/2025]
Abstract
Disorders of lipid metabolism, including hyperlipidemia, atherosclerosis, and metabolic dysfunction-associated steatotic liver disease, are increasing across the globe. Bempedoic acid (BPA) is a first-in-class drug for the treatment of hypercholesterolemia and cardiac risk reduction, which may particularly benefit those who do not tolerate statins. Inhibition of hepatic ATP-citrate lyase (ACLY) is widely accepted as the main mediator of its observed clinical effects. However, BPA treatment also has ACLY-independent effects on lipid metabolism, as the structural similarity of BPA to endogenous fatty acids allows it to trigger multiple lipid-signaling pathways. Here, we review the molecular targets of BPA and related 'decoy fatty acid' drugs and identify areas where further study is warranted as these molecules are evaluated for clinical indications.
Collapse
Affiliation(s)
- Julianna G Supplee
- Graduate Group in Biochemistry, Biophysics and Chemical Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Ronen Marmorstein
- Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA; Department of Biochemistry and Biophysics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| | - Kathryn E Wellen
- Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA; Department of Cancer Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
8
|
Guo Y, Xu N, Meng Q, Zhong M, Yang M, Xu F, Zhang L, Jiang M, Wu J, Ma Z, Xu Y, Li Y. Mechanisms of Inonotus obliquus (Fr.) Pilát polysaccharides in ameliorating lipid-induced skeletal muscle insulin resistance via PI3K/AKT and AMPK/ACC1/CPT1 signaling pathways. JOURNAL OF ETHNOPHARMACOLOGY 2025; 349:119938. [PMID: 40348308 DOI: 10.1016/j.jep.2025.119938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/14/2025] [Accepted: 05/07/2025] [Indexed: 05/14/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Inonotus obliquus (Fr.) Pilát, a traditional medicinal fungi, has been used to treat diabetes in China and Russia since the 16th century. Recent studies show Inonotus obliquus (Fr.) Pilát polysaccharides (IOP) have hypoglycemic and lipid-lowering effects in type 2 diabetes mellitus (T2DM) mice and can recover liver insulin resistance. AIM OF THE STUDY This study aims to assess the effect of IOP and its mechanisms in ameliorating insulin resistance and lipid metabolism disorders in T2DM. MATERIALS AND METHODS The potential targets of IOP for T2DM were identified by network pharmacology and molecular docking. In vitro, an insulin resistance model in C2C12 cells was induced, and IOP's effects on glucose uptake, glycogen, lipid content, and lipid metabolism-related mRNAs were assessed. In vivo, a T2DM mice model was established. Blood glucose, lipids, glucose tolerance, and insulin sensitivity were evaluated. Histopathology was used to assess morphological changes in mice skeletal muscle. Western blotting was utilized to evaluate the expression levels of PI3K/AKT and AMPK/ACC1/CPT1 signaling pathway proteins both in vivo and in vitro. RESULTS Network pharmacology results showed IOP and T2DM targets were enriched in PI3K/AKT, insulin resistance, and lipid metabolism pathways. Cell experiments showed IOP enhanced glucose uptake and glycogen content, reduced lipid content, and improved lipid deposition in insulin-resistant C2C12 cells. Animal experiments showed IOP improved hyperglycemia and hyperlipidemia, enhanced glucose tolerance and insulin sensitivity, and reduced insulin resistance in T2DM mice. Western blot showed IOP activated PI3K/AKT and AMPK/ACC1/CPT1 pathways, promoting GLUT4 expression and translocation, and GSK3β phosphorylation. CONCLUSIONS In summary, the results indicated that IOP was able to ameliorate lipid-induced skeletal muscle insulin resistance in T2DM. The mechanism may be related to the PI3K/AKT and AMPK/ACC1/CPT1 signaling pathways.
Collapse
Affiliation(s)
- Yiming Guo
- Department of Pathophysiology, School of Basic Medicine, Binzhou Medical University, Yantai, Shandong Province, China
| | - Nuo Xu
- Department of Intensive Care Medicine, The Second School of Clinical Medical, Binzhou Medical University, Yantai, Shandong Province, China
| | - Qinyu Meng
- Department of Pathophysiology, School of Basic Medicine, Binzhou Medical University, Yantai, Shandong Province, China
| | - Mengru Zhong
- Department of Pathophysiology, School of Basic Medicine, Binzhou Medical University, Yantai, Shandong Province, China
| | - Meizi Yang
- Department of Pathophysiology, School of Basic Medicine, Binzhou Medical University, Yantai, Shandong Province, China
| | - Fang Xu
- Department of Pathophysiology, School of Basic Medicine, Binzhou Medical University, Yantai, Shandong Province, China
| | - Lei Zhang
- Department of Intensive Care Medicine, The Second School of Clinical Medical, Binzhou Medical University, Yantai, Shandong Province, China
| | - Muchen Jiang
- Department of Pharmacy, School of Pharmacy, Binzhou Medical University, Yantai, Shandong Province, China
| | - Junze Wu
- Department of Clinical Medicine, The First School of Clinical Medical, Binzhou Medical University, Yantai, Shandong Province, China
| | - Zihan Ma
- Department of Clinical Medicine, The First School of Clinical Medical, Binzhou Medical University, Yantai, Shandong Province, China
| | - Yingjiang Xu
- Department of Clinical Medicine, The First School of Clinical Medical, Binzhou Medical University, Yantai, Shandong Province, China.
| | - Yana Li
- Department of Pathophysiology, School of Basic Medicine, Binzhou Medical University, Yantai, Shandong Province, China.
| |
Collapse
|
9
|
Wang Z, Shui K, Zhang Z, Chen Y, Yang N, Ji S, Shen P, Tian Q. Discovery of a novel binding pocket in PPARγ for partial agonists: structure-based virtual screening identifies ginsenoside Rg5 as a partial agonist promoting beige adipogenesis. Front Chem 2025; 13:1579445. [PMID: 40405893 PMCID: PMC12095147 DOI: 10.3389/fchem.2025.1579445] [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: 02/19/2025] [Accepted: 04/22/2025] [Indexed: 05/26/2025] Open
Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ) is a key target for metabolic disorders that contribute to obesity and type 2 diabetes mellitus (T2DM). However, full agonists such as thiazolidinediones (TZDs) have limitations in terms of side effects. Selective PPARγ modulators (SPPARγMs) that target alternative binding pockets offer the potential for safer partial agonists. Here, we employed six computational algorithms (Fpocket, DeepSite, CavityPlus, DoGSiteScorer, CASTpFold, POCASA) to identify a novel allosteric pocket (pocket 6-5) in the PPARγ ligand-binding domain (LBD), localized at the helix 3 (H3), helix 2 (H2), helix 2'(H2'), and β-sheet interface. A virtual screening of 4,097 natural compounds from traditional Chinese medicine (TCM) libraries was conducted, which led to the identification of ginsenoside Rg5 (TWSZ-5) as a top hit. Molecular docking and molecular dynamics (MD) dynamics revealed TWSZ-5 stabilizes pocket 6-5 through hydrogen bonds with Ser342, Gln345, Lys261, and Lys263. TWSZ-5 promoted beige adipocyte differentiation in adipose-derived stem cells (ADSCs) in vitro, upregulating Ucp1, Prdm16, Cpt1α, and Pgc1α. The present study identifies TWSZ-5 as a novel SPPARγM that utilizes an allosteric binding pocket to enhance thermogenesis while mitigating adverse effects. These findings emphasize the potential of TCM derivatives and structure-based screening strategies to develop safer antidiabetic therapies with precision pharmacology.
Collapse
Affiliation(s)
- Zhen Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Urology, The Affiliated Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing, China
| | - Kexin Shui
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Urology, The Affiliated Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing, China
| | - Zehui Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Urology, The Affiliated Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing, China
| | - Yihan Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Urology, The Affiliated Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing, China
| | - Nanfei Yang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Urology, The Affiliated Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing, China
| | - Shiliang Ji
- Suzhou Research Center of Medical School, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, China
| | - Pingping Shen
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Urology, The Affiliated Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing, China
| | - Qiang Tian
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Urology, The Affiliated Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, School of Life Sciences, Nanjing University, Nanjing, China
- Department of Colorectal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
10
|
Chu Y, Yang S, Chen X. Fibroblast growth factor receptor signaling in metabolic dysfunction-associated fatty liver disease: Pathogenesis and therapeutic targets. Pharmacol Ther 2025; 269:108844. [PMID: 40113178 DOI: 10.1016/j.pharmthera.2025.108844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 02/08/2025] [Accepted: 02/20/2025] [Indexed: 03/22/2025]
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) has emerged as a significant hepatic manifestation of metabolic syndrome, with its prevalence increasing globally alongside the epidemics of obesity and diabetes. MAFLD represents a continuum of liver damage, spanning from uncomplicated steatosis to metabolic dysfunction-associated steatohepatitis (MASH). This condition can advance to more severe outcomes, including fibrosis and cirrhosis. Fibroblast growth factor receptors (FGFRs) are a family of four receptor tyrosine kinases (FGFR1-4) that interact with both paracrine and endocrine fibroblast growth factors (FGFs). This interaction activates the phosphorylation of tyrosine kinase residues, thereby triggering downstream signaling pathways, including RAS-MAPK, JAK-STAT, PI3K-AKT, and PLCγ. In the context of MAFLD, paracrine FGF-FGFR signaling is predominantly biased toward the development of liver fibrosis and carcinogenesis. In contrast, endocrine FGF-FGFR signaling is primarily biased toward regulating the metabolism of bile acids, carbohydrates, lipids, and phosphate, as well as maintaining the overall balance of energy metabolism in the body. The interplay between these biased signaling pathways significantly influences the progression of MAFLD. This review explores the critical functions of FGFR signaling in MAFLD from three perspectives: first, it examines the primary roles of FGFRs relative to their structure; second, it summarizes FGFR signaling in hepatic lipid metabolism, elucidating mechanisms underlying the occurrence and progression of MAFLD; finally, it highlights recent advancements in drug development aimed at targeting FGFR signaling for the treatment of MAFLD and its associated diseases.
Collapse
Affiliation(s)
- Yi Chu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology & College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Su Yang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology & College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Xiaodong Chen
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology & College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
11
|
Xiong Y, Knoedler S, Alfertshofer M, Kim BS, Jiang D, Liu G, Rinkevich Y, Mi B. Mechanisms and therapeutic opportunities in metabolic aberrations of diabetic wounds: a narrative review. Cell Death Dis 2025; 16:341. [PMID: 40280905 PMCID: PMC12032273 DOI: 10.1038/s41419-025-07583-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 01/28/2025] [Accepted: 03/21/2025] [Indexed: 04/29/2025]
Abstract
Metabolic aberrations are fundamental to the complex pathophysiology and challenges associated with diabetic wound healing. These alterations, induced by the diabetic environment, trigger a cascade of events that disrupt the normal wound-healing process. Key factors in this metabolic alternation include chronic hyperglycemia, insulin resistance, and dysregulated lipid and amino acid metabolism. In this review, we summarize the underlying mechanisms driving these metabolic changes in diabetic wounds, while emphasizing the broad implications of these disturbances. Additionally, we discuss therapeutic approaches that target these metabolic anomalies and how their integration with existing wound-healing treatments may yield synergistic effects, offering promising avenues for innovative therapies.
Collapse
Affiliation(s)
- Yuan Xiong
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Samuel Knoedler
- Division of Plastic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02152, USA
- Institute of Regenerative Biology and Medicine, Helmholtz Zentrum München, 81377, Munich, Germany
| | - Michael Alfertshofer
- Department of Hand, Plastic and Aesthetic Surgery, Ludwig-Maximilians-University Munich, 80336, Munich, Germany
| | - Bong-Sung Kim
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, Raemistrasse 100, 8091, Zurich, Switzerland
| | - Dongsheng Jiang
- Precision Research Centre for Refractory Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, China.
| | - Guohui Liu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Yuval Rinkevich
- Institute of Regenerative Biology and Medicine, Helmholtz Zentrum München, 81377, Munich, Germany.
| | - Bobin Mi
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
12
|
Yu W, Haoyu Y, Ling Z, Xing H, Pengfei X, Anzhu W, Lili Z, Linhua Z. Targeting lipid metabolic reprogramming to alleviate diabetic kidney disease: molecular insights and therapeutic strategies. Front Immunol 2025; 16:1549484. [PMID: 40352935 PMCID: PMC12061959 DOI: 10.3389/fimmu.2025.1549484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 03/14/2025] [Indexed: 05/14/2025] Open
Abstract
Diabetic kidney disease (DKD) is one of the major complications of diabetes, and its pathological progression is closely associated with lipid metabolic reprogramming. Under diabetic conditions, renal cells undergo significant lipid metabolic abnormalities, including increased lipid uptake, impaired fatty acid oxidation, disrupted cholesterol efflux, and enhanced lipid catabolism, as adaptive responses to metabolic stress. These changes result in the accumulation of lipids such as free fatty acids, diacylglycerol, and ceramides, leading to lipotoxicity that triggers inflammation and fibrosis. Hypoxia in the DKD microenvironment suppresses fatty acid oxidation and promotes lipid synthesis through the HIF-1α pathway, while chronic inflammation exacerbates lipid metabolic disturbances via inflammatory cytokines, inflammasomes, and macrophage polarization. Targeting lipid metabolism represents a promising therapeutic strategy for alleviating DKD; however, further clinical translational studies are warranted to validate the efficacy and safety of these approaches.
Collapse
Affiliation(s)
- Wei Yu
- Institute of Metabolic Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate College, Beijing University of Chinese Medicine, Beijing, China
| | - Yang Haoyu
- Institute of Metabolic Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhou Ling
- Institute of Metabolic Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate College, Beijing University of Chinese Medicine, Beijing, China
| | - Hang Xing
- Institute of Metabolic Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate College, Beijing University of Chinese Medicine, Beijing, China
| | - Xie Pengfei
- Institute of Metabolic Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate College, Beijing University of Chinese Medicine, Beijing, China
| | - Wang Anzhu
- Chinese-Japanese Friendship Hospital, Beijing, China
| | - Zhang Lili
- Institute of Metabolic Diseases, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhao Linhua
- Department of Endocrinology, The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin, China
| |
Collapse
|
13
|
Chen L, Zhang L, Zhao Y, He M, Wu H, Wang J, Chen Z, Zhao Y, Shen F, Zhang X. Impact of DNA methylation on digestive and metabolic gene expression in red pandas (Ailurus fulgens) during the transition from milk to bamboo diet. BMC Genomics 2025; 26:404. [PMID: 40275147 PMCID: PMC12023452 DOI: 10.1186/s12864-025-11606-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: 07/31/2024] [Accepted: 04/15/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND DNA methylation plays a crucial role in species development and environmental adaptation. In mammals, there are significant dietary changes from infancy to adulthood. Notably, the red panda transitions from milk consumption as juveniles to a bamboo-based diet as adults, with significant alterations in food characteristics and nutritional content. However, the regulatory role of DNA methylation in this process remains unclear. In this study, we investigate the regulatory role of DNA methylation on the expression of digestive and metabolic genes in the liver and pancreas during the red panda's dietary transition from suckling stage to adulthood. RESULTS Our findings reveal significant differences in DNA methylation patterns before and after dietary transition, highlighting the specific alterations in the methylation profiles of genes involved in lipid, carbohydrate, and amino acid metabolism. We found that perilipin-4 (PLIN4) is hypomethylated and highly expressed in the liver of adult red pandas, facilitating lipid droplet formation and storage, crucial for adapting to the low-fat content in bamboo. In contrast, genes like lipoprotein lipase (LPL), crucial for lipid breakdown, exhibited hypermethylated with low-expression patterns, reflecting a reduced lipid metabolism capacity in adults. Carbohydrate metabolism-related genes like ADH4 and FAM3C are hypomethylated and highly expressed in adults, enhancing glycogen production and glucose utilization. Genes involved in protein metabolism like CTSZ and GLDC, exhibit hypomethylated with high-expression and hypermethylated with low-expression patterns in the pancreas of adults, respectively, contributing to protein metabolism balance post-weaning. CONCLUSION This study reveals the regulatory role of DNA methylation in the dietary transition of red pandas from milk to bamboo and provides methylation evidence for the molecular regulation of adaptive expression of digestive and metabolic genes in red pandas with specialized diets.
Collapse
Affiliation(s)
- Lei Chen
- Key Laboratory of Bio-resources and Eco-environment, Ministry of Education, College of Life Science, Sichuan University, Chengdu, 610064, China
| | - Liang Zhang
- Sichuan Key Laboratory for Conservation Biology of Endangered Wildlife, Chengdu Research Base of Giant Panda Breeding, 1375 Panda Road, Northern Suburb, Chengdu, 610081, China
| | - Yanni Zhao
- Key Laboratory of Bio-resources and Eco-environment, Ministry of Education, College of Life Science, Sichuan University, Chengdu, 610064, China
| | - Ming He
- China Conservation and Research Center for the Giant Panda, Dujiangyan, 611800, China
| | - Honglin Wu
- China Conservation and Research Center for the Giant Panda, Dujiangyan, 611800, China
| | - Jingheng Wang
- Key Laboratory of Bio-resources and Eco-environment, Ministry of Education, College of Life Science, Sichuan University, Chengdu, 610064, China
| | - Zhoulong Chen
- Key Laboratory of Bio-resources and Eco-environment, Ministry of Education, College of Life Science, Sichuan University, Chengdu, 610064, China
| | - Yongqi Zhao
- Key Laboratory of Bio-resources and Eco-environment, Ministry of Education, College of Life Science, Sichuan University, Chengdu, 610064, China
| | - Fujun Shen
- Sichuan Key Laboratory for Conservation Biology of Endangered Wildlife, Chengdu Research Base of Giant Panda Breeding, 1375 Panda Road, Northern Suburb, Chengdu, 610081, China
| | - Xiuyue Zhang
- Key Laboratory of Bio-resources and Eco-environment, Ministry of Education, College of Life Science, Sichuan University, Chengdu, 610064, China.
| |
Collapse
|
14
|
Yang R, Bian H, Zhou Z, Yang Y, Wang X, Xu J, Zhong W, Zhu L. Underlying mechanisms of metabolic dysfunction-associated steatotic liver disease induced by 2-ethylhexyl diphenyl phosphate and its hydroxylated metabolite in zebrafish (Danio rerio). JOURNAL OF HAZARDOUS MATERIALS 2025; 493:138407. [PMID: 40300519 DOI: 10.1016/j.jhazmat.2025.138407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 04/22/2025] [Accepted: 04/23/2025] [Indexed: 05/01/2025]
Abstract
2-Ethylhexyl diphenyl phosphate (EHDPHP) is ubiquitous in various environmental media and organisms. Due to its susceptibility to biotransformation, its primary product 2-ethyl-5-hydroxyhexyl diphenyl phosphate (5-OH-EHDPHP) is almost at equal level in organisms. However, their hepatotoxicity remains unclear. In this study, adult zebrafish were exposed to 5, 35, or 245 µg/L of EHDPHP for 28 days. Distinct metabolic dysfunction-associated steatotic liver disease (MASLD) was observed in treated zebrafish, indicated by increased hepatic lipid levels (total cholesterol, triglycerides, nonesterified fatty acids, and fat droplets), steatosis (hepatic ballooning), and inflammation (tnf-α and il-6). Combined the in vitro hepatic cell test, molecular docking and molecular dynamics simulation, it was revealed that peroxisome proliferator-activated receptor gamma (pparγ) was upregulated upon EHDPHP exposure, thereby facilitating lipid synthesis and hepatic lipid accumulation. Notably, its main metabolite 5-OH-EHDPHP induced stronger hepatocyte toxicity and PPARγ transcription. Additionally, serious liver function damage was observed, with aspartate aminotransferase, alanine transaminase, albumin, and γ-glutamyl transferase levels markedly disrupted. This increases the risk of development of cardiovascular disease, hepatic cirrhosis or other chronic conditions. Collectively, the results demonstrate that EHDPHP may cause strong hepatic toxicities, which may be pounded by its hydroxylated metabolites.
Collapse
Affiliation(s)
- Rongyan Yang
- Key Laboratory of Pollution Processes and Environmental Criteria of Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering of Nankai University, Tianjin 300350, China
| | - Hanfei Bian
- Key Laboratory of Pollution Processes and Environmental Criteria of Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering of Nankai University, Tianjin 300350, China
| | - Zhou Zhou
- Key Laboratory of Pollution Processes and Environmental Criteria of Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering of Nankai University, Tianjin 300350, China
| | - Yi Yang
- Key Laboratory of Pollution Processes and Environmental Criteria of Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering of Nankai University, Tianjin 300350, China
| | - Xiao Wang
- Key Laboratory of Pollution Processes and Environmental Criteria of Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering of Nankai University, Tianjin 300350, China
| | - Jingshu Xu
- Key Laboratory of Pollution Processes and Environmental Criteria of Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering of Nankai University, Tianjin 300350, China
| | - Wenjue Zhong
- Key Laboratory of Pollution Processes and Environmental Criteria of Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering of Nankai University, Tianjin 300350, China.
| | - Lingyan Zhu
- Key Laboratory of Pollution Processes and Environmental Criteria of Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering of Nankai University, Tianjin 300350, China.
| |
Collapse
|
15
|
Bao X, San M, Wang S, Zhuo Y, Liu Z, Zheng Y, Li D. Oxytocin receptor enhances adipocyte browning and energy metabolism in mice. Exp Cell Res 2025; 447:114534. [PMID: 40122503 DOI: 10.1016/j.yexcr.2025.114534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 02/14/2025] [Accepted: 03/20/2025] [Indexed: 03/25/2025]
Abstract
Obesity is characterized by abnormal adipose tissue development and disrupted energy metabolism, involving multiple factors. Oxytocin receptor (OXTR) influences social behaviors, mammary gland development and reproduction. In this study, a transgenic mouse model with universal OXTR overexpression under the β-actin promoter (++Oxtr) was employed. Both ++Oxtr males and females exhibited a lean phenotype with reduced fat accumulation, despite unchanged food consumption. OXTR overexpression enhanced energy expenditure, adaptive thermogenesis and glucose tolerance. Morphologically, OXTR overexpression induced adipose tissue browning, marked by increased cell density and smaller adipocytes. Gene expression analysis revealed elevated levels of Brown Adipose Tissue (BAT) markers, fatty acid transport proteins and glucose transporters in adipose tissues. High OXTR ameliorated high-fat diet (HFD)-induced obesity with improvement of metabolic parameters. Mechanistically, OXTR overexpression led to an activation of PPAR signaling, increased energy expenditure, reduced fat deposition and promoted weight loss. These findings identify OXTR as a critical regulator of energy metabolism and thermogenesis. The ability of OXTR to enhance adaptive thermogenesis and energy metabolism suggests it may serve as a novel therapeutic target for metabolic disorders.
Collapse
Affiliation(s)
- Xinyue Bao
- Department of Basic Medicine, Shenyang Medical College, Shenyang, Liaoning, 110034, China
| | - Mingjun San
- Transgenic Research Center, Northeast Normal University, Changchun, Jilin, 130024, China
| | - Shuilian Wang
- Department of Basic Medicine, Shenyang Medical College, Shenyang, Liaoning, 110034, China
| | - Yanli Zhuo
- Room of drug inspection (II), Shenyang Institute for Food and Drug Control, Shenyang, Liaoning, 110000, China
| | - Ziying Liu
- Department of Basic Medicine, Shenyang Medical College, Shenyang, Liaoning, 110034, China
| | - Yaowu Zheng
- Transgenic Research Center, Northeast Normal University, Changchun, Jilin, 130024, China.
| | - Dan Li
- Department of Basic Medicine, Shenyang Medical College, Shenyang, Liaoning, 110034, China.
| |
Collapse
|
16
|
Wang D, Su T, Zhan M, Luo S, Tan H, Lin J, Lai X. Correlation of perfluoroalkyl and polyfluoroalkyl substance levels during pregnancy with gestational diabetes mellitus: a systematic review and meta-analysis. BMC Pregnancy Childbirth 2025; 25:448. [PMID: 40229775 PMCID: PMC11998447 DOI: 10.1186/s12884-025-07551-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 03/31/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND Perfluoroalkyl and polyfluoroalkyl substances (PFAS) are a major class of contaminants in recent years. Pregnant women are more susceptible to the influence of these compounds, which could heighten the risk of developing gestational diabetes mellitus (GDM). This study aims to conduct an updated systematic review and meta-analysis to determine the correlation between PFAS exposure during pregnancy and the risk of developing GDM and delve into their dose-response relationship. METHODS Pubmed, EMBASE, Web of Science, and Cochrane Library databases were searched. Data were statistically analyzed using Stata 15.0. Fixed-effects (FEM) or random-effects (REM) models were used to combine STD mean difference (SMD) or odds ratio (OR) and 95% confidence intervals (CIs) according to heterogeneity. Dose-response meta-analyses were performed when applicable. RESULTS A total of 12 papers were included in this study. Meta-analysis results indicated significantly higher levels of PFOA, PFBS, and PFUnDA in GDM patients compared to healthy pregnant women. Pregnant women exposed to high levels of PFOA and PFBS had a significantly increased risk of developing GDM, with ORs of 1.513 and 1.436, respectively. Dose-response analyses indicated that for each 1 ng/ml increase in PFOA and PFBS exposure, the risk of GDM increased by 0.3% and 11.7%, respectively. In contrast, no significant associations were observed between high exposure to other PFAS compounds, such as PFNA, PFHxS, and PFOS, and the development of GDM. Subgroup analyses suggested that PFOA, PFBS, and PFOS levels were higher in GDM patients from China compared to those from Western countries. The differences in PFOA and PFOS levels between GDM and normal pregnant women were more pronounced during late pregnancy. CONCLUSION Exposure to PFOA, PFBS, and PFUnDA during pregnancy is associated with an increased risk of GDM. Given the elevated risk, particularly in the Chinese population, it is crucial to reduce exposure to these substances, especially from the preconception period onward.
Collapse
Affiliation(s)
- Dongying Wang
- Department of Traditional Chinese Medicine, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510000, China
| | - Ting Su
- Department of Traditional Chinese Medicine, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510000, China
- First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Meiqi Zhan
- Department of Traditional Chinese Medicine, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510000, China
| | - Sining Luo
- First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Hongyu Tan
- Department of Traditional Chinese Medicine, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510000, China
| | - Jinglin Lin
- Department of Traditional Chinese Medicine, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China.
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510000, China.
- , No. 26, Erheng Road, Yuancun, Tianhe District, Guangzhou, Guangdong, China.
| | - Xin Lai
- Department of Traditional Chinese Medicine, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655, China.
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510000, China.
- , No. 26, Erheng Road, Yuancun, Tianhe District, Guangzhou, Guangdong, China.
| |
Collapse
|
17
|
Luong TVT, Yang S, Kim J. Lipotoxicity as a therapeutic target in the type 2 diabetic heart. J Mol Cell Cardiol 2025; 201:105-121. [PMID: 40020774 DOI: 10.1016/j.yjmcc.2025.02.010] [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: 08/28/2024] [Revised: 01/07/2025] [Accepted: 02/24/2025] [Indexed: 03/03/2025]
Abstract
Cardiac lipotoxicity, characterized by excessive lipid accumulation in the cardiac tissue, is a critical contributor to the pathogenesis of diabetic heart. Recent research has highlighted the key mechanisms underlying lipotoxicity, including mitochondrial dysfunction, endoplasmic reticulum stress, inflammation, and cell apoptosis, which ultimately impair the cardiac function. Various therapeutic interventions have been developed to target these pathways, mitigate lipotoxicity, and improve cardiovascular outcomes in diabetic patients. Given the global escalation in the prevalence of diabetes and the urgent demand for effective therapeutic approaches, this review focuses on how targeting cardiac lipotoxicity may be a promising avenue for treating diabetes.
Collapse
Affiliation(s)
- Trang Van T Luong
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Seonbu Yang
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Jaetaek Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Chung-Ang University, Seoul, Republic of Korea.
| |
Collapse
|
18
|
Zhang QR, Dong Y, Fan JG. Early-life exposure to gestational diabetes mellitus predisposes offspring to pediatric nonalcoholic fatty liver disease. Hepatobiliary Pancreat Dis Int 2025; 24:128-137. [PMID: 38195352 DOI: 10.1016/j.hbpd.2023.12.007] [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: 08/05/2023] [Accepted: 12/28/2023] [Indexed: 01/11/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD) has emerged as the prevailing chronic liver disease in the pediatric population due to the global obesity pandemic. Evidence shows that prenatal and postnatal exposure to maternal abnormalities leads to a higher risk of pediatric NAFLD through persistent alterations in developmental programming. Gestational diabetes mellitus (GDM) is a hyperglycemic syndrome which has become the most prevalent complication in pregnant women. An increasing number of both epidemiologic investigations and animal model studies have validated adverse and long-term outcomes in offspring following GDM exposure in utero. Similarly, GDM is considered a crucial risk factor for pediatric NAFLD. This review aimed to summarize currently published studies concerning the inductive roles of GDM in offspring NAFLD development during childhood and adolescence. Dysregulations in hepatic lipid metabolism and gut microbiota in offspring, as well as dysfunctions in the placenta are potential factors in the pathogenesis of GDM-associated pediatric NAFLD. In addition, potentially effective interventions for GDM-associated offspring NAFLD are also discussed in this review. However, most of these therapeutic approaches still require further clinical research for validation.
Collapse
Affiliation(s)
- Qian-Ren Zhang
- Center for Fatty Liver, Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Yan Dong
- Department of Endocrinology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Shanghai Institute for Pediatric Research, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Jian-Gao Fan
- Center for Fatty Liver, Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Shanghai Key Lab of Pediatric Gastroenterology and Nutrition, Shanghai 200092, China.
| |
Collapse
|
19
|
Yang Y, Jiao L, Huang Y, Shang H, Li E, Chang H, Cui H, Wan Y. Evaluation of FXR Activity in Pollutants Identified in Sewage Sludge and Subsequent in Vitro and in Vivo Characterization of Metabolic Effects of Triphenyl Phosphate. ENVIRONMENTAL HEALTH PERSPECTIVES 2025; 133:47005. [PMID: 40048564 PMCID: PMC12010937 DOI: 10.1289/ehp15435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 01/03/2025] [Accepted: 01/27/2025] [Indexed: 04/15/2025]
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most common liver disease worldwide, and increasing evidence suggests that exposure to environmental pollutants is associated with the increased incidence of MASLD. The farnesoid X receptor (FXR) plays an important role in the development of MASLD by regulating bile acids (BAs) and lipid metabolism. However, whether FXR-active pollutants are the environmental drivers of MASLD remains unclear. OBJECTIVES This study aimed to determine whether FXR-active pollutants exist in the environment and evaluate their ability to trigger MASLD development in mice. METHODS An FXR protein affinity pull-down assay and nontargeted mass spectrometry (MS) analysis were used to identify environmental FXR ligands in sewage sludge. A homogeneous time-resolved fluorescence coactivator recruitment assay and cell-based dual-luciferase reporter assay were used to determine the FXR activities of the identified pollutants. Targeted analysis of BAs, MS imaging, lipidomic analysis, 16S rRNA sequencing, and quantitative polymerase chain reaction were conducted to assess the ability of FXR-active pollutants to induce metabolic disorders of BAs and lipids and to contribute to MASLD development in C57BL/6N mice. RESULTS We identified 19 compounds in the sewage sludge that had FXR-antagonistic activity, and triphenyl phosphate (TPHP) was the FXR antagonist with the highest efficacy. Mice exposed to either 10 or 50 mg / kg TPHP for 30 d had higher levels of conjugated primary BAs in enterohepatic circulation, and the BA pool showed FXR antagonistic activities. The exposed mice also had greater lipogenesis (more Oil Red O staining and high triglyceride levels) in liver. CONCLUSIONS Nineteen FXR-antagonistic pollutants were identified in sewage sludge. FXR inhibition by the strongest antagonist TPHP may have a role in promoting MASLD development in mice by inducing a positive feedback loop between the FXR and BAs. https://doi.org/10.1289/EHP15435.
Collapse
Affiliation(s)
- Yi Yang
- Laboratory for Earth Surface Processes, College of Urban and Environmental Sciences, Peking University, Beijing, China
| | - Ling Jiao
- Laboratory for Earth Surface Processes, College of Urban and Environmental Sciences, Peking University, Beijing, China
| | - Yixuan Huang
- Laboratory for Earth Surface Processes, College of Urban and Environmental Sciences, Peking University, Beijing, China
| | - Hailin Shang
- Laboratory for Earth Surface Processes, College of Urban and Environmental Sciences, Peking University, Beijing, China
| | - Enrui Li
- College of Environmental Science and Engineering, Beijing Forestry University, Beijing, China
| | - Hong Chang
- College of Environmental Science and Engineering, Beijing Forestry University, Beijing, China
| | - Hongyang Cui
- Laboratory for Earth Surface Processes, College of Urban and Environmental Sciences, Peking University, Beijing, China
| | - Yi Wan
- Laboratory for Earth Surface Processes, College of Urban and Environmental Sciences, Peking University, Beijing, China
| |
Collapse
|
20
|
Tani-Ichi S, Abe S, Miyachi H, Kitano S, Shimba A, Ejima A, Hara T, Cui G, Kado T, Hori S, Tobe K, Ikuta K. IL-7Rα signaling in regulatory T cells of adipose tissue is essential for systemic glucose homeostasis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025; 214:666-679. [PMID: 40107286 DOI: 10.1093/jimmun/vkae064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 12/17/2024] [Indexed: 03/22/2025]
Abstract
Regulatory T cells (Tregs) mediate tissue homeostasis and repair. The function of the interleukin-7 receptor α (IL-7Rα) in nonlymphoid tissue Tregs is still unknown, although low expression of IL-7Rα is a widely accepted marker for Tregs. Here, we show that IL-33R (ST2)-expressing Tregs in the visceral adipose tissue (VAT) express the IL-7Rα at high levels. Treg-specific IL-7Rα-deficient mice exhibited reduced adipose ST2+ Tregs and impaired glucose tolerance, whereas IL-7Rα was dispensable for Tregs in lymphoid tissues. Mice deficient in thymic stromal lymphopoietin (TSLP), an additional ligand for IL-7Rα, displayed a modest decrease in adipose ST2+ Tregs and a reduced accumulation of adipose eosinophils, accompanied by slightly impaired glucose tolerance. In the VAT, mesothelial cells expressed IL-7, whereas adipose stem cells and folate receptor β-expressing tissue-resident macrophages expressed TSLP. Thus, this study indicates the significance of IL-7Rα signaling in the maintenance of VAT Tregs and glucose homeostasis, revealing a novel role for IL-7 and TSLP in immunometabolism.
Collapse
Affiliation(s)
- Shizue Tani-Ichi
- Laboratory of Immune Regulation, Department of Virus Research, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
- Department of Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shinya Abe
- Laboratory of Immune Regulation, Department of Virus Research, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Hitoshi Miyachi
- Reproductive Engineering Team, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Satsuki Kitano
- Reproductive Engineering Team, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Akihiro Shimba
- Laboratory of Immune Regulation, Department of Virus Research, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
- Department of Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Aki Ejima
- Laboratory of Immune Regulation, Department of Virus Research, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Takahiro Hara
- Laboratory of Immune Regulation, Department of Virus Research, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Guangwei Cui
- Laboratory of Immune Regulation, Department of Virus Research, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Tomonobu Kado
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Shohei Hori
- Laboratory of Immunology and Microbiology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Kazuyuki Tobe
- Research Center for Pre-Disease Science, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Koichi Ikuta
- Laboratory of Immune Regulation, Department of Virus Research, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| |
Collapse
|
21
|
Zhang Y, Yang J, Min J, Huang S, Li Y, Liu S. The emerging role of E3 ubiquitin ligases and deubiquitinases in metabolic dysfunction-associated steatotic liver disease. J Transl Med 2025; 23:368. [PMID: 40133964 PMCID: PMC11938720 DOI: 10.1186/s12967-025-06255-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 02/17/2025] [Indexed: 03/27/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most common chronic liver disease worldwide, with a prevalence as high as 32.4%. MASLD encompasses a spectrum of liver pathologies, ranging from steatosis to metabolic dysfunction-associated steatohepatitis (MASH), fibrosis, and, in some cases, progression to end-stage liver disease (cirrhosis and hepatocellular carcinoma). A comprehensive understanding of the pathogenesis of this highly prevalent liver disease may facilitate the identification of novel targets for the development of improved therapies. E3 ubiquitin ligases and deubiquitinases (DUBs) are key regulatory components of the ubiquitin‒proteasome system (UPS), which plays a pivotal role in maintaining intracellular protein homeostasis. Emerging evidence implicates that aberrant expression of E3 ligases and DUBs is involved in the progression of MASLD. Here, we review abnormalities in E3 ligases and DUBs by (1) discussing their targets, mechanisms, and functions in MASLD; (2) summarizing pharmacological interventions targeting these enzymes in preclinical and clinical studies; and (3) addressing challenges and future therapeutic strategies. This review synthesizes current evidence to highlight the development of novel therapeutic strategies based on the UPS for MASLD and progressive liver disease.
Collapse
Affiliation(s)
- Yu Zhang
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, CSU-Sinocare Research Center for Nutrition and Metabolic Health, Furong Laboratory, Changsha, Hunan, 410011, China
| | - Jiahui Yang
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, CSU-Sinocare Research Center for Nutrition and Metabolic Health, Furong Laboratory, Changsha, Hunan, 410011, China
| | - Jiali Min
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, CSU-Sinocare Research Center for Nutrition and Metabolic Health, Furong Laboratory, Changsha, Hunan, 410011, China
| | - Shan Huang
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, CSU-Sinocare Research Center for Nutrition and Metabolic Health, Furong Laboratory, Changsha, Hunan, 410011, China
| | - Yuchen Li
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, CSU-Sinocare Research Center for Nutrition and Metabolic Health, Furong Laboratory, Changsha, Hunan, 410011, China
| | - Shanshan Liu
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, CSU-Sinocare Research Center for Nutrition and Metabolic Health, Furong Laboratory, Changsha, Hunan, 410011, China.
| |
Collapse
|
22
|
Lai S, Tang D, Feng J. Mitochondrial targeted therapies in MAFLD. Biochem Biophys Res Commun 2025; 753:151498. [PMID: 39986088 DOI: 10.1016/j.bbrc.2025.151498] [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: 11/26/2024] [Revised: 01/24/2025] [Accepted: 02/15/2025] [Indexed: 02/24/2025]
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) is a clinical-pathological syndrome primarily characterized by excessive accumulation of fat in hepatocytes, independent of alcohol consumption and other well-established hepatotoxic agents. Mitochondrial dysfunction is widely acknowledged as a pivotal factor in the pathogenesis of various diseases, including cardiovascular diseases, cancer, neurodegenerative disorders, and metabolic diseases such as obesity and obesity-associated MAFLD. Mitochondria are dynamic cellular organelles capable of modifying their functions and structures to accommodate the metabolic demands of cells. In the context of MAFLD, the excess production of reactive oxygen species induces oxidative stress, leading to mitochondrial dysfunction, which subsequently promotes metabolic disorders, fat accumulation, and the infiltration of inflammatory cells in liver and adipose tissue. This review aims to systematically analyze the role of mitochondria-targeted therapies in MAFLD, evaluate current therapeutic strategies, and explore future directions in this rapidly evolving field. We specifically focus on the molecular mechanisms underlying mitochondrial dysfunction, emerging therapeutic approaches, and their clinical implications. This is of significant importance for the development of new therapeutic approaches for these metabolic disorders.
Collapse
Affiliation(s)
- Sien Lai
- Guangdong Provincial Engineering and Technology Research Center for Gene Editing, School of Medicine, Foshan University, 528000, Foshan, China.
| | - Dongsheng Tang
- Guangdong Provincial Engineering and Technology Research Center for Gene Editing, School of Medicine, Foshan University, 528000, Foshan, China.
| | - Juan Feng
- Guangdong Provincial Engineering and Technology Research Center for Gene Editing, School of Medicine, Foshan University, 528000, Foshan, China.
| |
Collapse
|
23
|
Leszczynska A, Alle T, Kaufmann B, Sung H, Stoess C, Reca A, Kim A, Kim S, Tran C, Oukoloff K, Monti L, Lucero B, Gertsman I, Momper JD, Hartmann P, Feldstein AE, Dohil R, Ballatore C. d 4-Cystamine: A Deuterated Cystamine Derivative with Improved Anti-Inflammatory and Anti-Fibrotic Activities in a Murine Model of Fibrosing Steatohepatitis. ACS Pharmacol Transl Sci 2025; 8:885-898. [PMID: 40109735 PMCID: PMC11915185 DOI: 10.1021/acsptsci.4c00738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/27/2025] [Accepted: 02/03/2025] [Indexed: 03/22/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a multifactorial chronic disease that can progress to metabolic dysfunction-associated steatohepatitis (MASH) and liver fibrosis, ultimately leading to liver cirrhosis and hepatocellular carcinoma. Oxidative stress is believed to play an important role in the development of MASH. Small aminothiol compounds such as cysteamine and its oxidized precursor, cystamine, are known pleiotropic compounds that exhibit relatively potent antioxidant and other effects. Herein, we evaluate the efficacy of cystamine, as well as two deuterated derivatives, in a choline-deficient, L-amino acid-defined, high-fat-diet (CDAA-HFD) mouse model of rapidly progressing liver fibrosis. Compared to control mice, daily oral administration of isotopically reinforced cystamine derivatives (200 mg/kg) led to a significant reduction of liver fibrosis and inflammation as well as oxidative stress. Moreover, the efficacy of treatment appeared to increase with the deuteration state of cystamine, with the tetradeuterated derivative, d 4 -cystamine, being the most effective. These results indicate that deuterated cystamine derivatives hold promise as potential candidates for the treatment of MASH.
Collapse
Affiliation(s)
- Aleksandra Leszczynska
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Thibault Alle
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Benedikt Kaufmann
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Hana Sung
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Christian Stoess
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Agustina Reca
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Andrea Kim
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Sun Kim
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Chelsea Tran
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Killian Oukoloff
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Ludovica Monti
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Bobby Lucero
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Ilya Gertsman
- Clarus Analytical LLC, 8545 Arjons Dr. Suite A, San Diego, California 92126, United States
| | - Jeremiah D Momper
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Phillipp Hartmann
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Ariel E Feldstein
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
- Global Drug Discovery, Novo Nordisk, Copenhagen DK-2880, Denmark
| | - Ranjan Dohil
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Carlo Ballatore
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| |
Collapse
|
24
|
Dai J, Zhao Y, Chen Y, Jiang Y, Sun R, Tang X, Cui Y, Mao H, Peng XG. Irisin reverses high-fat diet-induced metabolic dysfunction via activation of brown adipose tissue in mice. Int J Obes (Lond) 2025:10.1038/s41366-025-01739-z. [PMID: 40082597 DOI: 10.1038/s41366-025-01739-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 02/01/2025] [Accepted: 02/25/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND High-fat diet (HFD) induces negative effects on the activity of interscapular brown adipose tissue (iBAT) and systemic energy metabolism. Irisin, a small hormonal agent known to modulate metabolism has been used for intervening HFD-induced obesity. However, its mechanism of action on iBAT function remains to be fully elucidated. This study sought to investigate whether irisin intervention could restore the thermogenic function of iBAT in mice with HFD-induced obesity, thereby regulating systemic metabolism. METHODS Magnetic resonance imaging (MRI) and positron emission tomography/computed tomography (PET/CT) were used to monitor changes of thermogenic capacity of iBAT and systemic metabolism in mice with HFD-induced obesity and iBAT deficiency during 2-week or 4-week irisin intervention. Pathological and molecular biology analyses were performed on tissue and blood samples. RESULTS Prolonged HFD feeding in mice induced obesity and impaired the thermogenic capacity of iBAT. MRI results showed that irisin intervention for 4-week reduced lipid content in iBAT, increased uncoupling protein 1 (UCP 1) expression and enhanced glucose analogue uptake capacity. These improvements of functions in iBAT activity were accompanied by an improvement in systemic metabolism. The positive effects of irisin appears to be dependent on the length of intervention time. When iBAT was removed, the beneficial effects of irisin were partially suppressed, suggesting that irisin regulates metabolism through the restoration of the thermogenic function of iBAT. CONCLUSIONS HFD results in reduced thermogenic capacity of iBAT, while irisin intervention can effectively restore iBAT function, leading to improvement in overall glucose and lipid metabolism.
Collapse
Affiliation(s)
- Jingyue Dai
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Yufei Zhao
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Yue Chen
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Yang Jiang
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Rui Sun
- Department of Pharmacy, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, 215002, China
| | - Xingzhe Tang
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Ying Cui
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Hui Mao
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA, 30322, USA
| | - Xin-Gui Peng
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China.
- Department of Radiology, Nanjing Lishui People's Hospital, Zhongda Hospital Lishui Branch, Southeast University, Nanjing, 211200, China.
| |
Collapse
|
25
|
Jiang Q, Zhang D, Hu N, Ma Y, Jin H, Zhao Y, Zhang M, Li B, Huang Z, Yuan B, Zhu Y, Tian J, Miao X. Environmental Chemical-Induced Cardiometabolic Disorders: Combined Epidemiological and Experimental Evidence. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2025; 59:3853-3868. [PMID: 39977603 DOI: 10.1021/acs.est.4c09728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
Screening environmental pollutants that are harmful to the cardiometabolic status and understanding their key toxic pathways are crucial for effective clinical intervention. Based on exposure data of 46 chemicals in a nationally representative 13,286 people, logistic regression and mixture modeling were used to preliminarily identify environmental pollutants with significant impacts on 12 indicators for cardiometabolic disorders. A total of 15 chemicals were found to be associated with the integrated latent class, among which four chemicals (perfluorononanoic acid [PFNA], perfluorooctanoic acid [PFOA], thiocyanate, and thallium) also contributed significantly to the mixture effect. We constructed the adverse outcome pathways (AOPs) for nine significant toxicants in both models of individual chemicals and the mixture for each cardiometabolic disorder. Notably, fluoroalkyl substances affect multiple aspects of hyperlipidemia by activating PPARα. We performed molecular docking and in vitro experiments to verify and supplement the toxicological mechanism of PFNA. Through binding to PPARα, PFNA increased the levels of downstream molecules including CD36 (fatty acid transfer), ACSL1 (fatty acid activation), and CPT1a (intracellular transfer for β-oxidation) and ultimately promoted the accumulation of triglycerides and lipid droplets in HepG2 cells. These markers, together with key events for other metabolic phenotypes, may be potential targets for scientific research or clinical treatment.
Collapse
Affiliation(s)
- Qi Jiang
- Department of Epidemiology and Biostatistics, School of Public Health, Research Center of Public Health, Renmin Hospital of Wuhan University, Department of Gastrointestinal Oncology, Zhongnan Hospital of Wuhan University, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei 430071, China
| | - Donghui Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Research Center of Public Health, Renmin Hospital of Wuhan University, Department of Gastrointestinal Oncology, Zhongnan Hospital of Wuhan University, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei 430071, China
| | - Naifan Hu
- Department of Epidemiology and Biostatistics, School of Public Health, Research Center of Public Health, Renmin Hospital of Wuhan University, Department of Gastrointestinal Oncology, Zhongnan Hospital of Wuhan University, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei 430071, China
| | - Yunfei Ma
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan 430071, China
| | - Huibin Jin
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan 430071, China
| | - Yunhao Zhao
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan 430071, China
| | - Ming Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan 430071, China
| | - Bin Li
- Department of Epidemiology and Biostatistics, School of Public Health, Wuhan University, Wuhan 430071, China
| | - Zhenzhen Huang
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Wuhan 430071, China
| | - Bifeng Yuan
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Wuhan 430071, China
| | - Ying Zhu
- Department of Epidemiology and Biostatistics, School of Public Health, Research Center of Public Health, Renmin Hospital of Wuhan University, Department of Gastrointestinal Oncology, Zhongnan Hospital of Wuhan University, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei 430071, China
| | - Jianbo Tian
- Department of Epidemiology and Biostatistics, School of Public Health, Research Center of Public Health, Renmin Hospital of Wuhan University, Department of Gastrointestinal Oncology, Zhongnan Hospital of Wuhan University, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei 430071, China
| | - Xiaoping Miao
- Department of Epidemiology and Biostatistics, School of Public Health, Research Center of Public Health, Renmin Hospital of Wuhan University, Department of Gastrointestinal Oncology, Zhongnan Hospital of Wuhan University, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei 430071, China
| |
Collapse
|
26
|
Huang M, Ma Y, Fan Q, Che S, Zhang J, Ding S, Zhu S, Li X. Effects of nanopolystyrene and/or phoxim exposure on digestive function of Eriocheir sinensis. Comp Biochem Physiol C Toxicol Pharmacol 2025; 289:110102. [PMID: 39653099 DOI: 10.1016/j.cbpc.2024.110102] [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: 09/29/2024] [Revised: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 12/12/2024]
Abstract
Nanopolystyrene (NP) and phoxim (PHO) are pervasive environmental contaminants that pose a significant threat to the health of aquatic organisms, prompting widespread concern among researchers and the public alike. The hepatopancreas play important roles in the Chinese mitten crab (Eriocheir sinensis), such as digestion, absorption and detoxification. This study assessed the hepatopancreatic toxicity caused by the exposure of Eriocheir sinensis to environmentally relevant concentrations of NP and/or PHO. After a 21-day exposure period, NP (1.0 × 1010 particles/L) and PHO (24 μg/L) exposure resulted in reduced number of blister-like, resorptive, and fibrillar cells and an elevation in lipid droplets within the hepatopancreas compared to the control group. Furthermore, trypsin and lipase activity decreased, amylase activity increased, and a significantly decrease in the expression of digestion-related genes, including CHT, CarL, and CarB, suggested impairment in both digestive and metabolic functions. The marked upregulation of key genes, including PPARγ, GYK, PEPCK, and SCD, as well as key metabolites such as 4-methylzymosterol-carboxylate, zymosterone, lathosterol, 7-dehydro-desmosterol, vitamin D2, 24-methylene-cycloartanol, 5-dehydroepisterol, and sitosterol in the lipid metabolic pathway, showed that the peroxisome proliferator-activated receptor (PPAR) and steroid biosynthesis signaling pathways were highly affected by exposure to NP and/or PHO. These findings indicated that exposure to NP and/or PHO might adversely affect the hepatopancreatic physiological homeostasis in E. sinensis by causing tissue damage and interfering with digestive and metabolic functions. Our results provide ecotoxicological insights into the effects of nanopolystyrene and/or phoxim exposure on the digestive function of Eriocheir sinensis.
Collapse
Affiliation(s)
- Mengting Huang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Yuan Ma
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Qianru Fan
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Shunli Che
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Jun Zhang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; Engineering Technology Research Center of Healthy Aquaculture, Anhui Agricultural University, Hefei 230036, China
| | - Shuquan Ding
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; Engineering Technology Research Center of Healthy Aquaculture, Anhui Agricultural University, Hefei 230036, China
| | - Shuren Zhu
- Shandong Freshwater Fisheries Research Institute, Jinan 250013, China.
| | - Xilei Li
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; Engineering Technology Research Center of Healthy Aquaculture, Anhui Agricultural University, Hefei 230036, China.
| |
Collapse
|
27
|
Zheng Y, Shao M, Zheng Y, Sun W, Qin S, Sun Z, Zhu L, Guan Y, Wang Q, Wang Y, Li L. PPARs in atherosclerosis: The spatial and temporal features from mechanism to druggable targets. J Adv Res 2025; 69:225-244. [PMID: 38555000 PMCID: PMC11954843 DOI: 10.1016/j.jare.2024.03.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 04/02/2024] Open
Abstract
BACKGROUND Atherosclerosis is a chronic and complex disease caused by lipid disorder, inflammation, and other factors. It is closely related to cardiovascular diseases, the chief cause of death globally. Peroxisome proliferator-activated receptors (PPARs) are valuable anti-atherosclerosis targets that showcase multiple roles at different pathological stages of atherosclerosis and for cell types at different tissue sites. AIM OF REVIEW Considering the spatial and temporal characteristics of the pathological evolution of atherosclerosis, the roles and pharmacological and clinical studies of PPARs were summarized systematically and updated under different pathological stages and in different vascular cells of atherosclerosis. Moreover, selective PPAR modulators and PPAR-pan agonists can exert their synergistic effects meanwhile reducing the side effects, thereby providing novel insight into future drug development for precise spatial-temporal therapeutic strategy of anti-atherosclerosis targeting PPARs. KEY SCIENTIFIC Concepts of Review: Based on the spatial and temporal characteristics of atherosclerosis, we have proposed the importance of stage- and cell type-dependent precision therapy. Initially, PPARs improve endothelial cells' dysfunction by inhibiting inflammation and oxidative stress and then regulate macrophages' lipid metabolism and polarization to improve fatty streak. Finally, PPARs reduce fibrous cap formation by suppressing the proliferation and migration of vascular smooth muscle cells (VSMCs). Therefore, research on the cell type-specific mechanisms of PPARs can provide the foundation for space-time drug treatment. Moreover, pharmacological studies have demonstrated that several drugs or compounds can exert their effects by the activation of PPARs. Selective PPAR modulators (that specifically activate gene subsets of PPARs) can exert tissue and cell-specific effects. Furthermore, the dual- or pan-PPAR agonist could perform a better role in balancing efficacy and side effects. Therefore, research on cells/tissue-specific activation of PPARs and PPAR-pan agonists can provide the basis for precision therapy and drug development of PPARs.
Collapse
Affiliation(s)
- Yi Zheng
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Mingyan Shao
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yanfei Zheng
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Wenlong Sun
- Institute of Biomedical Research, School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255000, China
| | - Si Qin
- Lab of Food Function and Nutrigenomics, College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Ziwei Sun
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Linghui Zhu
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yuanyuan Guan
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Qi Wang
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Yong Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China; First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming 650500, China.
| | - Lingru Li
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing 100029, China.
| |
Collapse
|
28
|
Duan H, Gong M, Yuan G, Wang Z. Sex Hormone: A Potential Target at Treating Female Metabolic Dysfunction-Associated Steatotic Liver Disease? J Clin Exp Hepatol 2025; 15:102459. [PMID: 39722783 PMCID: PMC11667709 DOI: 10.1016/j.jceh.2024.102459] [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: 08/10/2024] [Accepted: 11/13/2024] [Indexed: 12/28/2024] Open
Abstract
The global prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD) is rising due to rapid lifestyle changes. Although females may be less prone to MASLD than males, specific studies on MASLD in females should still be conducted. Previous research has shown that sex hormone levels are strongly linked to MASLD in females. By reviewing a large number of experimental and clinical studies, we summarized the pathophysiological mechanisms of estrogen, androgen, sex hormone-binding globulin, follicle-stimulating hormone, and prolactin involved in the development of MASLD. We also analyzed the role of these hormones in female MASLD patients with polycystic ovarian syndrome or menopause, and explored the potential of targeting sex hormones for the treatment of MASLD. We hope this will provide a reference for further exploration of mechanisms and treatments for female MASLD from the perspective of sex hormones.
Collapse
Affiliation(s)
- Huiyan Duan
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Minmin Gong
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gang Yuan
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhi Wang
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
29
|
Xu X, Shen S, Dong Y, Jiang L, Wang J, Shao Y. Protective effect of puerarin in diabetic nephropathy: A systematic review and meta-analysis of animal studies. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 138:156385. [PMID: 39823801 DOI: 10.1016/j.phymed.2025.156385] [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: 10/14/2024] [Revised: 12/18/2024] [Accepted: 01/10/2025] [Indexed: 01/20/2025]
Abstract
BACKGROUND Puerarin is a crucial constituent separated from the Chinese herbaceous plant, Pueraria lobata (Willd.) Ohwi, which exhibits multiple biological activities. Previous studies have indicated that puerarin has a function to alleviate renal damage in animal models of diabetic nephropathy (DN). However, there is still a dearth of systematic preclinical studies. PURPOSE This study was designed to assess the effectiveness of puerarin on DN through meta-analysis. Furthermore, it aimed to reveal the underlying mechanisms of puerarin's efficacy in treating DN. METHODS The animal studies were retrieved from 5 electronic databases. In total, 26 studies were included in our analysis. STATA 18.0 software was employed to evaluate crucial parameters, including fasting blood glucose (FBG), serum creatinine (SCr), blood urea nitrogen (BUN), proteinuria, oxidative stress, inflammatory responses, and lipid metabolism. RESULTS The results indicate that puerarin significantly improves FBG, SCr, BUN, proteinuria, and kidney index (KI). Additionally, puerarin enhances indicators of oxidative stress including superoxide dismutase (SOD), glutathione (GSH), and catalase (CAT), while reduces inflammatory indicators like interleukin 6 (IL-6), interleukin 1β (IL-1β), and tumor necrosis factor alpha (TNF-α). Moreover, puerarin lowers triglycerides (TG), total cholesterol (TC), and low-density lipoprotein cholesterol (LDL) levels. CONCLUSION In conclusion, puerarin has the effect of improving renal function in DN animals possibly through antioxidative, anti-inflammatory, and the regulation of renal lipid accumulation, which offers preclinical support for its possible therapeutic use in the management of DN.
Collapse
Affiliation(s)
- Xiaoying Xu
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China; Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, China
| | - Siqi Shen
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China; Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, China
| | - Yingying Dong
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, China
| | - Lan Jiang
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310007, China
| | - Jianwei Wang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, China.
| | - Yanfei Shao
- Hangzhou Hospital of Traditional Chinese Medicine (Affiliated Hangzhou Hospital of Traditional Chinese Medicine, Zhejiang Chinese Medical University), Hangzhou 310007, Zhejiang, China; Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, China.
| |
Collapse
|
30
|
Palumbo M, Ugolotti M, Zimetti F, Adorni MP. Anti-atherosclerotic effects of natural compounds targeting lipid metabolism and inflammation: Focus on PPARs, LXRs, and PCSK9. ATHEROSCLEROSIS PLUS 2025; 59:39-53. [PMID: 39877131 PMCID: PMC11773090 DOI: 10.1016/j.athplu.2024.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 12/17/2024] [Accepted: 12/19/2024] [Indexed: 01/31/2025]
Abstract
A large body of evidence has shown that modulation of the nuclear receptors peroxisome proliferator-activated receptors (PPARs), the liver X receptors (LXRs), the proprotein convertase subtilisin/kexin type 9 (PCSK9) and inflammatory processes by natural compounds has hypolipidemic and anti-atherosclerotic effects. These beneficial outcomes are certainly related to the crucial function of these targets in maintaining cholesterol homeostasis and regulating systemic inflammation. Currently, the therapeutic scenario for cardiovascular diseases (CVD) offers a plethora of widely validated and functional pharmacological treatments to improve the health status of patients. However, patients are increasingly sceptical of pharmacological treatments which are often associated with moderate to severe side effects. The aim of our review is to provide a collection of the most recent scientific evidence on the most common phytochemicals, used for centuries in the Mediterranean diet and traditional chinese medicine that act on these key regulators of cholesterol homeostasis and systemic inflammation, which could constitute important tools for CVD management.
Collapse
Affiliation(s)
| | | | | | - Maria Pia Adorni
- Department of Medicine and Surgery, Unit of Neuroscience, University of Parma, Italy
| |
Collapse
|
31
|
Knezović E, Hefer M, Blažanović S, Petrović A, Tomičić V, Srb N, Kirner D, Smolić R, Smolić M. Drug Pipeline for MASLD: What Can Be Learned from the Successful Story of Resmetirom. Curr Issues Mol Biol 2025; 47:154. [PMID: 40136408 PMCID: PMC11941580 DOI: 10.3390/cimb47030154] [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: 02/02/2025] [Revised: 02/18/2025] [Accepted: 02/20/2025] [Indexed: 03/27/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) and its progressive form, metabolic dysfunction-associated steatohepatitis (MASH), represent a growing global health problem linked to obesity, insulin resistance, and dyslipidemia. MASLD often leads to fibrosis, cirrhosis, and hepatocellular carcinoma. Currently, therapeutic options are limited, emphasizing the need for novel, targeted pharmacological interventions. Resmetirom, a selective thyroid hormone receptor beta (THR-β) agonist, offers a promising approach by specifically enhancing hepatic metabolism while minimizing systemic effects. Clinical trials have demonstrated its capacity to reduce hepatic triglyceride accumulation and improve lipid profiles. Early- and advanced-phase studies, including the MAESTRO program, highlight significant reductions in hepatic fat content and favorable impacts on noninvasive biomarkers of fibrosis with minimal side effects. This review highlights evidence from pivotal studies, explores resmetirom's mechanism of action, and compares its efficacy and safety with other emerging therapeutic agents. While resmetirom marks a breakthrough in non-cirrhotic MASH management, further long-term studies are essential to fully evaluate its clinical benefits and potential regulatory approval for broader use in MASLD and MASH.
Collapse
Affiliation(s)
- Elizabeta Knezović
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia; (E.K.); (M.H.); (S.B.); (A.P.); (V.T.); (N.S.); (D.K.); (R.S.)
- Clinical Institute of Translational Medicine, University Hospital Osijek, 31000 Osijek, Croatia
| | - Marija Hefer
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia; (E.K.); (M.H.); (S.B.); (A.P.); (V.T.); (N.S.); (D.K.); (R.S.)
| | - Suzana Blažanović
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia; (E.K.); (M.H.); (S.B.); (A.P.); (V.T.); (N.S.); (D.K.); (R.S.)
| | - Ana Petrović
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia; (E.K.); (M.H.); (S.B.); (A.P.); (V.T.); (N.S.); (D.K.); (R.S.)
| | - Vice Tomičić
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia; (E.K.); (M.H.); (S.B.); (A.P.); (V.T.); (N.S.); (D.K.); (R.S.)
| | - Nika Srb
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia; (E.K.); (M.H.); (S.B.); (A.P.); (V.T.); (N.S.); (D.K.); (R.S.)
| | - Damir Kirner
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia; (E.K.); (M.H.); (S.B.); (A.P.); (V.T.); (N.S.); (D.K.); (R.S.)
| | - Robert Smolić
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia; (E.K.); (M.H.); (S.B.); (A.P.); (V.T.); (N.S.); (D.K.); (R.S.)
| | - Martina Smolić
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia; (E.K.); (M.H.); (S.B.); (A.P.); (V.T.); (N.S.); (D.K.); (R.S.)
| |
Collapse
|
32
|
Bezerra HVA, Ramírez-Zamudio GD, Santana MHDA, Polizel GHG, de Oliveira RÍG, Rissi GP, Frujuelle Filho AR, Poleti MD, Velloso L, da Silva TH, Gallo SB, da Luz E Silva S, Cônsolo NRB, Leme PR. Lamb Performance and Meat Quality: The Impact of Chromium in High-Concentrate Diets and Its Molecular Effects on Skeletal Muscle During Finishing Phase. Biol Trace Elem Res 2025:10.1007/s12011-025-04556-7. [PMID: 40011411 DOI: 10.1007/s12011-025-04556-7] [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: 11/14/2024] [Accepted: 02/16/2025] [Indexed: 02/28/2025]
Abstract
Chromium is an essential trace mineral in insulin-mediated glucose metabolism, potentially affecting protein synthesis and improving performance and meat quality. This study aimed to assess the impact of chromium on performance, carcass characteristics, meat quality, and the gene expression in the skeletal muscle of lambs fed a high-concentrate diet. Sixteen just just-weaned Dorper × Santa Inês crossbred lambs, weighing 24.95 ± 1.97 kg, were allocated into a control group, a basal diet without chromium in the diet supplementation (CTL) or chromium supplementation (Cr3.0), with the addition of 3.0 mg of chromium per kg of dry matter (Cr3.0DM). The animals were housed in individual pens and fed a diet comprising 6% forage and 94% concentrate for 60 days. We evaluated average daily gain (ADG), dry matter intake (DMI), feed efficiency (FE), carcass characteristics, meat quality, and muscle transcriptome. Chromium supplementation increased ADG (P = 0.048) and tended to improve FE (P = 0.08). Further, chromium supplementation increased kidney, pelvic, and heart fat (P ≤ 0.05). In meat, a reduction in cooking losses and an increase in shear force were observed in the meat of lambs treated with chromium-treated lambs (P ≤ 0.05). Transcriptome analysis revealed 21 differentially expressed genes, with 9 downregulated genes, including COX6C, TMED5, ODC1, HNRNPA1, and 12 upregulated genes, such as RPL39, RPL35A, RPS25, NSA2, RRM2, GCNT1, associated with pathways of ribosome biogenesis, protein synthesis, collagen remodeling, and fat metabolism. In conclusion, dietary chromium supplementation at 3.0 mg/kg DM can improve performance and efficiency without affecting carcass characteristics. In meat, chromium reduced cooking losses, potentially increasing its juiciness, but reduced its juiciness-related tenderness, negatively impacting the tenderness of the meat.
Collapse
Affiliation(s)
- Helena Viel Alves Bezerra
- Department of Animal Science, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, SP, 13635-900, Brazil.
| | - German Darío Ramírez-Zamudio
- Department of Animal Science, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, SP, 13635-900, Brazil.
| | - Miguel Henrique de Almeida Santana
- Department of Animal Science, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, SP, 13635-900, Brazil
| | - Guilherme Henrique Gebim Polizel
- Department of Animal Science, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, SP, 13635-900, Brazil
| | - Regner Ítalo Gonçalves de Oliveira
- Department of Animal Science, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, SP, 13635-900, Brazil
| | - Guilherme Pegoraro Rissi
- Department of Animal Science, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, SP, 13635-900, Brazil
| | - André Ricardo Frujuelle Filho
- Department of Animal Science, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, SP, 13635-900, Brazil
| | - Mirele Daiana Poleti
- Department of Biosciences, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, SP, 13635-900, Brazil
| | - Leonardo Velloso
- Department of Food Engineering, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, SP, 13635-900, Brazil
| | - Thiago Henrique da Silva
- Department of Animal Science, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, SP, 13635-900, Brazil
| | - Sarita Bonagurio Gallo
- Department of Animal Science, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, SP, 13635-900, Brazil
| | - Saulo da Luz E Silva
- Department of Animal Science, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, SP, 13635-900, Brazil
| | - Nara Regina Brandão Cônsolo
- Department of Animal Nutrition and Production, Faculty of Veterinary Medicine and Animal Science, University of São Paulo, Pirassununga, SP, 13635-900, Brazil
| | - Paulo Roberto Leme
- Department of Animal Science, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, SP, 13635-900, Brazil
| |
Collapse
|
33
|
Liu X, Chen S, Liu X, Wu X, Jiang X, Li Y, Yang Z. Enpp1 ameliorates MAFLD by regulating hepatocyte lipid metabolism through the AMPK/PPARα signaling pathway. Cell Biosci 2025; 15:22. [PMID: 39972484 PMCID: PMC11841222 DOI: 10.1186/s13578-025-01364-3] [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: 08/09/2024] [Accepted: 02/07/2025] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND Metabolic dysfunction-associated fatty liver disease (MAFLD) has become the leading chronic liver disease globally, and there are no approved pharmacotherapies to treat this disease. Ectonucleotide pyrophosphatase/phosphodiesterase 1 (Enpp1) has been found to be related to insulin resistance and lipid accumulation. However, the role and mechanism of Enpp1 in the development of MAFLD remain unknown. RESULTS Here we discovered that Enpp1 is lowly expressed in the liver of MAFLD patients by clinical investigation. Knocking out Enpp1 in the liver of mice aggravated obesity, insulin resistance and hepatic steatosis, and these effects were reversed by liver-specific Enpp1 overexpression. Through transcriptomic data mining and experimental validation, we demonstrated that Enpp1 deficiency inhibited the expression of AMPK (energy receptor) and PPARα (nuclear transcription factor for lipid metabolism), thereby promoting the transcription of lipid synthesis factors and mediating the progression of MAFLD. Mechanistically, Enpp1 enhanced the activity of AMPK by increasing the AMP-to-ATP ratio, which in turn raised PPARα levels and promoted the transcription of its downstream lipid metabolism factors. Pharmacological inhibition of AMPK activity abolished the promoting effect of Enpp1 on PPARα protein expression. CONCLUSIONS This study indicate that Enpp1 can effectively ameliorate MAFLD through effects on AMPK/PPARα signaling pathway-mediated lipid metabolism, revealing the significance of Enpp1 as a promising therapeutic target against MAFLD.
Collapse
Affiliation(s)
- Xiaohui Liu
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) & Comparative Medicine Centre, Peking Union Medical College (PUMC), Beijing, 100021, China
| | - Shuai Chen
- Fuyang People's Hospital affiliated to Anhui Medical University, Fuyang, China
| | - Xing Liu
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) & Comparative Medicine Centre, Peking Union Medical College (PUMC), Beijing, 100021, China
| | - Xianxian Wu
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) & Comparative Medicine Centre, Peking Union Medical College (PUMC), Beijing, 100021, China
| | - Xiaoliang Jiang
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) & Comparative Medicine Centre, Peking Union Medical College (PUMC), Beijing, 100021, China
| | - Yuhan Li
- Department of Clinical Laboratory, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China.
| | - Zhiwei Yang
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences (CAMS) & Comparative Medicine Centre, Peking Union Medical College (PUMC), Beijing, 100021, China.
| |
Collapse
|
34
|
Wang Y, Liao B, Shan X, Ye H, Wen Y, Guo H, Xiao F, Zhu H. Revealing rutaecarpine's promise: A pathway to parkinson's disease relief through PPAR modulation. Int Immunopharmacol 2025; 147:114076. [PMID: 39809102 DOI: 10.1016/j.intimp.2025.114076] [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: 10/23/2024] [Revised: 01/04/2025] [Accepted: 01/08/2025] [Indexed: 01/16/2025]
Abstract
The pathological mechanisms of Parkinson's disease (PD) is complex, and no definitive cure currently exists. This study identified Rutaecarpine (Rut), an alkaloid extracted from natural plants, as a potential therapeutic agent for PD. To elucidate its mechanisms of action and specific effects in PD, network pharmacology, molecular docking, and experimental validation methods were employed. Our findings demonstrated the efficacy of Rut in ameliorating PD symptoms. Network pharmacology analysis indicated that Rut exerts its therapeutic effects through the PPAR signaling pathway and the lipid pathway. Molecular docking results revealed that Rut forms stable protein-ligand complexes with PPARα and PPARγ. Animal experiments showed that Rut improved motor function in PD mice, protected dopaminergic neurons, ameliorated lipid metabolism disorders, and reduced neuroinflammation. This study identified the critical molecular mechanisms and therapeutic targets of Rut in the treatment of PD, providing a theoretical foundation for future investigations into the pharmacodynamics of Rut as a potential anti-PD agent.
Collapse
Affiliation(s)
- Yeying Wang
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006 Jiangxi, China; The Second Clinical Medical College of Nanchang University, Nanchang 330006 Jiangxi, China.
| | - Bin Liao
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006 Jiangxi, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang 330006 Jiangxi, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang 330006 Jiangxi, China; Institute of Neuroscience, Nanchang University, Nanchang 330006 Jiangxi, China.
| | - Xuesong Shan
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006 Jiangxi, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang 330006 Jiangxi, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang 330006 Jiangxi, China; Institute of Neuroscience, Nanchang University, Nanchang 330006 Jiangxi, China.
| | - Haonan Ye
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006 Jiangxi, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang 330006 Jiangxi, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang 330006 Jiangxi, China; Institute of Neuroscience, Nanchang University, Nanchang 330006 Jiangxi, China.
| | - Yuqi Wen
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006 Jiangxi, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang 330006 Jiangxi, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang 330006 Jiangxi, China; Institute of Neuroscience, Nanchang University, Nanchang 330006 Jiangxi, China.
| | - Hua Guo
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006 Jiangxi, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang 330006 Jiangxi, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang 330006 Jiangxi, China; Institute of Neuroscience, Nanchang University, Nanchang 330006 Jiangxi, China.
| | - Feng Xiao
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006 Jiangxi, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang 330006 Jiangxi, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang 330006 Jiangxi, China; Institute of Neuroscience, Nanchang University, Nanchang 330006 Jiangxi, China.
| | - Hong Zhu
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006 Jiangxi, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang 330006 Jiangxi, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang 330006 Jiangxi, China; Institute of Neuroscience, Nanchang University, Nanchang 330006 Jiangxi, China.
| |
Collapse
|
35
|
Zhu Z, Guan Y, Gao S, Guo F, Liu D, Zhang H. Impact of natural compounds on peroxisome proliferator-activated receptor: Molecular effects and its importance as a novel therapeutic target for neurological disorders. Eur J Med Chem 2025; 283:117170. [PMID: 39700874 DOI: 10.1016/j.ejmech.2024.117170] [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: 07/16/2024] [Revised: 12/10/2024] [Accepted: 12/12/2024] [Indexed: 12/21/2024]
Abstract
Neurological disorders refer to the pathological changes of the nervous system involving multiple pathological mechanisms characterized by complex pathogenesis and poor prognosis. Peroxisome proliferator-activated receptor (PPAR) is a ligand-activated transcription factor that is a member of the nuclear receptor superfamily. PPAR has attracted considerable attention in the past decades as one of the potential targets for the treatment of neurological disorders. Several in vivo and in vitro studies have confirmed that PPARs play a neuroprotective role by regulating multiple pathological mechanisms. Several selective PPAR ligands, such as thiazolidinediones and fibrates, have been approved as pharmacological agonists. Nevertheless, PPAR agonists cause a variety of adverse effects. Some natural PPAR agonists, including wogonin, bergenin, jujuboside A, asperosaponin VI, monascin, and magnolol, have been introduced as safe agonists, as evidenced by clinical or preclinical experiments. This review summarizes the effects of phytochemicals on PPAR receptors in treating various neurological disorders. Further, it summarizes recent advances in phytochemicals as potential, safe, and promising PPAR agonists to provide insights into understanding the PPAR-dependent and independent cascades mediated by phytochemicals. The phytochemicals exhibited potential for treating neurological disorders by inhibiting neuroinflammation, exerting anti-oxidative stress and anti-apoptotic activities, promoting autophagy, preventing demyelination, and reducing brain edema and neurotoxicity. This review presents data that will help clarify the potential mechanisms by which phytochemicals act as pharmacological agonists of PPARs in the treatment of neurological disorders. It also provides insights into developing new drugs, highlighting phytochemicals as potential, safe, and promising PPAR agonists. Additionally, this review aims to enhance understanding of both PPAR-dependent and independent pathways mediated by phytochemicals.
Collapse
Affiliation(s)
- Zhe Zhu
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Yadi Guan
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Songlan Gao
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Feng Guo
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| | - Dong Liu
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| | - Honglei Zhang
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| |
Collapse
|
36
|
Qian X, Zhou Q, Ouyang Y, Wu X, Sun X, Wang S, Duan Y, Hu Z, Hou Y, Wang Z, Chen X, Wang KL, Shen Y, Dong B, Lin Y, Wen T, Tian Q, Guo Z, Li M, Xiao L, Wu Q, Meng Y, Liu G, Ying H, Zhou Y, Zhang W, Duan S, Bai X, Liu T, Zhan P, Lu Z, Xu D. Transferrin promotes fatty acid oxidation and liver tumor growth through PHD2-mediated PPARα hydroxylation in an iron-dependent manner. Proc Natl Acad Sci U S A 2025; 122:e2412473122. [PMID: 39888917 PMCID: PMC11804496 DOI: 10.1073/pnas.2412473122] [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: 06/22/2024] [Accepted: 01/02/2025] [Indexed: 02/02/2025] Open
Abstract
Tumor cells reshape iron and lipid metabolism for their rapid proliferation. However, how tumor cells coordinate the interplay between tumor cell-specific iron homeostasis and lipid metabolism reprogramming to counteract energy shortages remains unclear. Here, we demonstrated that glucose deprivation in hepatocellular carcinoma (HCC) cells induced AMPK-dependent Transferrin S685 phosphorylation, which exposed Transferrin nuclear localization signal (NLS) for binding to importin α7 and subsequent nuclear translocation. Nucleus-translocated Transferrin interacts with PPARα and enhance its protein stability to increase fatty acid oxidation (FAO) upon glucose deprivation. Mechanistically, PPARα-associated Transferrin upregulates iron-dependent PHD2-mediated PPARα P87 hydroxylation and subsequently disrupts the binding of MDM2 to PPARα, therefore inhibiting MDM2-mediated PPARα ubiquitination and degradation. Reconstitution of Transferrin S685A and NLS mutation or knock-in expression of PPARα P87A inhibited PPARα-mediated FAO upon energy stress, enhanced HCC cell apoptosis, and impeded liver tumor growth in mice. Importantly, combined treatment with Transferrin pS685 blocking peptide suppressing AMPK-Transferrin-PPARα axis could synergize with a well-established AMPK activator Metformin to inhibit tumor growth. Additionally, Transferrin pS685-mediated PPARα P87 hydroxylation is positively correlated with PPARα expression levels in human HCC specimens and poor patient prognosis. These findings revealed a mechanism by which Transferrin can sense energy stress to promote the hydroxylation and protein stability of PPARα through iron-dependent activation of PHD2 and underscore the moonlighting function of Transferrin in lipid catabolism and liver tumor development.
Collapse
Affiliation(s)
- Xu Qian
- Department of Clinical Laboratory, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou310022, China
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang310029, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, Zhejiang310029, China
| | - Qimin Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200011, China
| | - Yuan Ouyang
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai200125, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Xiaohong Wu
- National Health Commission (NHC) Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, Heilongjiang150081, China
| | - Xue Sun
- Department of Surgical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang150081, China
| | - Shuo Wang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong250012, China
| | - Yuran Duan
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang310029, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, Zhejiang310029, China
| | - Zhiqiang Hu
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang310029, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, Zhejiang310029, China
| | - Yueru Hou
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang310029, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, Zhejiang310029, China
| | - Zheng Wang
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang310029, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, Zhejiang310029, China
| | - Xiaohan Chen
- Department of Surgical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang150081, China
| | | | - Yuli Shen
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang310029, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, Zhejiang310029, China
| | - Bofei Dong
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang310029, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, Zhejiang310029, China
| | - Yanni Lin
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang310029, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, Zhejiang310029, China
| | - Ting Wen
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang310029, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, Zhejiang310029, China
| | - Qi Tian
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang310029, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, Zhejiang310029, China
| | - Zhanpeng Guo
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang310029, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, Zhejiang310029, China
| | - Min Li
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang310029, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, Zhejiang310029, China
| | - Liwei Xiao
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang310029, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, Zhejiang310029, China
| | - Qingang Wu
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang310029, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, Zhejiang310029, China
| | - Ying Meng
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang310029, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, Zhejiang310029, China
| | - Guijun Liu
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang310029, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, Zhejiang310029, China
| | - Hangjie Ying
- Department of Clinical Laboratory, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou310022, China
| | - Yahui Zhou
- Department of Clinical Laboratory, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou310022, China
| | - Wuchang Zhang
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai200125, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Shengzhong Duan
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou310000, China
| | - Xueli Bai
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang310029, China
| | - Tong Liu
- Department of Surgical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang150081, China
- National Health Commission (NHC) Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, Heilongjiang150081, China
| | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong250012, China
| | - Zhimin Lu
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang310029, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, Zhejiang310029, China
| | - Daqian Xu
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang310029, China
- Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, Zhejiang310029, China
- National Health Commission (NHC) Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, Heilongjiang150081, China
| |
Collapse
|
37
|
Attema B, de la Rosa Rodriguez MA, van Schothorst EM, Grefte S, Hooiveld GJ, Kersten S. Deficiency of the mitochondrial transporter SLC25A47 minimally impacts hepatic lipid metabolism in fasted and diet-induced obese mice. Mol Metab 2025; 92:102092. [PMID: 39746607 PMCID: PMC11773045 DOI: 10.1016/j.molmet.2024.102092] [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: 10/30/2024] [Revised: 12/20/2024] [Accepted: 12/27/2024] [Indexed: 01/04/2025] Open
Abstract
OBJECTIVE The peroxisome proliferator-activated receptor-alpha (PPARα) plays a central role in lipid metabolism in the liver by stimulating the expression of hundreds of genes. Accordingly, regulation by PPARα could be a screening tool to identify novel genes involved in hepatic lipid metabolism. Previously, the mitochondrial transporter SLC25A47 was suggested to play a role in energy metabolism and liver-specific uncoupling, but further research is lacking. METHODS We explored the potential role of SLC25A47 through in vitro studies and using mice overexpressing and lacking SLC25A47. RESULTS SLC25A47 was identified as a PPARα-regulated and fasting-induced gene in human and mouse hepatocytes. Adenoviral-mediated overexpression of SLC25A47 minimally impacted metabolic parameters during fasting and high-fat feeding. During high-fat feeding, SLC25A47 ablation also did not influence any metabolic parameters, apart from a minor improvement in glucose tolerance. In fasted mice, SLC25A47 ablation was associated with modest, reproducible, and likely indirect reductions in plasma triglycerides and glycerol. SLC25A47 ablation did not influence energy expenditure. Depending on the nutritional status, metabolomic analysis showed modest alterations in plasma, liver, and hepatic mitochondrial levels of various metabolites related to amino acid metabolism, TCA cycle, and fatty acid metabolism. No major and consistent alterations in levels of specific metabolites were found that establish the substrate for and function of SLC25A47. CONCLUSION Collectively, our results hint at a role of SLC25A47 in amino acid and fatty acid metabolism, yet suggest that SLC25A47 is dispensable for hepatic lipid homeostasis during fasting and high-fat feeding.
Collapse
Affiliation(s)
- Brecht Attema
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Stippeneng 4, 6708 WE Wageningen, the Netherlands
| | - Montserrat A de la Rosa Rodriguez
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Stippeneng 4, 6708 WE Wageningen, the Netherlands
| | | | - Sander Grefte
- Human and Animal Physiology, Wageningen University, Wageningen, the Netherlands
| | - Guido Jej Hooiveld
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Stippeneng 4, 6708 WE Wageningen, the Netherlands
| | - Sander Kersten
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Stippeneng 4, 6708 WE Wageningen, the Netherlands; Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
38
|
Li YM, Yan MM, Luo T, Zhu W, Jiang JG. Comparative hepatoprotective effects of flavonoids-rich fractions from flowers and leaves of Penthorum chinense Pursh in vitro. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118960. [PMID: 39426574 DOI: 10.1016/j.jep.2024.118960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 10/21/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Penthorum chinense Pursh is a traditional Miao ethnomedicine rich in bioactive components, widely recognized for its hepatoprotective properties. However, the hepatoprotective effects of its flowers and leaves have not been individually elucidated. AIMS OF THE STUDY The objective of this study was to isolate and purify flavonoids-rich fractions from the flowers (PFF) and leaves (PLF) of P. chinense, and to assess their potential protective effects against oxidative, alcohol-induced, and free fatty acid (FFA) induced injury in hepatic cells. MATERIALS AND METHODS The P. chinense flowers and leaves flavonoids-rich fractions were extracted by the method optimized by response surface methodology, and the extracts were subsequently purified using petroleum ether and microporous column. The physical characteristics and component composition of PFF and PLF were analyzed by FT-IR and UPLC-MS/MS. The hepatoprotective activities of PFF and PLF were evaluated by the alcohol, H2O2, and FFA-induced hepatocyte injury cellular model in vitro. The protective effects of PFF and PLF on the hepatic cells were evaluated by assessing cell apoptosis rate, enzymes activities, mitochondrial membrane potential, and mRNA expression in relevant signaling pathways. RESULTS The results revealed that PFF was mainly composed of pinocembrin, quercitrin and quercetin, while PLF was predominantly composed of quercetin, pinocembrin, and kaempferol and their derivatives. PFF and PLF exhibited distinct effects on increasing the cell proliferation rate, regulating the MDA, GOT and GPT levels, and modulating the mRNA expression in apoptosis and antioxidant pathways in alcohol damaged LO2 cells. PFF exhibited superior efficacy in reducing cell apoptosis in alcohol-damage cells compared to PLF. Both PFF and PLF alleviated mitochondrial stress in H2O2-induced LO2 cells. Additionally, the PFF and PLF attenuated lipid accumulation and activated mRNA expressions in PPARα/ACOX1/CPT-1 lipid metabolism pathways, as well as Nrf2/ARE oxidative stress pathways. CONCLUSION This study compared the hepatoprotective activities of flavonoids-rich fractions purified from the flowers and leaves of P. chinense. The results contribute to the enhanced development and utilization of various parts of P. chinense aimed at medical and health food applications.
Collapse
Affiliation(s)
- Yi-Meng Li
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510640, China; Dermatology Hospital of Southern Medical University, Guangzhou, 510091, China; The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Mao-Mao Yan
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510640, China
| | - Ting Luo
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510640, China
| | - Wei Zhu
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510120, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, 510120, China.
| | - Jian-Guo Jiang
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510640, China.
| |
Collapse
|
39
|
Bhardwaj M, Mazumder PM. An insight on the additive impact of type 2 diabetes mellitus and nonalcoholic fatty liver disease on cardiovascular consequences. Mol Biol Rep 2025; 52:169. [PMID: 39873861 DOI: 10.1007/s11033-025-10249-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 01/10/2025] [Indexed: 01/30/2025]
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) and type 2 diabetes mellitus (T2DM) are associated with a multifactorial complicated aetiology that is often coexisting and has a strong and distinct connection with cardiovascular diseases (CVDs). In order to accomplish effective and appropriate therapeutic strategies, a deeper understanding of the bidirectional interaction between NAFLD patients, NAFLD patients with T2DM, and NAFLD patients with CVDs is required to control the concomitant rise in prevalence of these conditions worldwide. This article also aims to shed light on the epidemiology and mechanisms behind the relationship between T2DM, NAFLD and the related cardiovascular consequences. METHOD Literature was collected from PubMed, Medline, Embase, Web of Science and Google scholar from inception to June, 2024. For surveying literature different combinations and formats of terms including NAFLD, NASH, T2DM and CVDs were used. RESULTS In the recent decade, clinical and epidemiological studies have been conducted and provide strong evidence that NAFLD is closely linked with CVD progression along with associated morbidity and mortality in both patients with and without T2DM. Several mechanistic approaches contribute to cardiovascular consequences and abnormalities in cardiac biomarkers in T2DM and NAFLD patients, including adipose tissue malfunction, mitochondrial dysfunction, the microbiota, genetic and epigenetic alterations contributing to insulin resistance, glucotoxicity and lipotoxicity. CONCLUSION The study reveals a complex interplay between diabetes, hepatic and cardiovascular complications, leading to significant morbidity and mortality in diabetic and NAFLD patients. This pandemic necessitates further research to identify mitigating variables and develop effective treatment approaches.
Collapse
Affiliation(s)
- Monika Bhardwaj
- Department of Pharmaceutical Sciences & Technology, BIT Mesra, Ranchi, 835215, India
| | - Papiya Mitra Mazumder
- Department of Pharmaceutical Sciences & Technology, BIT Mesra, Ranchi, 835215, India.
| |
Collapse
|
40
|
Wang Y, Zhang Y, Wang Y. Overexpression of Apolipoprotein A-I Alleviates Insulin Resistance in MASLD Mice Through the PPARα Pathway. Int J Mol Sci 2025; 26:1051. [PMID: 39940822 PMCID: PMC11817368 DOI: 10.3390/ijms26031051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 01/21/2025] [Accepted: 01/22/2025] [Indexed: 02/16/2025] Open
Abstract
Insulin resistance (IR) is one of the important causes of metabolic dysfunction-associated steatotic liver disease (MASLD). Apolipoprotein A-I (apoA-I) is secreted primarily by hepatocytes and plays an essential role in reverse cholesterol transport. Our previous studies revealed that apoA-I can mitigate the progression of metabolic dysfunction-associated steatohepatitis (MASH). However, there is no clear evidence to explain the relationship between apoA-I and IR. Here, we investigated the effects of apoA-I overexpression on IR in both HepG2 cells and mice. In vitro experiment results revealed that apoA-I overexpression can promote cellular glucose uptake in oleic acid-induced IR in HepG2 cells. High-fat, high-cholesterol, and high-fructose diets were used to induce IR in mice. The results showed that apoA-I overexpression improved glucose tolerance, reduced serum insulin levels, and ameliorated IR in diet-induced MASLD mice. Moreover, apoA-I promoted the expression of peroxisome proliferator-activated receptor α (PPARα) in the nucleus both in vitro and in vivo. In conclusion, apoA-I could alleviate MASLD by reducing IR in mice and might exert this effect through the PPARα pathway.
Collapse
Affiliation(s)
| | - Yudian Zhang
- Municipal Laboratory for Liver Protection and Regulation of Regeneration, Department of Cell Biology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China;
| | - Yutong Wang
- Municipal Laboratory for Liver Protection and Regulation of Regeneration, Department of Cell Biology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China;
| |
Collapse
|
41
|
Hu L, Zhang X, Zhang W, Jin S, Zhao J, Zheng J, Song W, Shen Z. Targeting TCMR-associated cytokine genes for drug screening identifies PPARγ agonists as novel immunomodulatory agents in transplantation. Front Immunol 2025; 16:1539645. [PMID: 39911401 PMCID: PMC11794815 DOI: 10.3389/fimmu.2025.1539645] [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: 12/04/2024] [Accepted: 01/06/2025] [Indexed: 02/07/2025] Open
Abstract
Objective T cell-mediated rejection (TCMR) remains a significant challenge in organ transplantation. This study aimed to define a TCMR-associated cytokine gene set and identify drugs to prevent TCMR through drug repurposing. Methods Gene expression profiles from kidney, heart, and lung transplant biopsies were obtained from the Gene Expression Omnibus (GEO) database. Differentially expressed genes (DEGs) between TCMR and non-TCMR groups were identified, and their intersection with cytokine-related genes yielded an 11-gene TCMR-associated cytokine gene set (TCMR-Cs). To evaluate the effectiveness of this gene set, a diagnostic predictive model was constructed using Lasso regression and multivariate logistic regression, with validation in independent datasets. Connectivity Map (CMap) analysis was employed to screen drugs targeting TCMR-Cs. Experimental validation of the identified drug was performed in vitro using T cell activation and Th1 differentiation assays, and in vivo in a mouse skin transplant model with survival analysis. Results The TCMR-Cs exhibited outstanding predictive performance for TCMR, achieving an AUC of 0.99 in the training cohorts and maintaining strong performance in the test cohorts. CMap analysis identified peroxisome proliferator-activated receptor gamma (PPARγ) agonists as potential therapeutic candidates. Experimental validation showed that the PPARγ agonist rosiglitazone significantly suppressed T cell activation and reduced Th1 differentiation in vitro without cytotoxic effects. The combination of rosiglitazone and rapamycin significantly prolonged graft survival. Conclusions This study defined a novel TCMR-associated cytokine gene set that effectively predicts TCMR and identified PPARγ agonists, which prevent TCMR and improve graft survival when combined with rapamycin.
Collapse
Affiliation(s)
- Lu Hu
- The First Central Clinical School, Tianjin Medical University, Tianjin, China
- Research Institute of Transplant Medicine, School of Medicine, Nankai University, Tianjin, China
| | - Xiaohan Zhang
- Research Institute of Transplant Medicine, School of Medicine, Nankai University, Tianjin, China
- School of Medicine, Nankai University, Tianjin, China
| | - Weiqi Zhang
- Research Institute of Transplant Medicine, School of Medicine, Nankai University, Tianjin, China
- School of Medicine, Nankai University, Tianjin, China
| | - Shuai Jin
- The First Central Clinical School, Tianjin Medical University, Tianjin, China
- Research Institute of Transplant Medicine, School of Medicine, Nankai University, Tianjin, China
| | - Jie Zhao
- Department of Renal Transplantation, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Jianming Zheng
- Department of Renal Transplantation, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Wenli Song
- Department of Renal Transplantation, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Zhongyang Shen
- Research Institute of Transplant Medicine, School of Medicine, Nankai University, Tianjin, China
- NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Tianjin, China
- Key Laboratory of Transplant Medicine, Chinese Academy of Medical Sciences, Tianjin, China
| |
Collapse
|
42
|
Wu M, He C, Yu H, Zhang Y, Tang L, Liu M, Gao M, Wu J, Zeng F, Chen H, Jiang S, Zhu Z. Therapeutic targets of antidiabetic drugs and kidney stones: A druggable mendelian randomization study and experimental study in rats. Eur J Pharmacol 2025; 987:177197. [PMID: 39662658 DOI: 10.1016/j.ejphar.2024.177197] [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: 09/07/2024] [Revised: 11/22/2024] [Accepted: 12/09/2024] [Indexed: 12/13/2024]
Abstract
Diabetes is known to increase the risk of kidney stones, but the influence of antidiabetic drugs on this risk remains uncertain. Genetic instruments for antidiabetic drugs were identified as variants, which were associated with both the expression of genes encoding target proteins of drugs and glycated hemoglobin level (HbA1c). Here, we investigated the effect of antidiabetic drugs on kidney stones in a mendelian randomization (MR) framework, and further explore the potential effect on CaOx stone rat models induced by glyoxylic acid. Genetically proxied thiazolidinediones (PPARG agonists) significantly reduced the risk of kidney stones (OR = 0.42; P=0.004) per 1-SD decrement in HbA1c, while no significant association was noted in sulfonylureas, SGLT2 inhibitors, or GLP-1 analogs. Other antidiabetic drugs were not analyzed due to unclear pharmacological targets or no identified instruments. Additionally, PPARG agonists pioglitazone ameliorated CaOx nephrocalcinosis in glyoxylic acid-induced rats. The summary-data-based MR (SMR) results showed that PPARG mRNA expression in blood or kidney was not associated with kidney stone risk, and thus we performed mediation MR of PPARG agonists, circulating metabolites, and kidney stones. Among 249 circulating metabolites, we identified an indirect effect of PPARG agonists on kidney stones through increasing phospholipids to total lipids ratio in very large VLDL, with a mediated proportion of 6.87% (P = 0.018). Our study provided evidence that PPARG agonists reduced the risk of kidney stones partially via regulating lipid metabolism, and PPARG agonists may be a promising study subject in clinical studies for the prevention of kidney stones.
Collapse
Affiliation(s)
- Maolan Wu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Cheng He
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Hao Yu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Youjie Zhang
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Liang Tang
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Minghui Liu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Meng Gao
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Jian Wu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Feng Zeng
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Hequn Chen
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Shilong Jiang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China; The Hunan Institute of Pharmacy Practice and Clinical Research, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Zewu Zhu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China; Department of Internal Medicine, Section Endocrinology, Yale University School of Medicine, New Haven, CT, 06519, USA.
| |
Collapse
|
43
|
Du Y, Huo Y, Yang Y, Lin P, Liu W, Wang Z, Zeng W, Li J, Liang Z, Yuan C, Zhu J, Luo Z, Liu Y, Ma C, Yang C. Role of sirtuins in obesity and osteoporosis: molecular mechanisms and therapeutic targets. Cell Commun Signal 2025; 23:20. [PMID: 39799353 PMCID: PMC11724515 DOI: 10.1186/s12964-024-02025-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 12/30/2024] [Indexed: 01/15/2025] Open
Abstract
The prevalence of obesity and osteoporosis (OP) represents a significant public health concern on a global scale. A substantial body of evidence indicates that there is a complex relationship between obesity and OP, with a correlation between the occurrence of OP and obesity. In recent years, sirtuins have emerged as a prominent area of interest in the fields of aging and endocrine metabolism. Among the various research avenues exploring the potential of sirtuins, the effects of these proteins on obesity and OP have garnered significant attention from numerous researchers. Sirtuins regulate energy balance and lipid balance, which in turn inhibit the process of adipogenesis. Additionally, sirtuins regulate the balance between osteogenic and osteoblastic activity, which protects against the development of OP. However, no study has yet provided a comprehensive discussion of the relationship between the three: sirtuins, obesity, and OP. This paper will therefore describe the relationship between sirtuins and obesity, the relationship between sirtuins and OP, and a discussion focusing on the possibility of treating OP caused by obesity by targeting sirtuins. This will be based on the common influences on the occurrence of obesity and OP (such as mesenchymal stem cells, gut microbiota, and insulin). Finally, the potential of SIRT1, an important member of sirtuins, in polyphenolic natural products for the treatment of obesity and OP will be presented. This will contribute to a better understanding of the interactions between sirtuins and obesity and bone, which will facilitate the development of new therapeutic strategies for obesity and OP in the future.
Collapse
Grants
- Nos. 2021B1515140012, 2023A1515010083 the Natural Science Foundation of Guangdong Province
- No. 20211800905342 the Dongguan Science and Technology of Social Development Program
- No. A2024398 the Medical Scientific Research Foundation of Guangdong Province
- No. k202005 the Research and Development Fund of Dongguan People' s Hospital
- Nos. GDMU2021003, GDMU2021049, GDMU2022031, GDMU2022047, GDMU2022063, GDMU2022077, GDMU2022078, GDMU2023008, GDMU2023015, GDMU2023026, GDMU2023042, GDMU2023102 the Guangdong Medical University Students' Innovation and Entrepreneurship Training Program
- Nos. 202210571008, S202210571075, 202310571031, S202310571047, S202310571078, S202310571063, S202310571077 the Provincial and National College Students' Innovation and Entrepreneurship Training Program
- No. 4SG24028G the Guangdong Medical University-Southern Medical University twinning research team project
- No. PF100-2-01 "Climbing 100" Joint Merit Training Program Funded Project
- Nos. 2023ZYDS001, 2023FZDS001, 2023FYDB010 the Guangdong Medical University Students' Innovation Experiment Program
- the Research and Development Fund of Dongguan People’ s Hospital
- the Guangdong Medical University Students’ Innovation and Entrepreneurship Training Program
- the Provincial and National College Students’ Innovation and Entrepreneurship Training Program
- the Cai Limin National Traditional Chinese Medicine Inheritance Studio
- the Guangdong Medical University Students’ Innovation Experiment Program
Collapse
Affiliation(s)
- Yikuan Du
- Central Laboratory, The Tenth Affiliated Hospital of Southern Medical University, Dongguan, 523059, China
| | - Yuying Huo
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, The First Dongguan Affiliated Hospital, School of Basic Medical Sciences, Guangdong Medical University, Dongguan, 523808, China
| | - Yujia Yang
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, The First Dongguan Affiliated Hospital, School of Basic Medical Sciences, Guangdong Medical University, Dongguan, 523808, China
| | - Peiqi Lin
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, The First Dongguan Affiliated Hospital, School of Basic Medical Sciences, Guangdong Medical University, Dongguan, 523808, China
| | - Wuzheng Liu
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, The First Dongguan Affiliated Hospital, School of Basic Medical Sciences, Guangdong Medical University, Dongguan, 523808, China
| | - Ziqin Wang
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, The First Dongguan Affiliated Hospital, School of Basic Medical Sciences, Guangdong Medical University, Dongguan, 523808, China
| | - Wenqi Zeng
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, The First Dongguan Affiliated Hospital, School of Basic Medical Sciences, Guangdong Medical University, Dongguan, 523808, China
| | - Jiahui Li
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, The First Dongguan Affiliated Hospital, School of Basic Medical Sciences, Guangdong Medical University, Dongguan, 523808, China
| | - Zhonghan Liang
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, The First Dongguan Affiliated Hospital, School of Basic Medical Sciences, Guangdong Medical University, Dongguan, 523808, China
| | - Chenyue Yuan
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, The First Dongguan Affiliated Hospital, School of Basic Medical Sciences, Guangdong Medical University, Dongguan, 523808, China
| | - Jinfeng Zhu
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, The First Dongguan Affiliated Hospital, School of Basic Medical Sciences, Guangdong Medical University, Dongguan, 523808, China
| | - Ziyi Luo
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, The First Dongguan Affiliated Hospital, School of Basic Medical Sciences, Guangdong Medical University, Dongguan, 523808, China
| | - Yi Liu
- The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523808, China
| | - Chunling Ma
- The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523808, China
| | - Chun Yang
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, The First Dongguan Affiliated Hospital, School of Basic Medical Sciences, Guangdong Medical University, Dongguan, 523808, China.
| |
Collapse
|
44
|
Yang N, Wu T, Li M, Hu X, Ma R, Jiang W, Su Z, Yang R, Zhu C. Silver-quercetin-loaded honeycomb-like Ti-based interface combats infection-triggered excessive inflammation via specific bactericidal and macrophage reprogramming. Bioact Mater 2025; 43:48-66. [PMID: 39318638 PMCID: PMC11421951 DOI: 10.1016/j.bioactmat.2024.09.012] [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: 07/04/2024] [Revised: 08/26/2024] [Accepted: 09/08/2024] [Indexed: 09/26/2024] Open
Abstract
Excessive inflammation caused by bacterial infection is the primary cause of implant failure. Antibiotic treatment often fails to prevent peri-implant infection and may induce unexpected drug resistance. Herein, a non-antibiotic strategy based on the synergy of silver ion release and macrophage reprogramming is proposed for preventing infection and bacteria-induced inflammation suppression by the organic-inorganic hybridization of silver nanoparticle (AgNP) and quercetin (Que) into a polydopamine (PDA)-based coating on the 3D framework of porous titanium (SQPdFT). Once the planktonic bacteria (e.g., Escherichia coli, Staphylococcus aureus) reach the surface of SQPdFT, released Que disrupts the bacterial membrane. Then, AgNP can penetrate the invading bacterium and kill them, which further inhibits the biofilm formation. Simultaneously, released Que can regulate macrophage polarization homeostasis via the peroxisome proliferators-activated receptors gamma (PPARγ)-mediated nuclear factor kappa-B (NF-κB) pathway, thereby terminating excessive inflammatory responses. These advantages facilitate the adhesion and osteogenic differentiation of bone marrow-derived mesenchymal stem cells (BMSCs), concomitantly suppressing osteoclast maturation, and eventually conferring superior mechanical stability to SQPdFT within the medullary cavity. In summary, owing to its excellent antibacterial effect, immune remodeling function, and pro-osteointegration ability, SQPdFT is a promising protective coating for titanium-based implants used in orthopedic replacement surgery.
Collapse
Affiliation(s)
- Ning Yang
- Department of Orthopaedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Ting Wu
- CAS Key Lab for Biomedical Effects of Nanomaterials and Nanosafety, Center of Materials Science and Optoelectronics Engineering, National Center for Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100190, China
| | - Meng Li
- Department of Orthopaedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Xianli Hu
- Department of Orthopaedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Ruixiang Ma
- Department of Orthopaedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Wei Jiang
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
| | - Zheng Su
- Department of Orthopaedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Rong Yang
- CAS Key Lab for Biomedical Effects of Nanomaterials and Nanosafety, Center of Materials Science and Optoelectronics Engineering, National Center for Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100190, China
| | - Chen Zhu
- Department of Orthopaedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| |
Collapse
|
45
|
Choi H, An S, Hyun YE, Noh M, Jeong LS. Design, synthesis and biological evaluation of truncated 1'-homologated 4'-selenonucleosides as PPARγ/δ dual modulators. Bioorg Chem 2025; 154:108042. [PMID: 39705933 DOI: 10.1016/j.bioorg.2024.108042] [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: 09/23/2024] [Revised: 11/22/2024] [Accepted: 12/04/2024] [Indexed: 12/23/2024]
Abstract
This study explores the synthesis and evaluation of truncated 1'-homologated 4'-selenonucleosides as dual modulators of PPARγ and PPARδ. Starting with d-lyxose, a 4'-selenosugar was synthesized and condensed with a nucleobase via an SN2 reaction, followed by modifications at the C2- and N6-positions, yielding compounds 3a-l. Structure-activity trend analysis identified compound 3h, featuring 2-chloro and N6-3-iodobenzylamine substituents, as a potent PPARγ partial agonist and PPARδ antagonist (PPARγ Ki = 2.8 μM, PPARδ Ki = 43 nM). This compound significantly enhanced adiponectin production and promoted adipogenic differentiation in hBM-MSCs. The 4'-seleno substitution preserved ligand functionality while enhancing binding affinity and pharmacological efficacy. In silico docking studies supported these binding affinities, demonstrating optimal binding poses for 3h at both PPARγ and PPARδ. These findings underscore the potential of 4'-selenonucleosides as therapeutic agents for metabolic disorders associated with hypoadiponectinemia, meriting further investigation and clinical development.
Collapse
Affiliation(s)
- Hongseok Choi
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Seungchan An
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Young Eum Hyun
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Minsoo Noh
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Lak Shin Jeong
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea; Future Medicine Co., Ltd, 54 Changup-ro, Sujeong-gu, Seongnam, Gyeonggi-do 13449, Republic of Korea.
| |
Collapse
|
46
|
Shou J, Ma J, Wang X, Li X, Chen S, Kang B, Shaw P. Free Cholesterol-Induced Liver Injury in Non-Alcoholic Fatty Liver Disease: Mechanisms and a Therapeutic Intervention Using Dihydrotanshinone I. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2406191. [PMID: 39558866 PMCID: PMC11727260 DOI: 10.1002/advs.202406191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 08/28/2024] [Indexed: 11/20/2024]
Abstract
Build-up of free cholesterol (FC) substantially contributes to the development and severity of non-alcoholic fatty liver disease (NAFLD). Here, we investigate the specific mechanism by which FC induces liver injury in NAFLD and propose a novel therapeutic approach using dihydrotanshinone I (DhT). Rather than cholesterol ester (CE), we observed elevated levels of total cholesterol, FC, and alanine transaminase (ALT) in NAFLD patients and high-cholesterol diet-induced NAFLD mice compared to those in healthy controls. The FC level demonstrated a positive correlation with the ALT level in both patients and mice. Mechanistic studies revealed that FC elevated reactive oxygen species level, impaired the function of lysosomes, and disrupted lipophagy process, consequently inducing cell apoptosis. We then found that DhT protected mice on an HCD diet, independent of gut microbiota. DhT functioned as a potent ligand for peroxisome proliferator-activated receptor α (PPARα), stimulating its transcriptional function and enhancing catalase expression to lower reactive oxygen species (ROS) level. Notably, the protective effect of DhT was nullified in mice with hepatic PPARα knockdown. Thus, these findings are the first to report the detrimental role of FC in NAFLD, which could lead to the development of new treatment strategies for NAFLD by leveraging the therapeutic potential of DhT and PPARα pathway.
Collapse
Affiliation(s)
- Jia‐Wen Shou
- Li Dak Sum Yip Yio Chin R&D Centre for Chinese MedicineThe Chinese University of Hong KongHong Kong852852China
| | - Juncai Ma
- Centre for Cell and Developmental BiologyState Key Laboratory for AgrobiotechnologySchool of Life SciencesThe Chinese University of Hong KongHong Kong852852China
| | - Xuchu Wang
- Department of Laboratory Medicinethe Second Affiliated Hospital of Zhejiang UniversityHangzhou310000China
| | - Xiao‐Xiao Li
- Li Dak Sum Yip Yio Chin R&D Centre for Chinese MedicineThe Chinese University of Hong KongHong Kong852852China
- Research Center for Chinese Medicine InnovationThe Hong Kong Polytechnic UniversityHong Kong852852China
| | - Shu‐Cheng Chen
- School of NursingThe Hong Kong Polytechnic UniversityHong Kong852852China
| | - Byung‐Ho Kang
- Centre for Cell and Developmental BiologyState Key Laboratory for AgrobiotechnologySchool of Life SciencesThe Chinese University of Hong KongHong Kong852852China
| | - Pang‐Chui Shaw
- Li Dak Sum Yip Yio Chin R&D Centre for Chinese MedicineThe Chinese University of Hong KongHong Kong852852China
- School of Life SciencesThe Chinese University of Hong KongHong Kong852852China
- State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants and Institute of Chinese MedicineThe Chinese University of Hong KongHong Kong852852China
| |
Collapse
|
47
|
Nielsen MH, Nøhr-Meldgaard J, Møllerhøj MB, Oró D, Pors SE, Andersen MW, Kamzolas I, Petsalaki E, Vacca M, Harder LM, Perfield JW, Veidal S, Hansen HH, Feigh M. Characterization of six clinical drugs and dietary intervention in the nonobese CDAA-HFD mouse model of MASH and progressive fibrosis. Am J Physiol Gastrointest Liver Physiol 2025; 328:G51-G71. [PMID: 39404770 DOI: 10.1152/ajpgi.00110.2024] [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: 04/10/2024] [Revised: 09/13/2024] [Accepted: 09/23/2024] [Indexed: 12/17/2024]
Abstract
The choline-deficient l-amino acid defined-high-fat diet (CDAA-HFD) mouse model is widely used in preclinical metabolic dysfunction-associated steatohepatitis (MASH) research. To validate the CDAA-HFD mouse, we evaluated disease progression and responsiveness to dietary and pharmacological interventions with semaglutide, lanifibranor, elafibranor, obeticholic acid (OCA), firsocostat, and resmetirom. Disease phenotyping was performed in C57BL/6J mice fed CDAA-HFD for 3-20 wk and ranked using the MASLD Human Proximity Score (MHPS). Semaglutide, lanifibranor, elafibranor, OCA, firsocostat, or resmetirom were profiled as treatment intervention for 8 wk, starting after 6 wk of CDAA-HFD feeding. Semaglutide and lanifibranor were further evaluated as early (preventive) therapy for 9 wk, starting 3 wk after CDAA-HFD diet feeding. In addition, benefits of dietary intervention (chow reversal) for 8 wk were characterized following 6 wk of CDAA-HFD feeding. CDAA-HFD mice demonstrated a nonobese phenotype with fast onset and progression of MASH and fibrosis, high similarity to human MASH-fibrosis, and tumor development after 20 wk of diet-induction. Semaglutide and lanifibranor partially reversed fibrosis when administered as prevention but not as treatment intervention. Elafibranor was the only interventional drug therapy to improve fibrosis. In comparison, chow-reversal resulted in complete regression of steatosis with improved liver inflammation and fibrosis in CDAA-HFD mice. CDAA-HFD mice recapitulate histological hallmarks of advanced MASH with progressive severe fibrosis, however, in the absence of a clinical translational obese dysmetabolic phenotype. CDAA-HFD mice are suitable for profiling drug candidates directly targeting hepatic lipid metabolism, inflammation, and fibrosis. The timing of pharmacological intervention is critical for determining antifibrotic drug efficacy in the model.NEW & NOTEWORTHY The CDAA-HFD mouse model is widely used in preclinical MASH research, but validation of the model is lacking. This study presents the longitudinal characterization of disease progression. Furthermore, late-stage clinical compounds and dietary intervention (chow reversal) display distinct hepatoprotective effects in the model. Collectively, the study provides critical information guiding the use of the CDAA-HFD mouse model in preclinical drug discovery for MASH and fibrosis.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ioannis Kamzolas
- TVP Lab, WT/MRC Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Hinxton, United Kingdom
| | - Evangelia Petsalaki
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Hinxton, United Kingdom
| | - Michele Vacca
- TVP Lab, WT/MRC Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
- Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Bari, Italy
- Laboratory of Liver Metabolism and MASLD, Roger Williams Institute of Hepatology, London, United Kingdom
| | - Lea Mørch Harder
- Research and Early Development, Novo Nordisk A/S, Måløv, Denmark
| | - James W Perfield
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States
| | - Sanne Veidal
- Research and Early Development, Novo Nordisk A/S, Måløv, Denmark
| | | | | |
Collapse
|
48
|
Leng Y, Zhang Y, Cheng Y, Ye S, Zheng Y, He M, Wu E, Kong L, Zhang H. LIX1L aggravates MASH-HCC progression by reprogramming of hepatic metabolism and microenvironment via CD36. Pharmacol Res 2025; 211:107567. [PMID: 39725340 DOI: 10.1016/j.phrs.2024.107567] [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: 10/08/2024] [Revised: 12/19/2024] [Accepted: 12/24/2024] [Indexed: 12/28/2024]
Abstract
Limb expression 1-like protein (LIX1L) is an essential player in liver disorders, but its function in metabolic dysfunction-associated steatohepatitis (MASH) and associated hepatocellular carcinoma (HCC) progression remains obscure. Here, we identify LIX1L as a key integrative regulator linking lipid metabolism and inflammation, adipose tissue and hepatic microenvironment, which promotes MASH progression. LIX1L significantly upregulates in MASH patients, mouse models, and palmitic acid-stimulated hepatocytes. Lix1l deletion inhibits hepatic lipid accumulation, inflammation, and fibrosis as well as adipocyte differentiation by downregulating CD36, alleviating MASH and associated HCC progression in mice. Mechanistically, metabolic stress promotes PARP1-mediated poly-ADP-ribosylation of LIX1L to increase stability and RNA binding ability of LIX1L. Subsequently, LIX1L binds to AU-rich element in the 3'UTR and CDS of CD36 mRNA, thus mitigating CD36 mRNA decay. Furthermore, LIX1L deficiency-mediated downregulation of CD36 reprograms the tumor-prone liver microenvironment with increased cytotoxic T lymphocytes and reduced immunosuppressive cell proportions. These data indicate a systematic function of LIX1L in the pathogenesis of MASH and underscore targeting PARP1/LIX1L/CD36 axis as a feasible strategy for treatment of MASH and associated HCC.
Collapse
Affiliation(s)
- Yingrong Leng
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yanqiu Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yang Cheng
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Shengtao Ye
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Ying Zheng
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Mengmeng He
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Enyi Wu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Lingyi Kong
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| | - Hao Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
49
|
Guo Z, Chen E, Xie X, Guo Y, Zhang M, Zhu Y, Wang Y, Fang F, Yan L, Liu X. Flll32, a curcumin analog, improves adipose tissue thermogenesis. Biochem Biophys Res Commun 2024; 737:150919. [PMID: 39486136 DOI: 10.1016/j.bbrc.2024.150919] [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: 10/14/2024] [Revised: 10/17/2024] [Accepted: 10/27/2024] [Indexed: 11/04/2024]
Abstract
Adipose tissue is a key regulator of systemic energy homeostasis and improving adipose tissue function provides a brand-new theoretical reference for the prevention and treatment of obesity. FLLL32, a curcumin analog, can hinder various carcinogenic processes, however, its role in adipose tissue has not been fully elucidated. In this study, we observed that FLLL32 treatment significantly improved cold intolerance and reduced white adipose tissue (WAT) adipocyte size in mice, but had no effect on body weight and adipose tissues weight. Furthermore, FLLL32 treatment upregulated the expression level of uncoupling protein 1 and downregulated the expression level of peroxisome proliferator-activated receptor gamma in adipose tissue. Additionally, FLLL32 promoted the mRNA level of transferrin receptor protein 1, a key iron transporter on the cell membrane, and the lipid peroxidation in inguinal WAT. Finally, FLLL32 significantly inhibited the differentiation and maturation of preadipocytes. In summary, our results demonstrated that FLLL32 plays a crucial role in regulating adipose tissue function.
Collapse
Affiliation(s)
- Zeyu Guo
- Department of Biochemistry & Molecular Biology, State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences & School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Enhui Chen
- Department of Pathophysiology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences & School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Xianghong Xie
- Department of Biochemistry & Molecular Biology, State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences & School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Yanfang Guo
- Department of Biochemistry & Molecular Biology, State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences & School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Minglong Zhang
- Department of Pathophysiology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences & School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Yinghan Zhu
- Department of Pathophysiology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences & School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Yiting Wang
- Department of Biochemistry & Molecular Biology, State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences & School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Fude Fang
- Department of Biochemistry & Molecular Biology, State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences & School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Li Yan
- Department of Pathophysiology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences & School of Basic Medicine Peking Union Medical College, Beijing, 100005, China.
| | - Xiaojun Liu
- Department of Biochemistry & Molecular Biology, State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences & School of Basic Medicine Peking Union Medical College, Beijing, 100005, China.
| |
Collapse
|
50
|
Melini S, Pirozzi C, Lama A, Comella F, Opallo N, Del Piano F, Di Napoli E, Mollica MP, Paciello O, Ferrante MC, Mattace Raso G, Meli R. Co-Micronized Palmitoylethanolamide and Rutin Associated With Hydroxytyrosol Recover Diabesity-Induced Hepatic Dysfunction in Mice: In Vitro Insights Into the Synergistic Effect. Phytother Res 2024; 38:6035-6047. [PMID: 39474783 PMCID: PMC11634826 DOI: 10.1002/ptr.8361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 09/05/2024] [Accepted: 10/04/2024] [Indexed: 12/13/2024]
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) and diabesity (diabetes related to obesity) are interrelated since glucose and lipid alterations play a vital role in the development of both disorders. Due to their multi-variant metabolic features, more than one drug or natural product may be required to achieve proper therapeutic effects. This study aimed to evaluate the effectiveness of a formulation containing co-micronized palmitoylethanolamide and rutin (PEA-Rut) associated with hydroxytyrosol (HT), namely NORM3, against hepatic damage and metabolic alterations in high-fat diet (HFD)-induced diabesity in mice. NORM3 decreased the body weight and fat mass of obese mice. The formulation improved HFD-altered insulin sensitivity and hepatic glucose production and metabolism, as shown by glucose, insulin, pyruvate tolerance tests, Western blot, and real-time PCR. In the liver, NORM3 limited macro- and micro-vacuolar steatosis, as revealed by morphological analysis, and reduced the associated hepatic inflammation. NORM3 counteracted lipid dysfunctions of HFD animals, activating AMPK, a key cellular energy sensor, and normalizing the expression of carnitine palmitoyl-transferase (CPT)1, a rate-limiting enzyme of fatty acid β-oxidation, and other genes involved in lipid homeostasis. Relevantly, the hepatic antioxidant activity of NORM3 was proved (reduced ROS and increased detoxifying factors and enzymes). Finally, in vitro synergistic protective effects of the components (PEA-Rut and HT) on H2O2-induced oxidative challenge in HepG2 were determined (ROS production, inflammation, and antioxidant defense). Our results show the beneficial effect of NORM3 and its potential as an innovative phytotherapeutic combination in limiting hepatic damage progression and counteracting glucose and lipid dysmetabolism associated with diabesity.
Collapse
Affiliation(s)
- S. Melini
- Department of Pharmacy, School of MedicineUniversity of Naples Federico IINaplesItaly
| | - C. Pirozzi
- Department of Pharmacy, School of MedicineUniversity of Naples Federico IINaplesItaly
| | - A. Lama
- Department of Pharmacy, School of MedicineUniversity of Naples Federico IINaplesItaly
| | - F. Comella
- Department of Pharmacy, School of MedicineUniversity of Naples Federico IINaplesItaly
| | - N. Opallo
- Department of Pharmacy, School of MedicineUniversity of Naples Federico IINaplesItaly
| | - F. Del Piano
- Department of Veterinary Medicine and Animal ProductionsUniversity of Naples Federico IINaplesItaly
| | - E. Di Napoli
- Department of Veterinary Medicine and Animal ProductionsUniversity of Naples Federico IINaplesItaly
| | - M. P. Mollica
- Department of BiologyUniversity of Naples Federico IINaplesItaly
| | - O. Paciello
- Department of Veterinary Medicine and Animal ProductionsUniversity of Naples Federico IINaplesItaly
| | - M. C. Ferrante
- Department of Veterinary Medicine and Animal ProductionsUniversity of Naples Federico IINaplesItaly
| | - G. Mattace Raso
- Department of Pharmacy, School of MedicineUniversity of Naples Federico IINaplesItaly
| | - R. Meli
- Department of Pharmacy, School of MedicineUniversity of Naples Federico IINaplesItaly
| |
Collapse
|