1
|
Amirkhizi F, Taghizadeh M, Khalese-Ranjbar B, Hamedi-Shahraki S, Asghari S. The clinical value of serum sirtuin-1 concentration in the diagnosis of metabolic dysfunction-associated steatotic liver disease. BMC Gastroenterol 2025; 25:27. [PMID: 39844087 PMCID: PMC11753077 DOI: 10.1186/s12876-025-03613-8] [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] [Accepted: 01/14/2025] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most prevalent chronic liver disease and can affect individuals without producing any symptoms. We aimed to explore the value of serum sirtuin-1 (Sirt-1) in the diagnosis of MASLD. METHODS This case-control study analyzed data collected from 190 individuals aged 20 to 60 years. Anthropometric parameters, demographic information, and serum biochemical variables-including glycemic parameters, lipid profiles, liver enzymes, and Sirt-1 levels-were assessed. The correlation between serum Sirt-1 and biochemical variables was examined using Pearson's correlation coefficient. Receiver operating characteristic (ROC) curve analysis was employed to evaluate the diagnostic value of serum Sirt-1 in the context of MASLD. RESULTS Serum Sirt-1 levels was significantly lower in the MASLD group (p < 0.001) and was inversely correlated with serum insulin (r = -0.163, p = 0.025), HOMA-IR (r = -0.169, p = 0.020) and triglyceride (r = -0.190, p = 0.009) and positively correlated with serum levels of high-density lipoprotein cholesterol (HDL-C) (r = 0.214, p = 0.003). The area under the curve (AUC) of Sirt-1 to predict the presence of MASLD was 0.76 (p < 0.001, 95% CI: 0.69, 0.82) with a sensitivity of 78.9, specificity of 61.1, positive predictive value (PPV) of 67.0%, and negative predictive value (NPV) of 74.0%. The optimal cutoff, determined using Youden's index, was 23.75 ng/mL. This indicates that serum Sirt-1 levels below 23.75 ng/mL may be indicative of MASLD. CONCLUSIONS The present study demonstrated that serum Sirt-1 levels were significantly lower in patients with MASLD. Furthermore, these levels were correlated with various metabolic parameters, including insulin resistance and the serum lipid profile. A serum Sirt-1 level below the cutoff of 23.75 ng/mL exhibited a significant association with the presence of MASLD, suggesting its potential utility in identifying patients with this condition.
Collapse
Affiliation(s)
- Farshad Amirkhizi
- Department of Nutrition, School of Public Health, Zabol University of Medical Sciences, Zabol, Iran
| | - Mahdiyeh Taghizadeh
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, No#44, Hojjatdoust St., Naderi St., Keshavarz Blvd, Tehran, 141556117, Iran
| | - Banafshe Khalese-Ranjbar
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, No#44, Hojjatdoust St., Naderi St., Keshavarz Blvd, Tehran, 141556117, Iran
| | - Soudabeh Hamedi-Shahraki
- Department of Epidemiology and Biostatistics, School of Public Health, Zabol University of Medical Sciences, Zabol, Iran
| | - Somayyeh Asghari
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, No#44, Hojjatdoust St., Naderi St., Keshavarz Blvd, Tehran, 141556117, Iran.
| |
Collapse
|
2
|
Liu Y, Zhou F, Zhao H, Song J, Song M, Zhu J, Wang Y, Man Hoi MP, Lin L, Zhang Q. Dimeric guaianolide sesquiterpenoids from the flowers of Chrysanthemum indicum ameliorate hepatic steatosis through mitigating SIRT1-mediated lipid accumulation and ferroptosis. J Adv Res 2025:S2090-1232(24)00625-8. [PMID: 39788286 DOI: 10.1016/j.jare.2024.12.047] [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: 08/09/2024] [Revised: 10/04/2024] [Accepted: 12/28/2024] [Indexed: 01/12/2025] Open
Abstract
INTRODUCTION Non-alcoholic fatty liver disease (NAFLD) acts as the primary contributor to non-alcoholic steatohepatitis, fibrosis, cirrhosis, and potentially hepatocellular carcinoma. The flowers of Chrysanthemum indicum, a traditional edible medicinal herb, have been widely used in China for more than 2000 years. However, the function of C. indicum in managing NAFLD has seldom been investigated. OBJECTIVES To reveal the novel active components and underlying mechanisms of C. indicum in treating NAFLD. METHODS An MS/MS-based molecular networking-guided strategy was used for the chemical investigation. The structure identification of the new compounds involved high resolution electrospray ionization mass spectrometry (HRESIMS), 1D and 2D nuclear magnetic resonance (NMR) spectra, electronic circular dichroism (ECD), and X-ray crystallographic analysis. The biological evaluation was performed using Nile Red staining, flow cytometry, commercial kits, western blotting, co-immunoprecipitation, isothermal titration calorimetry, cellular thermal shift assay, drug affinity responsive target stability assay, molecular docking, and confocal immunofluorescence. RESULTS A total of 27 new dimeric sesquiterpenoids, chryindicolides A-Z (1-26) and chrysanthemolide C (27), together with seven known compounds, were isolated from the flowers of C. indicum under the guide of MS/MS-based molecular networking. Among them, compounds 1-7 were rare chlorine-containing guaianolide dimers. Chryindicolide O (15) directly bound and activated the deacetylase Sirtuin 1 (SIRT1) to reduce de novo lipogenesis, enhance fatty acid β-oxidation, and inhibit ferroptosis in palmitic acid and oleic acid (P/O)-induced AML12 hepatocytes. In addition, chryindicolide O significantly ameliorated liver steatosis in high-fat diet-fed zebrafish. CONCLUSION Novel guaianolide dimers from C. indicum alleviated hepatic steatosis through mitigating SIRT1-mediated lipid accumulation and ferroptosis, suggesting that they could be further developed as candidates against NAFLD.
Collapse
Affiliation(s)
- Yu Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao 999078, China; Guangdong-Hong Kong-Macau Join Laboratory for Pharmacodynamic Constituents of TCM and New Drugs Research, University of Macau, Avenida da Universidade, Taipa, Macao 999078, China
| | - Fei Zhou
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao 999078, China
| | - Haoyu Zhao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao 999078, China
| | - Jianguo Song
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China; Guangdong-Hong Kong-Macau Join Laboratory for Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou 510632, China
| | - Min Song
- Guangdong Provincial Engineering Research Center for Modernization of TCM, Jinan University, Guangzhou 510632, China
| | - Jianzhong Zhu
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Ying Wang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China; Guangdong-Hong Kong-Macau Join Laboratory for Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou 510632, China
| | - Maggie Pui Man Hoi
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao 999078, China; Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macao 999078, China
| | - Ligen Lin
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao 999078, China; Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macao 999078, China.
| | - Qingwen Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao 999078, China; Guangdong-Hong Kong-Macau Join Laboratory for Pharmacodynamic Constituents of TCM and New Drugs Research, University of Macau, Avenida da Universidade, Taipa, Macao 999078, China; Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macao 999078, China.
| |
Collapse
|
3
|
Elmorsy EA, Elsisi HA, Alkhamiss AS, Alsoqih NS, Khodeir MM, Alsalloom AA, Almeman AA, Elghandour SR, Nadwa EH, Khalifa AK, Khaled BEA, Ramadan A, Kamal MM, Alsaeed TS, Alharbi MS, Abdel-Moneim AMH, Ellethy AT, Saber S. Activation of SIRT1 by SRT1720 alleviates dyslipidemia, improves insulin sensitivity and exhibits liver-protective effects in diabetic rats on a high-fat diet: New insights into the SIRT1/Nrf2/NFκB signaling pathway. Eur J Pharm Sci 2025; 206:107002. [PMID: 39778687 DOI: 10.1016/j.ejps.2025.107002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 11/29/2024] [Accepted: 01/05/2025] [Indexed: 01/11/2025]
Abstract
Insulin resistance and diabetes are associated with non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH) conditions, which are distinguished by metabolic dysfunction, oxidative stress and inflammation. Sirtuin 1 (SIRT1), a NAD+-dependent deacetylase, is fundamental in regulating metabolic pathways, reducing inflammation, and improving antioxidant defenses. This is the first study to investigate the effects of SRT1720, a SIRT1 activator, in diabetic rats on a high-fat diet. SRT1720 significantly lowered fasting blood glucose and insulin levels and enhanced glucose tolerance and HOMA-IR and QUICKI scores, indicating increased insulin sensitivity. The treatment also reduced total cholesterol, triglycerides, and LDL levels, showing amelioration of dyslipidemia. Moreover, SRT1720 lowered markers of liver fibrosis, including TGF-β, TIMP-1, Col1a1, and hydroxyproline, and decreased inflammation by reducing NFκB activity and pro-inflammatory cytokines (TNF-α and IL-6). Furthermore, SRT1720 augmented Nrf2 activity and HO-1 levels. Consequently, the SRT1720's protective role improved liver function and histology and prolonged rats' survival. These functions were suppressed by the co-administration of the SIRT1 inhibitor EX527, confirming that the beneficial effects of SRT1720 are SIRT1-dependent. Correlation analyses uncovered that increased SIRT1 activity was strongly associated with decreased oxidative stress, inflammation, insulin resistance, and fibrosis markers. To conclude, our results find that SRT1720 represents a promising therapeutic strategy for managing Type 2 diabetes in NAFLD or NASH patients possibly through the modulation of the SIRT1/Nrf2/NFκB signaling pathwa. SRT1720 could potentially halt or reverse the progression of these conditions and associated complications and merits further investigations.
Collapse
Affiliation(s)
- Elsayed A Elmorsy
- Department of Pharmacology and Therapeutics, College of Medicine, Qassim University, Buraidah 51452, Saudi Arabia; Department of Clinical Pharmacology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt.
| | - Hossam A Elsisi
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraidah 51452, Saudi Arabia; Department of Clinical Pharmacology, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt.
| | - Abdullah S Alkhamiss
- Department of Pathology, College of Medicine, Qassim University, Buraidah 51452, Saudi Arabia.
| | - Norah Suliman Alsoqih
- Department of Pediatrics, College of Medicine, Qassim University, Buraidah 51452, Saudi Arabia.
| | - Mostafa M Khodeir
- Department of Pathology, College of Medicine, Qassim University, Buraidah 51452, Saudi Arabia; Department of Pathology, Faculty of Medicine, Cairo University, Cairo 11562, Egypt.
| | - Abdulaziz A Alsalloom
- Department of Pathology, College of Medicine, Qassim University, Buraidah 51452, Saudi Arabia.
| | - Ahmad A Almeman
- Department of Pharmacology and Therapeutics, College of Medicine, Qassim University, Buraidah 51452, Saudi Arabia.
| | - Sahar R Elghandour
- Department of Anatomy and Histology, College of Medicine, Qassim University, Buraidah 51452, Saudi Arabia.
| | - Eman Hassan Nadwa
- Department of Pharmacology and Therapeutics, College of Medicine, Jouf University, Sakaka, Saudi Arabia; Department of Medical Pharmacology, Faculty of Medicine, Cairo University, Cairo 11562, Egypt.
| | - Amira Karam Khalifa
- Department of Medical Pharmacology, Faculty of Medicine, Cairo University, Cairo 11562, Egypt; Department of Medical Pharmacology, Nahda Faculty of Medicine, Beni Suef, 62521, Egypt.
| | - Bahaa Eldin Ali Khaled
- Department of Anatomy, College of Medicine, Jouf University, Sakaka, Saudi Arabia; Department of Anatomy and Embryology, Faculty of Medicine, Cairo University, Cairo 11562, Egypt.
| | - Asmaa Ramadan
- Department of Biochemistry, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa 11152, Egypt.
| | - Manal M Kamal
- Department of Medical Physiology, Faculty of Medicine, Assiut University, Assiut, Egypt; Department of Physiology, College of Medicine, Qassim University, Buraidah 51452, Saudi Arabia.
| | - Thamir Saad Alsaeed
- Department of Biology and Immunology, College of Medicine, Qassim University, Buraidah 51452, Saudi Arabia.
| | - Mariam S Alharbi
- Department of Medicine, College of Medicine, Qassim University, Buraidah 51452, Saudi Arabia.
| | | | - Abousree T Ellethy
- Department of Basic Oral Sciences and Dental Education, Biochemistry Division, College of Dentistry, Qassim University, Buraidah 51452, Saudi Arabia.
| | - Sameh Saber
- Department of Biochemistry, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa 11152, Egypt.
| |
Collapse
|
4
|
Ali BM, Elbaz EM, Al-Mokaddem AK, El-Emam SZ, Awny MM. Delphinidin or α-amyrin attenuated liver steatosis and metabolic disarrangement in rats fed a high-fat diet. Biofactors 2025; 51:e2133. [PMID: 39431734 DOI: 10.1002/biof.2133] [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: 03/08/2024] [Accepted: 10/01/2024] [Indexed: 10/22/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a liver pathology concomitant with metabolic disarrangement. This study assessed the therapeutic impacts of delphinidin, an anthocyanin, or α-amyrin, a pentacyclic triterpenoid, on NAFLD in rats and the underlying mechanisms involved. NAFLD was established by feeding a high-fat diet (HFD) for 10 weeks, either alone or in combination with delphinidin (40 mg/kg, oral) or α-amyrin (20 mg/kg, oral). Delphinidin or α-amyrin ameliorated the metabolic and histopathological perturbations induced by HFD. These compounds markedly attenuated NAFLD-induced hepatic steatosis, as evidenced by a substantial decrease in body weight, insulin resistance, and liver and adipose tissue indices. Alongside normalization of the atherogenic index, both improved HFD-mediated abnormalities in serum lipids, liver enzymes, leptin, and ghrelin levels. Moreover, their intervention activated the NFE2 like bZIP transcription factor 2 and heme oxygenase 1 pathways and abrogated HFD-triggered activation of mitogen-activated protein kinase 1 signaling. These remedies inhibited hepatic apoptosis and modulated the gene expression of lipogenic enzymes. Furthermore, histological analysis corroborated the suppression of lipid accumulation and amelioration of hepatic architecture in the treated rats. Our findings highlight the hepatoprotective value of delphinidin or α-amyrin against NAFLD and related metabolic diseases through their insulin-sensitizing, anti-inflammatory, antioxidant, and antiapoptotic effects.
Collapse
Affiliation(s)
- Bassam Mohamed Ali
- Department of Biochemistry, Faculty of Pharmacy, October 6 University, Giza, Egypt
| | - Eman M Elbaz
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Asmaa K Al-Mokaddem
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Soad Z El-Emam
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, October 6 University, Giza, Egypt
| | - Magdy M Awny
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, October 6 University, Giza, Egypt
| |
Collapse
|
5
|
Yang X, Huang J, Wang J, Sun H, Li J, Li S, Tang YE, Wang Z, Song Q. Effect of glucose selenol on hepatic lipid metabolism disorder induced by heavy metal cadmium in male rats. Biochim Biophys Acta Mol Cell Biol Lipids 2024; 1870:159589. [PMID: 39674492 DOI: 10.1016/j.bbalip.2024.159589] [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: 08/12/2024] [Revised: 12/10/2024] [Accepted: 12/11/2024] [Indexed: 12/16/2024]
Abstract
This study used 24 male rats to determine the protective effects of a new selenium molecule (glucose selenol) on cadmium (Cd) induced hepatic toxicity. The rats were randomly divided into four groups: control group, Cd group, Cd + 0.15 Se group, and Cd + 0.4 Se group. The results showed that glucose selenol supplementation alleviated the adverse impact of Cd on lipid metabolism, including decreased serum triacylglycerol and cholesterol levels. Transcriptome analysis revealed that, compared to the control group, Cd changed the expression of 1379 genes - discernibly affecting lipid metabolism pathways. Proteomic analysis primarily indicated alterations in lipid metabolism-related pathways. In conclusion, glucose selenol restored lipid metabolism disorders induced by Cd, thus rescuing hepatic damage. This integrated analysis identified the influence of glucose selenol on Cd-induced hepatic toxicity and provided its potential application prospects in alleviating the impact of heavy metal pollution, such as Cd, on human health.
Collapse
Affiliation(s)
- Xinyi Yang
- College of Life Science, Hunan Normal University, Changsha 410006, Hunan, China.
| | - Jinzhou Huang
- College of Life Science, Hunan Normal University, Changsha 410006, Hunan, China
| | - Juan Wang
- College of Life Science, Hunan Normal University, Changsha 410006, Hunan, China
| | - Huimin Sun
- College of Life Science, Hunan Normal University, Changsha 410006, Hunan, China
| | - JinJin Li
- College of Life Science, Hunan Normal University, Changsha 410006, Hunan, China
| | - Shunfeng Li
- College of Life Science, Hunan Normal University, Changsha 410006, Hunan, China
| | - Yun-E Tang
- College of Life Science, Hunan Normal University, Changsha 410006, Hunan, China
| | - Zhi Wang
- College of Life Science, Hunan Normal University, Changsha 410006, Hunan, China.
| | - Qisheng Song
- Division of Plant Sciences and Technology, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
6
|
Elhemiely AA, Darwish A. Pharmacological and biochemical insights into lead-induced hepatotoxicity: Pathway interplay and the protective effects of arbutin via the oral and intraperitoneal routes in silico and in vivo. Int Immunopharmacol 2024; 142:112968. [PMID: 39226827 DOI: 10.1016/j.intimp.2024.112968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 08/12/2024] [Accepted: 08/15/2024] [Indexed: 09/05/2024]
Abstract
INTRODUCTION Lead acetate (PbAc), a hazardous heavy metal, poses significant threats to human health and the environment because of widespread industrial exposure. PbAc exposure leads to liver injury primarily through oxidative stress and the disruption of key regulatory pathways. Understanding these mechanisms and exploring protective agents are vital for mitigating PbAc-induced hepatotoxicity. Therefore, we aimed to investigate the molecular pathways implicated in PbAc-induced liver damage, focusing on Sirt-1, Nrf2 (HO-1, NQO1, and SOD), Akt-1/GSK3β, m-TOR, and P53. Additionally, we aimed to assess the hepatoprotective effects of arbutin, which is administered orally and intraperitoneally, to determine the most effective delivery method. METHODOLOGY In silico analyses were conducted to identify relevant protein networks associated with Sirt-1 and AKT-1/GSK-3B pathways. The pharmacodynamic properties of arbutin were examined, followed by molecular docking studies to elucidate its interactions with the selected protein network. In vivo preclinical studies were carried out on adult male rats randomly assigned to 6 different treatment groups, including PbAc exposure and PbAc exposure treated with arbutin either orally or intraperitoneally. RESULTS PbAc exposure led to hepatic oxidative stress, as evidenced by elevated MDA levels and SIRT-1 inhibition, disrupting antioxidant pathways and activating antiautophagic and proapoptotic pathways, ultimately resulting in hepatocyte necrosis. Both oral and intraperitoneal arbutin administration effectively modifed these effects, with intraperitoneal delivery showing superior efficacy in mitigating PbAc-induced histological, immunological, and biochemical alterations. CONCLUSION This study provides insights into the molecular mechanisms underlying PbAc-induced liver injury and highlights the hepatoprotective potential of arbutin. These findings suggest that arbutin, particularly when administered intraperitoneally, holds promise as a therapeutic agent for combating PbAc-induced hepatotoxicity.
Collapse
Affiliation(s)
| | - Alshaymaa Darwish
- Department of Biochemistry, Faculty of Pharmacy, Sohag University, Sohag, Egypt.
| |
Collapse
|
7
|
Yang Z, Zarbl H, Kong B, Taylor R, Black K, Kipen H, Basaly V, Fang M, Guo GL. Liver-gut axis signaling regulates circadian energy metabolism in shift workers. FASEB J 2024; 38:e70203. [PMID: 39588921 PMCID: PMC11590413 DOI: 10.1096/fj.202402102r] [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: 09/06/2024] [Revised: 11/03/2024] [Accepted: 11/13/2024] [Indexed: 11/27/2024]
Abstract
Circadian rhythm is critical to maintaining the whole-body metabolic homeostasis of an organism. Chronic disruption of circadian rhythm by shift work is an important risk factor for metabolic diseases. Fibroblast growth factor 15/19 (FGF15/19), a key component in the liver-gut axis, potently suppresses bile acid (BA) synthesis and improves insulin sensitivity. FGF15/19 emerges as a novel pharmaceutical target for prevention and treatment of metabolic diseases. The nicotinamide adenine dinucleotide (NAD+)-dependent sirtuin 1 (SIRT1) deacetylase plays an important role in the maintenance of hepatic homeostasis by linking hepatic metabolism to circadian rhythm. Here, our clinical study identified that circadian rhythmicity and levels of plasma FGF19 and BA profiling, and cellular NAD+-dependent SIRT1 signaling were disturbed in night shift (NS, n = 10) compared to day shift (DS, n = 12) nurses. Our in vitro data showed that recombinant FGF19 protein rescued cellular circadian rhythm disrupted by SIRT1 inhibitors. Furthermore, we determined the effect of FGF15 on circadian rhythm and hepatic metabolism in wild-type (WT), Fgf15 knockout (KO), and Fgf15 transgenic (TG) mice. The expressions of circadian-controlled genes (CCGs) involved in SIRT1 signaling, BA and lipid metabolism, and inflammation were disrupted in Fgf15 KO compared to WT and/or Fgf15 TG mice. Moreover, systemic FGF15 deficiency led to the circadian disturbance of NAD+-dependent SIRT1 signaling and significant reduction during nighttime in mice. These findings suggest that FGF15/19 regulates the circadian energy metabolism, which warrants further studies as a putative prognostic biomarker and pharmaceutical target for preventing against metabolic diseases associated with chronic shift work.
Collapse
Affiliation(s)
- Zhenning Yang
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, RutgersThe State University of New JerseyPiscatawayNew JerseyUSA
- Environmental and Occupational Health Sciences Institute, RutgersThe State University of New JerseyPiscatawayNew JerseyUSA
| | - Helmut Zarbl
- Environmental and Occupational Health Sciences Institute, RutgersThe State University of New JerseyPiscatawayNew JerseyUSA
- Department of Environmental and Occupational Health and Justice, School of Public Health, RutgersThe State University of New JerseyPiscatawayNew JerseyUSA
| | - Bo Kong
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, RutgersThe State University of New JerseyPiscatawayNew JerseyUSA
| | - Rulaiha Taylor
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, RutgersThe State University of New JerseyPiscatawayNew JerseyUSA
| | - Kathleen Black
- Environmental and Occupational Health Sciences Institute, RutgersThe State University of New JerseyPiscatawayNew JerseyUSA
| | - Howard Kipen
- Environmental and Occupational Health Sciences Institute, RutgersThe State University of New JerseyPiscatawayNew JerseyUSA
- Department of Environmental and Occupational Health and Justice, School of Public Health, RutgersThe State University of New JerseyPiscatawayNew JerseyUSA
| | - Veronia Basaly
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, RutgersThe State University of New JerseyPiscatawayNew JerseyUSA
| | - Mingzhu Fang
- Environmental and Occupational Health Sciences Institute, RutgersThe State University of New JerseyPiscatawayNew JerseyUSA
- Department of Environmental and Occupational Health and Justice, School of Public Health, RutgersThe State University of New JerseyPiscatawayNew JerseyUSA
| | - Grace L. Guo
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, RutgersThe State University of New JerseyPiscatawayNew JerseyUSA
- Environmental and Occupational Health Sciences Institute, RutgersThe State University of New JerseyPiscatawayNew JerseyUSA
- VA New Jersey Health Care SystemVeterans Administration Medical CenterEast OrangeNew JerseyUSA
| |
Collapse
|
8
|
Patidar P, Hirani N, Bharti S, Baig MS. Key regulators of hepatic stellate cell activation in alcohol liver Disease: A comprehensive review. Int Immunopharmacol 2024; 141:112938. [PMID: 39163683 DOI: 10.1016/j.intimp.2024.112938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/27/2024] [Accepted: 08/12/2024] [Indexed: 08/22/2024]
Abstract
Alcoholic liver disease (ALD) is a broad category of disorders that begin with liver injury, lead to liver fibrosis, and ultimately conclude in alcohol-induced liver cirrhosis, the most chronic and irreversible liver damage. Liver fibrosis (LF) is a common pathological characteristic observed in most chronic liver inflammatory conditions that involve prolonged inflammation. In this review, we have summarized ethanol-mediated hepatic stellate cell (HSCs) activation and its role in liver fibrosis progression. We highlight important molecular mechanisms that are modulated by ethanol, play a role in the activation of HSCs and the progression of liver fibrosis and identifying potential targets to ameliorate liver fibrosis.
Collapse
Affiliation(s)
- Pramod Patidar
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Nik Hirani
- MRC Centre for Inflammation Research, Queen'sMedicalResearch Institute, University of Edinburgh, Edinburgh, EH164TJ, UK
| | - Shreya Bharti
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Mirza S Baig
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India.
| |
Collapse
|
9
|
Dou JY, Liu SH, Guo J, Wang CY, Dai X, Lian LH, Cui ZY, Nan JX, Wu YL. Dietary supplementation of pterostilbene, a major component in small berries, prevents alcohol-induced liver injury associated with lipid accumulation and inflammation. Food Funct 2024; 15:11206-11219. [PMID: 39449622 DOI: 10.1039/d4fo03898c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
Pterostilbene (PTE), a natural stilbene found in small berries, exhibits multiple pharmacological activities, particularly antioxidant and anti-inflammatory activities. This study explores the dietary supplementation of PTE to ameliorate acute and chronic alcohol-associated liver disease (ALD). C57BL/6 mice were administrated with PTE and subjected to acute or chronic alcohol stimulation. They were intragastrically administered with alcohol (5 g kg-1, 3 times per 24 h) to induce acute alcohol liver injury or fed a Lieber-DeCarli liquid diet containing 5% ethanol for 4 weeks and a single binge to induce chronic alcoholic liver injury. In the acute ethanol model, PTE decreased the serum transaminase and triglyceride (TG) levels and ameliorated lipid droplet accumulation. PTE ameliorated acute ethanol-induced hepatic steatosis by inhibiting the expression of SREBP1 and its target genes and up-regulating PPARα expression. PTE could reverse the inflammatory response by inhibiting NLRP3 activation, inflammatory factor secretion, and macrophage recruitment caused by acute ethanol exposure. PTE could synergistically activate the SIRT1-AMPK and LXR/FXR axis in mice with acute ethanol exposure. In the chronic-binge ethanol feeding model, PTE also decreased serum transaminase and TG levels and ameliorated hepatocellular ballooning, macrovesicular steatosis, lipid accumulation and inflammation. Chronic-binge ethanol feeding could induce extracellular matrix dysfunction with an increase in α-SMA, collagen I and TIMP-1 expression, which was decreased by PTE. PTE increased SIRT1 expression and AMPK phosphorylation and activated the LXRs/FXR axis, which could be reduced by chronic-binge ethanol feeding. PTE could prevent liver injury caused by alcohol regardless of acute or chronic exposure. These results suggest that PTE can be utilized as a dietary health supplement to avoid ALD and promote health and quality of life.
Collapse
Affiliation(s)
- Jia-Yi Dou
- Key Laboratory for Traditional Chinese Korean Medicine Research (State Ethnic Affairs), College of Pharmacy, Yanbian University, Yanji, Jilin Province 133002, China.
| | - Sai-Hu Liu
- Key Laboratory for Traditional Chinese Korean Medicine Research (State Ethnic Affairs), College of Pharmacy, Yanbian University, Yanji, Jilin Province 133002, China.
| | - Jia Guo
- Key Laboratory for Traditional Chinese Korean Medicine Research (State Ethnic Affairs), College of Pharmacy, Yanbian University, Yanji, Jilin Province 133002, China.
| | - Chen-Yu Wang
- Key Laboratory for Traditional Chinese Korean Medicine Research (State Ethnic Affairs), College of Pharmacy, Yanbian University, Yanji, Jilin Province 133002, China.
| | - Xu Dai
- Key Laboratory for Traditional Chinese Korean Medicine Research (State Ethnic Affairs), College of Pharmacy, Yanbian University, Yanji, Jilin Province 133002, China.
| | - Li-Hua Lian
- Key Laboratory for Traditional Chinese Korean Medicine Research (State Ethnic Affairs), College of Pharmacy, Yanbian University, Yanji, Jilin Province 133002, China.
| | - Zhen-Yu Cui
- Key Laboratory for Traditional Chinese Korean Medicine Research (State Ethnic Affairs), College of Pharmacy, Yanbian University, Yanji, Jilin Province 133002, China.
- Jilin Vocational and Technical College, Yanji, Jilin Province 133002, China
| | - Ji-Xing Nan
- Key Laboratory for Traditional Chinese Korean Medicine Research (State Ethnic Affairs), College of Pharmacy, Yanbian University, Yanji, Jilin Province 133002, China.
| | - Yan-Ling Wu
- Key Laboratory for Traditional Chinese Korean Medicine Research (State Ethnic Affairs), College of Pharmacy, Yanbian University, Yanji, Jilin Province 133002, China.
| |
Collapse
|
10
|
Porto-Barbosa T, Ramos LF, Pansa CC, Molica LR, Malaspina O, Moraes KCM. Inhibition of the miR-1914-5p increases the oxidative metabolism in cellular model of steatosis by modulating the Sirt1-PGC-1α pathway and systemic cellular activity. PLoS One 2024; 19:e0313185. [PMID: 39514580 PMCID: PMC11548759 DOI: 10.1371/journal.pone.0313185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 10/20/2024] [Indexed: 11/16/2024] Open
Abstract
Metabolic associated fatty liver disease (MAFLD) is considered an indicator of metabolic syndrome, which affects millions of people around the world and no effective treatment is currently available. MAFLD involves a wide spectrum of liver damage, that initiates from steatosis (fatty live) and may progress to more complex pathophysiology. Then, details in lipid metabolism controlling should be explored aiming to control the fatty liver. In this context, the miR-1914-5p can be considered a potential biotechnology tool to control lipid metabolism in hepatic cells. This miRNA finds potential mRNA binding sequences in more than 100 molecules correlated with energy production and lipid metabolism pointed in bioinformatic platforms. The present study addressed the miR-1914-5p effects in hepatic HepG2/LX-2 co-cultured cells in a in vitro steatotic environment stablished by the addition of 400 μM of a mixture of oleic and palmitic acids. The analyses demonstrated that the inhibition of the miRNA reduced energetic metabolites such as total lipids, triglycerides, cholesterol and even glucose. In addition, the miR-inhibitor-transfected cells did not present any deleterious effect in cellular environment by controlling reactive oxygen species production (ROS), mitochondrial membrane potential (ΔΨm) and even the pro-inflammatory environment. Moreover, the functional effect of the investigated miR, suggested its close connection to the modulation of Sirt-1-PGC1-α pathway, a master switch metabolic route that controlls cellular energetic metabolism. Our assays also suggested a synergistic effect of this miR-1914-5p in cell metabolism, which should be considered as a strong candidate to control steatotic environment in future clinical trials.
Collapse
Affiliation(s)
- Thais Porto-Barbosa
- Departamento de Biologia Geral e Aplicada, Instituto de Biociências, Universidade Estadual Paulista “Júlio de Mesquita Filho”—Campus Rio Claro, Rio Claro, SP, Brazil
- Instituto de Química, Universidade Estadual Paulista “Júlio de Mesquita Filho”–Graduate Program in Biotecnology, Campus Araraquara, Araraquara, SP, Brazil
| | - Letícia Ferreira Ramos
- Departamento de Biologia Geral e Aplicada, Instituto de Biociências, Universidade Estadual Paulista “Júlio de Mesquita Filho”—Campus Rio Claro, Rio Claro, SP, Brazil
- Instituto de Biociências, Universidade Estadual Paulista “Júlio de Mesquita Filho”–Graduate Program in Cellular, Molecular and Microbiology, Campus Rio Claro, Rio Claro SP, Brazil
| | - Camila Cristiane Pansa
- Departamento de Biologia Geral e Aplicada, Instituto de Biociências, Universidade Estadual Paulista “Júlio de Mesquita Filho”—Campus Rio Claro, Rio Claro, SP, Brazil
- Instituto de Biociências, Universidade Estadual Paulista “Júlio de Mesquita Filho”–Graduate Program in Cellular, Molecular and Microbiology, Campus Rio Claro, Rio Claro SP, Brazil
| | - Letícia Ramos Molica
- Departamento de Biologia Geral e Aplicada, Instituto de Biociências, Universidade Estadual Paulista “Júlio de Mesquita Filho”—Campus Rio Claro, Rio Claro, SP, Brazil
- Instituto de Química, Universidade Estadual Paulista “Júlio de Mesquita Filho”–Graduate Program in Biotecnology, Campus Araraquara, Araraquara, SP, Brazil
| | - Osmar Malaspina
- Departamento de Biologia Geral e Aplicada, Instituto de Biociências, Universidade Estadual Paulista “Júlio de Mesquita Filho”—Campus Rio Claro, Rio Claro, SP, Brazil
- Instituto de Biociências, Universidade Estadual Paulista “Júlio de Mesquita Filho”–Graduate Program in Cellular, Molecular and Microbiology, Campus Rio Claro, Rio Claro SP, Brazil
| | - Karen C. M. Moraes
- Departamento de Biologia Geral e Aplicada, Instituto de Biociências, Universidade Estadual Paulista “Júlio de Mesquita Filho”—Campus Rio Claro, Rio Claro, SP, Brazil
- Instituto de Química, Universidade Estadual Paulista “Júlio de Mesquita Filho”–Graduate Program in Biotecnology, Campus Araraquara, Araraquara, SP, Brazil
- Instituto de Biociências, Universidade Estadual Paulista “Júlio de Mesquita Filho”–Graduate Program in Cellular, Molecular and Microbiology, Campus Rio Claro, Rio Claro SP, Brazil
| |
Collapse
|
11
|
Zhao W, Shen Y, Bao Y, Monroig Ó, Zhu T, Sun P, Tocher DR, Zhou Q, Jin M. Fucoidan alleviates hepatic lipid deposition by modulating the Perk-Eif2α-Atf4 axis via Sirt1 activation in Acanthopagrus schlegelii. Int J Biol Macromol 2024; 282:137266. [PMID: 39505163 DOI: 10.1016/j.ijbiomac.2024.137266] [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: 06/06/2024] [Revised: 11/03/2024] [Accepted: 11/04/2024] [Indexed: 11/08/2024]
Abstract
With the increasing use of high-fat diets (HFD), fatty liver disease has become common in fish, and fucoidan is of interest as a natural sulfated polysaccharide with lipid-lowering activity. To explore the molecular regulatory mechanisms of fucoidan's alleviation of HFD-induced lipid deposition in liver, black seabream (Acanthopagrus schlegelii) was used to construct in vivo and in vitro HFD models. In vivo HFD stimulated the protein kinase RNA-like endoplasmic reticulum kinase (Perk) pathway, and up-regulated proliferator-activated receptor gamma (Pparγ) nuclear translocation and expression of lipogenic genes, while it down-regulated Ppar alpha (Pparα) nuclear translocation and expression of lipolytic genes. However, fucoidan reversed these effects of HFD and significantly alleviated HFD-induced lipid accumulation in liver. Moreover, after sirtuin 1 (sirt1) knockdown, these effects of fucoidan disappeared. In the in vitro HFD model, GSK2606414 (GSK)-specific inhibition of the Perk pathway, decreased Pparγ nuclear translocation and increased Pparα nuclear translocation. Overall, fucoidan mitigated HFD-induced, Perk pathway-mediated lipid deposition in the liver of black seabream by activating Sirt1. The findings provided a new prospect for the application of green polysaccharides in aquatic animal feeds.
Collapse
Affiliation(s)
- Wenli Zhao
- Laboratory of Fish and Shellfish Nutrition, School of Marine Sciences, Ningbo University, Ningbo 315211, China; Key Laboratory of Aquacultural Biotechnology Ministry of Education, Ningbo University, Ningbo 315211, China; Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural, Ningbo University, Ningbo 315211, China
| | - Yuedong Shen
- Laboratory of Fish and Shellfish Nutrition, School of Marine Sciences, Ningbo University, Ningbo 315211, China; Key Laboratory of Aquacultural Biotechnology Ministry of Education, Ningbo University, Ningbo 315211, China; Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural, Ningbo University, Ningbo 315211, China
| | - Yangguang Bao
- Laboratory of Fish and Shellfish Nutrition, School of Marine Sciences, Ningbo University, Ningbo 315211, China; Key Laboratory of Aquacultural Biotechnology Ministry of Education, Ningbo University, Ningbo 315211, China; Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural, Ningbo University, Ningbo 315211, China
| | - Óscar Monroig
- Instituto de Acuicultura Torre de la Sal (IATS), CSIC, 12595 Ribera de Cabanes, Castellon, Spain
| | - Tingting Zhu
- Laboratory of Fish and Shellfish Nutrition, School of Marine Sciences, Ningbo University, Ningbo 315211, China; Key Laboratory of Aquacultural Biotechnology Ministry of Education, Ningbo University, Ningbo 315211, China; Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural, Ningbo University, Ningbo 315211, China
| | - Peng Sun
- Laboratory of Fish and Shellfish Nutrition, School of Marine Sciences, Ningbo University, Ningbo 315211, China; Key Laboratory of Aquacultural Biotechnology Ministry of Education, Ningbo University, Ningbo 315211, China; Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural, Ningbo University, Ningbo 315211, China
| | - Douglas R Tocher
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, Shantou University, Shantou 515063, China
| | - Qicun Zhou
- Laboratory of Fish and Shellfish Nutrition, School of Marine Sciences, Ningbo University, Ningbo 315211, China; Key Laboratory of Aquacultural Biotechnology Ministry of Education, Ningbo University, Ningbo 315211, China; Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural, Ningbo University, Ningbo 315211, China
| | - Min Jin
- Laboratory of Fish and Shellfish Nutrition, School of Marine Sciences, Ningbo University, Ningbo 315211, China; Key Laboratory of Aquacultural Biotechnology Ministry of Education, Ningbo University, Ningbo 315211, China; Key Laboratory of Green Mariculture (Co-construction by Ministry and Province), Ministry of Agriculture and Rural, Ningbo University, Ningbo 315211, China.
| |
Collapse
|
12
|
Zhou R, Barnes K, Gibson S, Fillmore N. Dual-edged role of SIRT1 in energy metabolism and cardiovascular disease. Am J Physiol Heart Circ Physiol 2024; 327:H1162-H1173. [PMID: 39269450 DOI: 10.1152/ajpheart.00001.2024] [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: 01/01/2024] [Revised: 08/13/2024] [Accepted: 09/06/2024] [Indexed: 09/15/2024]
Abstract
Regulation of energy metabolism is pivotal in the development of cardiovascular diseases. Dysregulation in mitochondrial fatty acid oxidation has been linked to cardiac lipid accumulation and diabetic cardiomyopathy. Sirtuin 1 (SIRT1) is a deacetylase that regulates the acetylation of various proteins involved in mitochondrial energy metabolism. SIRT1 mediates energy metabolism by directly and indirectly affecting multiple aspects of mitochondrial processes, such as mitochondrial biogenesis. SIRT1 interacts with essential mitochondrial energy regulators such as peroxisome proliferator-activated receptor-α (PPARα), PPARγ coactivator-1α, estrogen-related receptor-α, and their downstream targets. Apart from that, SIRT1 regulates additional proteins, including forkhead box protein O1 and AMP-activated protein kinase in cardiac disease. Interestingly, studies have also shown that the expression of SIRT1 plays a dual-edged role in energy metabolism. Depending on the physiological state, SIRT1 expression can be detrimental or protective. This review focuses on the molecular pathways through which SIRT1 regulates energy metabolism in cardiovascular diseases. We will review SIRT1 and discuss its role in cardiac energy metabolism and its benefits and detrimental effects in heart disease.
Collapse
Affiliation(s)
- Redemptor Zhou
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, North Dakota, United States
| | - Kaleb Barnes
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, North Dakota, United States
| | - Savannah Gibson
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, North Dakota, United States
| | - Natasha Fillmore
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, North Dakota, United States
| |
Collapse
|
13
|
Wang X, Zhang C, Li R, Qiu Y, Ma Y, Wang S, Li Y, Guo S, Li C. Down-regulation of miR-29 improves lipid metabolism in fatty liver of dairy cows. Anim Biotechnol 2024; 35:2396414. [PMID: 39205627 DOI: 10.1080/10495398.2024.2396414] [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: 01/15/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
In this study, we conducted a thorough investigation into the mechanisms by which miR-29 influences lipid metabolism. Thirty-two cows were selected and categorized into distinct groups based on their liver triglyceride (TG) content: healthy, mild fatty liver, and moderate fatty liver groups. Dairy cows with moderate fatty liver showed higher levels of hepatic lipid accumulation, MDA content and serum AST, ALT and ALP contents and lower hepatic catalase CAT and SOD activities. Subsequently, hepatocytes isolated from healthy calves were exposed to sodium oleate (SO) in the presence or absence of pre-incubation with miR-29 inhibitor or inhibitor NC. Pre-transfection with miR-29 inhibitor resulted in reduced hepatocyte lipid accumulation and MDA levels, as well as decreased levels of AST, ALT, and ALP in the supernatant. In the miR-29 inhibitor + SO group, there was an increase in the expression of SREBP-1, FAS, SCD1, and Sirt1. Meanwhile, the expression of PPARα, CPT1, CPT2, PGC-1α, NRF-1, UCP2, and miR-29 were observed to be decreased. In comparison to the miR-29 inhibitor + SO group, some of the measured indicators showed partial reversal in the miR-29 inhibitor + siSirt1 + SO group. Collectively, these findings provide evidence that miR-29 may play a crucial role in the pathogenesis of fatty liver in dairy cows.
Collapse
Affiliation(s)
- Xueying Wang
- Henan International Joint Laboratory of Animal Welfare and Health Breeding, Henan University of Science and Technology, Luoyang, China
| | - Cai Zhang
- Henan International Joint Laboratory of Animal Welfare and Health Breeding, Henan University of Science and Technology, Luoyang, China
| | - Rishun Li
- Henan International Joint Laboratory of Animal Welfare and Health Breeding, Henan University of Science and Technology, Luoyang, China
| | - Yan Qiu
- Henan International Joint Laboratory of Animal Welfare and Health Breeding, Henan University of Science and Technology, Luoyang, China
| | - Yanbo Ma
- Henan International Joint Laboratory of Animal Welfare and Health Breeding, Henan University of Science and Technology, Luoyang, China
- Longmen Laboratory, Innovative Research Team of Livestock Intelligent Breeding and Equipment, Luoyang, China
| | - Shuai Wang
- Henan International Joint Laboratory of Animal Welfare and Health Breeding, Henan University of Science and Technology, Luoyang, China
| | - Yuanxiao Li
- Henan International Joint Laboratory of Animal Welfare and Health Breeding, Henan University of Science and Technology, Luoyang, China
| | - Shuai Guo
- Henan International Joint Laboratory of Animal Welfare and Health Breeding, Henan University of Science and Technology, Luoyang, China
| | - Chenxu Li
- State Key Laboratory for Zoonotic Diseases, Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun Jilin, China
| |
Collapse
|
14
|
Ho PY, Lin PX, Koh YC, Lin WS, Tang KL, Chen YH, Weerawatanakorn M, Pan MH. Exploring the Effects of Whole Food-Based Dragon Fruit on Metabolic Disorders in High-Fat Diet-Induced Mice. Mol Nutr Food Res 2024; 68:e2400604. [PMID: 39363653 DOI: 10.1002/mnfr.202400604] [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] [Revised: 09/12/2024] [Indexed: 10/05/2024]
Abstract
SCOPE Metabolic syndrome (MetS) significantly contributes to premature mortality, with obesity being a major risk factor. Dragon fruit, cultivated globally, exhibits bioactivity in preventing obesity-related diseases. Traditional studies using organic solvents for extraction do not align with actual consumption patterns. METHOD AND RESULTS This study evaluates whole red dragon fruit's effectiveness in ameliorating metabolic disorders using a high-fat diet-induced obesity model in mice for 20 weeks. The experimental groups include the supernatant (RS), precipitate (RP), and pomace (PO) of red dragon fruit juice, compared to the supernatant of white dragon fruit juice (WS). The study finds that dragon fruit extracts reduced adipose tissue weight, body fat percentage, pro-inflammatory cytokines, and improved blood lipid profiles. RP is the most effective, reducing body weight by 4.33 g, improving lipid metabolism and glucose homeostasis, and altering gut microbiota to enhance beneficial bacteria and short-chain fatty acids. RP's efficacy in preventing MetS and obesity is attributed to its bioactive components. CONCLUSION These findings advocate for using whole fruits in developing functional products, amplifying the agricultural economic value of red dragon fruit.
Collapse
Affiliation(s)
- Pin-Yu Ho
- Institute of Food Science and Technology, National Taiwan University, Taipei, 10617, Taiwan, ROC
| | - Pin-Xuan Lin
- Institute of Food Science and Technology, National Taiwan University, Taipei, 10617, Taiwan, ROC
| | - Yen-Chun Koh
- Institute of Food Science and Technology, National Taiwan University, Taipei, 10617, Taiwan, ROC
| | - Wei-Sheng Lin
- Institute of Food Science and Technology, National Taiwan University, Taipei, 10617, Taiwan, ROC
- Department of Food Science, National Quemoy University, Quemoy County, 89250, Taiwan, ROC
| | - Kai-Liang Tang
- Taichung District Agricultural Research and Extension station, Ministry of Agriculture, Songhuai Road, Dacun Township, Changhua County, 515008, Taiwan, ROC
| | - Yu-Hsin Chen
- Taichung District Agricultural Research and Extension station, Ministry of Agriculture, Songhuai Road, Dacun Township, Changhua County, 515008, Taiwan, ROC
| | - Monthana Weerawatanakorn
- Department of Agro-Industry, Naresuan University, 99 Moo 9, Thapho, Muang, Phitsanulok, 65000, Thailand
| | - Min-Hsiung Pan
- Institute of Food Science and Technology, National Taiwan University, Taipei, 10617, Taiwan, ROC
- Department of Public Health, China Medical University, Taichung, 40402, Taiwan, ROC
- Department of Food Nutrition and Health Biotechnology, Asia University, Taichung, 41354, Taiwan, ROC
| |
Collapse
|
15
|
Babula JJ, Bui D, Stevenson HL, Watowich SJ, Neelakantan H. Nicotinamide N-methyltransferase inhibition mitigates obesity-related metabolic dysfunction. Diabetes Obes Metab 2024; 26:5272-5282. [PMID: 39161060 PMCID: PMC11622326 DOI: 10.1111/dom.15879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/29/2024] [Accepted: 08/01/2024] [Indexed: 08/21/2024]
Abstract
AIM To assess the effects of a small-molecule nicotinamide N-methyltransferase (NNMT) inhibitor, 5A1MQ, on body composition, metabolic variables, fatty liver pathologies, and circulating biomarkers in diet-induced obese (DIO) mice, and characterize its plasma pharmacokinetics (PK) and tissue distribution in vivo. MATERIALS AND METHODS DIO mice were administered vehicle or 5A1MQ once daily for 28 days. Longitudinal measures of body composition, blood glucose and plasma insulin levels, and terminal measures of liver histopathology and serum markers, were evaluated. Plasma and tissue PK were established in age- and strain-matched mice after intravenous, oral, and subcutaneous dosing of 5A1MQ. RESULTS 5A1MQ treatment dose-dependently limited body weight and fat mass gains, improved oral glucose tolerance and insulin sensitivity, and suppressed hyperinsulinaemia in DIO mice. Liver histology from 5A1MQ-treated DIO mice showed attenuated hepatic steatosis and macrophage infiltration, and correspondingly reduced liver weight, size, and triglyceride levels. 5A1MQ treatment normalized circulating levels of alanine transaminase, aspartate transaminase, and ketone bodies, supporting an overall improvement in liver and metabolic functions. The pharmacodynamic effects of 5A1MQ were further corroborated by its high systemic exposure and effective distribution to metabolically active tissues, including adipose, muscle and liver, following subcutaneous dosing of mice. CONCLUSIONS This work validates NNMT inhibition as a viable pharmacological approach to ameliorate metabolic imbalances and improve liver pathologies that develop with obesity.
Collapse
Affiliation(s)
| | - Dinh Bui
- Ridgeline Therapeutics, Houston, TX, USA
| | - Heather L. Stevenson
- Department of Pathology, University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Stanley J. Watowich
- Ridgeline Therapeutics, Houston, TX, USA
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | | |
Collapse
|
16
|
Gan L, Guo H, Yang Q, Zhou X, Xie Y, Ma X, Gou L, Fang J, Zuo Z. Alkaline Mineral Complex Water Attenuates Transportation-Induced Hepatic Lipid Metabolism Dysregulation by AMPKα-SREBP-1c/PPARα Pathways. Int J Mol Sci 2024; 25:11373. [PMID: 39518926 PMCID: PMC11545688 DOI: 10.3390/ijms252111373] [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: 09/19/2024] [Revised: 10/19/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Transportation, an unavoidable process in livestock farming, causes metabolic disorders in the body, which then lead to endocrine disruption, being immunocompromised, and growth suppression. Lipid metabolism dysregulation is a critical phenotype induced by transportation. The liver is a vital organ in lipid metabolism, with a role in both lipid synthesis and lipolysis. However, the specific mechanisms by which transportation affects hepatic lipid metabolism remain unclear. This study employed rats as a model to investigate the effects of transportation on hepatic lipid metabolism. Rats subjected to transportation showed altered serum lipid profiles, including decreased serum triglyceride (TG), low-density lipoprotein cholesterol (VLDL-C), and non-esterified fatty acid (NEFA) immediately after transportation (IAT) and serum total cholesterol (TC) on day 3, and increasing serum TG, TC, and low-density lipoprotein cholesterol (LDL-C) on day 10. Meanwhile, fatty droplets in the liver were also reduced at IAT and increased on days 3 and 10. Notably, transportation also affected hepatic-lipid-metabolism-related enzyme activities and signaling pathways, such as increased AMP-activated protein kinase alpha (AMPKα) phosphorylation and modulations in key proteins and genes related to lipid metabolism, decreased hepatic acetyl-CoA carboxylase (ACC) and fatty acid synthase (FAS) activities at IAT, and increased carnitine palmitoyl transferase 1 alpha (CPT-1α) at IAT and ACC and CPT-1α activities on days 3 and 10. Supplementation with alkaline mineral complex water (AMC) before and after transportation mitigated the adverse effects on hepatic lipid metabolism by modulating the AMPKα-SREBP-1c/PPARα pathway, enhancing lipid synthesis, and reducing the oxidative catabolism of fatty acids. AMC inhibited the transportation-induced activation of AMPKα and restored the balance of lipid-metabolism-related enzymes and pathways. These findings highlight AMC's potential as a therapeutic intervention to alleviate transportation-induced lipid metabolism disorders, offering significant implications for improving animal welfare and reducing economic losses in livestock farming.
Collapse
Affiliation(s)
- Linli Gan
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China; (L.G.); (H.G.); (X.Z.); (Y.X.); (X.M.); (L.G.); (J.F.)
| | - Hongrui Guo
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China; (L.G.); (H.G.); (X.Z.); (Y.X.); (X.M.); (L.G.); (J.F.)
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu 611130, China
| | - Qiyuan Yang
- Sichuan Academy of Grassland Sciences, Chengdu 611731, China;
| | - Xueke Zhou
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China; (L.G.); (H.G.); (X.Z.); (Y.X.); (X.M.); (L.G.); (J.F.)
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu 611130, China
| | - Yue Xie
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China; (L.G.); (H.G.); (X.Z.); (Y.X.); (X.M.); (L.G.); (J.F.)
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu 611130, China
| | - Xiaoping Ma
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China; (L.G.); (H.G.); (X.Z.); (Y.X.); (X.M.); (L.G.); (J.F.)
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu 611130, China
| | - Liping Gou
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China; (L.G.); (H.G.); (X.Z.); (Y.X.); (X.M.); (L.G.); (J.F.)
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu 611130, China
| | - Jing Fang
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China; (L.G.); (H.G.); (X.Z.); (Y.X.); (X.M.); (L.G.); (J.F.)
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu 611130, China
| | - Zhicai Zuo
- College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu 611130, China; (L.G.); (H.G.); (X.Z.); (Y.X.); (X.M.); (L.G.); (J.F.)
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agriculture University, Wenjiang, Chengdu 611130, China
| |
Collapse
|
17
|
Ulusan M, Erdogan MA, Simsek O, Gunes V, Erbas O. Saccharomyces boulardii Mitigates Fructose-Induced Non-Alcoholic Fatty Liver in Rats. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1713. [PMID: 39459500 PMCID: PMC11509347 DOI: 10.3390/medicina60101713] [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: 09/12/2024] [Revised: 10/03/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024]
Abstract
Background and Objectives: Non-alcoholic fatty liver disease (NAFLD) is a growing global health concern closely linked to metabolic disorders, including obesity, insulin resistance, and dyslipidemia. Emerging evidence suggests that the gut-liver axis plays a critical role in the pathogenesis of NAFLD, with recent research highlighting the influence of gut microbiota, including fungal species such as Saccharomyces boulardii (S. boulardii). This study aimed to evaluate the effects of S. boulardii on lipid metabolism and oxidative stress in a rat model of fructose-induced NAFLD. Materials and Methods: Thirty Wistar rats were divided into three groups: a control group, a fatty liver group induced by 35% fructose supplementation, and a treatment group receiving S. boulardii (100 mg/kg/day) after fructose induction. Results: Biochemical analyses revealed that the treatment group exhibited significantly lower plasma levels of malondialdehyde (MDA), alanine aminotransferase (ALT), total triglycerides, and cholesterol compared to the untreated fatty liver group (p < 0.05). Furthermore, liver tissue analysis showed a marked reduction in lipid accumulation and fatty infiltration in the treatment group, with no visible lipid vacuoles in hepatocytes. The expression of aquaporin-8 (AQP8) and sirtuin-1 (SIRT1), key markers associated with hepatocyte function and lipid metabolism, was significantly higher in the S. boulardii group compared to the fatty liver group (p < 0.001). Conclusions: These findings indicate that S. boulardii supplementation mitigates the metabolic and oxidative stress-related alterations associated with fructose-induced NAFLD. In conclusion, our study suggests that S. boulardii exerts protective effects on the liver by reducing lipid accumulation and oxidative stress, highlighting its potential as a therapeutic intervention for NAFLD.
Collapse
Affiliation(s)
- Mehmet Ulusan
- Department of Internal Medicine, Faculty of Veterinary Medicine, Erciyes University, Kayseri 38280, Turkey; (M.U.); (V.G.)
- Department of Internal Medicine, Faculty of Veterinary Medicine, Burdur Mehmet Akif Ersoy University, Burdur 15030, Turkey
| | - Mumin Alper Erdogan
- Department of Physiology, Faculty of Medicine, Izmir Katip Celebi University, Izmir 35620, Turkey;
| | - Ozkan Simsek
- Department of Physiology, Faculty of Veterinary Medicine, Burdur Mehmet Akif Ersoy University, Burdur 15030, Turkey
| | - Vehbi Gunes
- Department of Internal Medicine, Faculty of Veterinary Medicine, Erciyes University, Kayseri 38280, Turkey; (M.U.); (V.G.)
- Experimental Research and Application Center (DEKAM), Erciyes University, Kayseri 38280, Turkey
| | - Oytun Erbas
- Department of Physiology, Faculty of Medicine, Istanbul Demiroglu Bilim University, Istanbul 34394, Turkey;
| |
Collapse
|
18
|
Kim B, Ronaldo R, Kweon BN, Yoon S, Park Y, Baek JH, Lee JM, Hyun CK. Mesenchymal Stem Cell-Derived Exosomes Attenuate Hepatic Steatosis and Insulin Resistance in Diet-Induced Obese Mice by Activating the FGF21-Adiponectin Axis. Int J Mol Sci 2024; 25:10447. [PMID: 39408777 PMCID: PMC11476820 DOI: 10.3390/ijms251910447] [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: 09/06/2024] [Revised: 09/26/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
Exosomes derived from mesenchymal stem cells have shown promise in treating metabolic disorders, yet their specific mechanisms remain largely unclear. This study investigates the protective effects of exosomes from human umbilical cord Wharton's jelly mesenchymal stem cells (hWJMSCs) against adiposity and insulin resistance in high-fat diet (HFD)-induced obese mice. HFD-fed mice treated with hWJMSC-derived exosomes demonstrated improved gut barrier integrity, which restored immune balance in the liver and adipose tissues by reducing macrophage infiltration and pro-inflammatory cytokine expression. Furthermore, these exosomes normalized lipid metabolism including lipid oxidation and lipogenesis, which alleviate lipotoxicity-induced endoplasmic reticulum (ER) stress, thereby decreasing fat accumulation and chronic tissue inflammation in hepatic and adipose tissues. Notably, hWJMSC-derived exosomes also promoted browning and thermogenic capacity of adipose tissues, which was linked to reduced fibroblast growth factor 21 (FGF21) resistance and increased adiponectin production. This process activated the AMPK-SIRT1-PGC-1α pathway, highlighting the role of the FGF21-adiponectin axis. Our findings elucidate the molecular mechanisms through which hWJMSC-derived exosomes counteract HFD-induced metabolic dysfunctions, supporting their potential as therapeutic agents for metabolic disorders.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Chang-Kee Hyun
- School of Life Science, Handong Global University, Pohang 37554, Gyungbuk, Republic of Korea
| |
Collapse
|
19
|
Guo R, Yu K, Huang K, Li J, Huang J, Yang X, Wu Y, Wang D. Regulatory mechanism of Sarmentosin and Quercetin on lipid accumulation in primary hepatocyte of GIFT tilapia (Oreochromis niloticus) with fatty liver. PLoS One 2024; 19:e0309976. [PMID: 39236049 PMCID: PMC11376590 DOI: 10.1371/journal.pone.0309976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 08/18/2024] [Indexed: 09/07/2024] Open
Abstract
Sarmentosin (SA) and Quercetin (QC) are two active components of Sedum Sarmentosum Bunge, which is a traditional Chinese herbal medicine. This study aimed to investigate the role and regulatory mechanism of SA and QC in fatty liver of Genetic Improvement of Farmed Tilapia (GIFT) tilapia. GIFT tilapia were randomly divided into two groups with three replicates per treatment (30 fish in each replicate): normal diet group (average weight 3.51±0.31 g) and high-fat diet group (average weight 3.44±0.09 g). After 8 weeks feeding trial, growth index, lipid deposition, and biochemical indexes were measured. Lipid deposition, and lipid and inflammation-related gene expression were detected in a primary hepatocyte model of fatty liver of GIFT tilapia treated with SA or QC. Our results showed that high-fat diet caused lipid deposition and peroxidative damage in the liver of GIFT tilapia. The cell counting kit-8 assay results indicated that 10 μM SA and 10 μM of QC both had the least effect on hepatocyte proliferation. Moreover, both 10 μM of SA and 10 μM of QC showed lipolytic effects and inhibited the expression of lipid-related genes (FAS, Leptin, SREBP-1c, and SREBP2) in fatty liver cells. Interestingly, QC induced autophagosome-like subcellular structure and increased the expression of IL-8 in fatty liver cells. In conclusion, this study confirmed that SA and QC improved fatty liver caused by high-fat diet, providing a novel therapeutic approach for fatty liver of GIFT tilapia.
Collapse
Affiliation(s)
- Ruijie Guo
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Kai Yu
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Kai Huang
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Jinghua Li
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Jiao Huang
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Xuhong Yang
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Yaoting Wu
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Dandan Wang
- College of Animal Science and Technology, Guangxi University, Nanning, China
| |
Collapse
|
20
|
Khatoon S, Das N, Chattopadhyay S, Joharapurkar A, Singh A, Patel V, Nirwan A, Kumar A, Mugale MN, Mishra DP, Kumaravelu J, Guha R, Jain MR, Chattopadhyay N, Sanyal S. Apigenin-6-C-glucoside ameliorates MASLD in rodent models via selective agonism of adiponectin receptor 2. Eur J Pharmacol 2024; 978:176800. [PMID: 38950835 DOI: 10.1016/j.ejphar.2024.176800] [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: 01/30/2024] [Revised: 06/13/2024] [Accepted: 06/28/2024] [Indexed: 07/03/2024]
Abstract
Adiponectin plays key roles in energy metabolism and ameliorates inflammation, oxidative stress, and mitochondrial dysfunction via its primary receptors, adiponectin receptors -1 and 2 (AdipoR1 and AdipoR2). Systemic depletion of adiponectin causes various metabolic disorders, including MASLD; however adiponectin supplementation is not yet achievable owing to its large size and oligomerization-associated complexities. Small-molecule AdipoR agonists, thus, may provide viable therapeutic options against metabolic disorders. Using a novel luciferase reporter-based assay here, we have identified Apigenin-6-C-glucoside (ACG), but not apigenin, as a specific agonist for the liver-rich AdipoR isoform, AdipoR2 (EC50: 384 pM) with >10000X preference over AdipoR1. Immunoblot analysis in HEK-293 overexpressing AdipoR2 or HepG2 and PLC/PRF/5 liver cell lines revealed rapid AMPK, p38 activation and induction of typical AdipoR targets PGC-1α and PPARα by ACG at a pharmacologically relevant concentration of 100 nM (reported cMax in mouse; 297 nM). ACG-mediated AdipoR2 activation culminated in a favorable modulation of key metabolic events, including decreased inflammation, oxidative stress, mitochondrial dysfunction, de novo lipogenesis, and increased fatty acid β-oxidation as determined by immunoblotting, QRT-PCR and extracellular flux analysis. AdipoR2 depletion or AMPK/p38 inhibition dampened these effects. The in vitro results were recapitulated in two different murine models of MASLD, where ACG at 10 mg/kg body weight robustly reduced hepatic steatosis, fibrosis, proinflammatory macrophage numbers, and increased hepatic glycogen content. Together, using in vitro experiments and rodent models, we demonstrate a proof-of-concept for AdipoR2 as a therapeutic target for MASLD and provide novel chemicobiological insights for the generation of translation-worthy pharmacological agents.
Collapse
Affiliation(s)
- Shamima Khatoon
- Division of Biochemistry and Structural Biology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Nabanita Das
- Division of Biochemistry and Structural Biology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Sourav Chattopadhyay
- Division of Biochemistry and Structural Biology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | | | - Abhinav Singh
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India; Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Vishal Patel
- Zydus Research Center, Moraiya, Ahmedabad, 382213, Gujarat, India
| | - Abhishek Nirwan
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India; Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Akhilesh Kumar
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Madhav Nilakanth Mugale
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India; Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Durga Prasad Mishra
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India; Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Jagavelu Kumaravelu
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India; Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Rajdeep Guha
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India; Laboratory Animal Facility, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | | | - Naibedya Chattopadhyay
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India; Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Sabyasachi Sanyal
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
21
|
Martín Barraza JI, Bars-Cortina D. Dietary Pattern's Role in Hepatic Epigenetic and Dietary Recommendations for the Prevention of NAFLD. Nutrients 2024; 16:2956. [PMID: 39275272 PMCID: PMC11396970 DOI: 10.3390/nu16172956] [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] [Revised: 08/25/2024] [Accepted: 08/29/2024] [Indexed: 09/16/2024] Open
Abstract
NAFLD has emerged as a significant public health concern, with its prevalence increasing globally. Emphasizing the complex relationship between dietary patterns and epigenetic modifications such as DNA methylation or miRNA expression can exert a positive impact on preventing and managing metabolic disorders, including NAFLD, within the 2030 Sustainable Development Goals. This review aims to evaluate the influence of dietary patterns on hepatic epigenetic gene modulation and provide dietary recommendations for the prevention and management of NAFLD in the general population. METHODS Comprehensive screening and eligibility criteria identified eleven articles focusing on epigenetic changes in NAFLD patients through dietary modifications or nutrient supplementation. RESULTS AND DISCUSSION Data were organized based on study types, categorizing them into evaluations of epigenetic changes in NAFLD patients through dietary pattern modifications or specific nutrient intake. CONCLUSIONS The study highlights the importance of dietary interventions in managing and preventing NAFLD, emphasizing the potential of dietary patterns to influence hepatic epigenetic gene modulation. This study provides valuable insights and recommendations to mitigate the risk of developing NAFLD: (i) eat a primarily plant-based diet; (ii) increase consumption of high-fiber foods; (iii) consume more polyunsaturated and monounsaturated fatty acids; (iv) limit processed foods, soft drinks, added sugars, and salt; and (v) avoid alcohol.
Collapse
Affiliation(s)
| | - David Bars-Cortina
- Oncology Data Analytics Program (ODAP), Unit of Biomarkers and Susceptibility (UBS), Catalan Institute of Oncology (ICO), L'Hospitalet del Llobregat, 08908 Barcelona, Spain
- ONCOBELL Program, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, 08908 Barcelona, Spain
- Department of Health Sciences, Universitat Oberta de Catalunya, 08018 Barcelona, Spain
| |
Collapse
|
22
|
Ashiqueali SA, Zhu X, Wiesenborn DS, Gesing A, Schneider A, Noureddine SA, Correa-Garcia CG, Masternak MM, Siddiqi SA. Calorie restriction and life-extending mutation downregulate miR-34a to facilitate lipid metabolism in the liver. Exp Gerontol 2024; 194:112506. [PMID: 38945410 PMCID: PMC11418173 DOI: 10.1016/j.exger.2024.112506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 06/26/2024] [Accepted: 06/27/2024] [Indexed: 07/02/2024]
Abstract
Ames dwarf mice (df/df) display delayed aging relative to their normal (N) siblings, living approximately 40-60 % longer. As such, investigating the mechanisms that enable these organisms to have extended lifespan is useful for the development of interventions to slow aging and deter age-related disease. Nonalcoholic fatty liver disease (NAFLD) is a condition that is characterized by the accumulation of excess adipose tissue in the liver. Previous studies highlight the potential of calorie restriction (CR) in promoting longevity, but little is known about its effects on the biomolecular processes that govern NAFLD. In this study, we examined the role of 6-month CR on genes regulating lipid metabolism in the livers of long-living df/df mice and their N littermates. Importantly, our findings showed significant downregulation of miR-34a-5p in N-CR mice and df/df mice regardless of dietary regimen. Alongside, our RT-PCR results indicated that downregulation of miR-34a-5p is correlated with the expression of metabolism-associated mRNAs involved in modulating the processes of de novo lipogenesis (DNL), fatty acid oxidation (FAO), very-low density lipoprotein transport (VLDL-T), and reverse cholesterol transport (RCT). To further verify the role of miR-34a-5p in regulating metabolic processes, we transfected the human liver cancer (HepG2) cell line with miR-34a mimic, and studied its effect on direct targets Sirt1, Ampk, and Ppara as well as downstream lipid transport regulating genes. Our findings suggest that CR and df/df life extending mutation are robust drivers of the miR-34a-5p signaling pathway and prevent the pathogenesis of age-related diseases by improving overall lipid homeostasis.
Collapse
Affiliation(s)
- Sarah A Ashiqueali
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Xiang Zhu
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Denise S Wiesenborn
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, USA; Department of Biotechnology, University of Applied Sciences Kaiserslautern, Zweibrücken, Germany
| | - Adam Gesing
- Department of Endocrinology of Ageing, Medical University of Lodz, Poland
| | - Augusto Schneider
- Department of Nutrition, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Sarah A Noureddine
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Christian G Correa-Garcia
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, USA; Department of Medicine, San Juan Bautista School of Medicine, Caguas, Puerto Rico
| | - Michal M Masternak
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, USA; Department of Head and Neck Surgery, Poznan University of Medical Sciences, 61-701 Poznan, Poland
| | - Shadab A Siddiqi
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, USA.
| |
Collapse
|
23
|
Henin G, Loumaye A, Deldicque L, Leclercq IA, Lanthier N. Unlocking liver health: Can tackling myosteatosis spark remission in metabolic dysfunction-associated steatotic liver disease? Liver Int 2024; 44:1781-1796. [PMID: 38623714 DOI: 10.1111/liv.15938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 03/12/2024] [Accepted: 04/02/2024] [Indexed: 04/17/2024]
Abstract
Myosteatosis is highly prevalent in metabolic dysfunction-associated steatotic liver disease (MASLD) and could reciprocally impact liver function. Decreasing muscle fat could be indirectly hepatoprotective in MASLD. We conducted a review to identify interventions reducing myosteatosis and their impact on liver function. Non-pharmacological interventions included diet (caloric restriction or lipid enrichment), bariatric surgery and physical activity. Caloric restriction in humans achieving a mean weight loss of 3% only reduces muscle fat. Lipid-enriched diet increases liver fat in human with no impact on muscle fat, except sphingomyelin-enriched diet which reduces both lipid contents exclusively in pre-clinical studies. Bariatric surgery, hybrid training (resistance exercise and electric stimulation) or whole-body vibration in human decrease both liver and muscle fat. Physical activity impacts both phenotypes by reducing local and systemic inflammation, enhancing insulin sensitivity and modulating the expression of key mediators of the muscle-liver-adipose tissue axis. The combination of diet and physical activity acts synergistically in liver, muscle and white adipose tissue, and further decrease muscle and liver fat. Several pharmacological interventions (patchouli alcohol, KBP-089, 2,4-dinitrophenol methyl ether, adipoRon and atglistatin) and food supplementation (vitamin D or resveratrol) improve liver and muscle phenotypes in pre-clinical studies by increasing fatty acid oxidation and anti-inflammatory properties. These interventions are effective in reducing myosteatosis in MASLD while addressing the liver disease itself. This review supports that disturbances in inter-organ crosstalk are key pathophysiological mechanisms involved in MASLD and myosteatosis pathogenesis. Focusing on the skeletal muscle might offer new therapeutic strategies to treat MASLD by modulating the interactions between liver and muscles.
Collapse
Affiliation(s)
- Guillaume Henin
- Service d'Hépato-Gastroentérologie, Cliniques universitaires Saint-Luc, UCLouvain, Brussels, Belgium
- Laboratory of Hepatogastroenterology, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Audrey Loumaye
- Service d'Endocrinologie, Diabétologie et Nutrition, Cliniques universitaires Saint-Luc, UCLouvain, Brussels, Belgium
| | | | - Isabelle A Leclercq
- Laboratory of Hepatogastroenterology, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Nicolas Lanthier
- Service d'Hépato-Gastroentérologie, Cliniques universitaires Saint-Luc, UCLouvain, Brussels, Belgium
- Laboratory of Hepatogastroenterology, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain (UCLouvain), Brussels, Belgium
| |
Collapse
|
24
|
Srinivas AN, Suresh D, Vishwanath PM, Satish S, Santhekadur PK, Koka S, Kumar DP. TACE inhibition: a promising therapeutic intervention against AATF-mediated steatohepatitis to hepatocarcinogenesis. Mol Oncol 2024; 18:1940-1957. [PMID: 38558505 PMCID: PMC11306524 DOI: 10.1002/1878-0261.13646] [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: 09/16/2023] [Revised: 01/03/2024] [Accepted: 03/18/2024] [Indexed: 04/04/2024] Open
Abstract
Metabolic dysfunction-associated steatohepatitis-driven hepatocellular carcinoma (MASH-HCC) is a global clinical challenge for which there is a limited understanding of disease pathogenesis and a subsequent lack of therapeutic interventions. We previously identified that tumor necrosis factor-alpha (TNF-α) upregulated apoptosis antagonizing transcription factor (AATF) in MASH. Here, we investigated the effect of TNF-α converting enzyme (TACE) inhibition as a promising targeted therapy against AATF-mediated steatohepatitis to hepatocarcinogenesis. A preclinical murine model that recapitulates human MASH-HCC was used in the study. C57Bl/6 mice were fed with chow diet normal water (CD) or western diet sugar water (WD) along with a low dose of carbon tetrachloride (CCl4; 0.2 μL·g-1, weekly) for 24 weeks. TACE activity, TNF-α levels, and AATF expression were measured. The mice were treated with the TACE inhibitor Marimastat for 12 weeks, followed by analyses of liver injury, fibrosis, inflammation, and oncogenic signaling. In vitro experiments using stable clones of AATF control and AATF knockdown were also conducted. We found that AATF expression was upregulated in WD/CCl4 mice, which developed severe MASH at 12 weeks and advanced fibrosis with HCC at 24 weeks. WD/CCl4 mice showed increased TACE activity with reduced hepatic expression of sirtuin 1 (Sirt1) and tissue inhibitor of metalloproteinase 3 (Timp3). The involvement of the SIRT1/TIMP3/TACE axis was confirmed by the release of TNF-α, which upregulated AATF, a key molecular driver of MASH-HCC. Interestingly, TACE inhibition by Marimastat reduced liver injury, dyslipidemia, AATF expression, and oncogenic signaling, effectively preventing hepatocarcinogenesis. Furthermore, Marimastat inhibited the activation of JNK, ERK1/2, and AKT, which are key regulators of tumorigenesis in WD/CCl4 mice and in AATF control cells, but had no effect on AATF knockdown cells. This study shows that TACE inhibition prevents AATF-mediated inflammation, fibrosis, and oncogenesis in MASH-HCC, offering a potential target for therapeutic intervention.
Collapse
Affiliation(s)
- Akshatha N. Srinivas
- Department of Biochemistry, CEMR Lab, JSS Medical CollegeJSS Academy of Higher Education and ResearchMysuruIndia
| | - Diwakar Suresh
- Department of Biochemistry, CEMR Lab, JSS Medical CollegeJSS Academy of Higher Education and ResearchMysuruIndia
| | - Prashant M. Vishwanath
- Department of Biochemistry, CEMR Lab, JSS Medical CollegeJSS Academy of Higher Education and ResearchMysuruIndia
| | - Suchitha Satish
- Department of Pathology, JSS Medical College and HospitalJSS Academy of Higher Education and ResearchMysuruIndia
| | - Prasanna K. Santhekadur
- Department of Biochemistry, CEMR Lab, JSS Medical CollegeJSS Academy of Higher Education and ResearchMysuruIndia
| | - Saisudha Koka
- Department of Pharmaceutical Sciences, Irma Lerma Rangel School of PharmacyTexas A&M UniversityKingsvilleTXUSA
| | - Divya P. Kumar
- Department of Biochemistry, CEMR Lab, JSS Medical CollegeJSS Academy of Higher Education and ResearchMysuruIndia
| |
Collapse
|
25
|
Yang J, Félix-Soriano E, Martínez-Gayo A, Ibañez-Santos J, Sáinz N, Martínez JA, Moreno-Aliaga MJ. SIRT1 and FOXO1 role on MASLD risk: effects of DHA-rich n-3 PUFA supplementation and exercise in aged obese female mice and in post-menopausal overweight/obese women. J Physiol Biochem 2024; 80:697-712. [PMID: 39264516 PMCID: PMC11502560 DOI: 10.1007/s13105-024-01044-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 08/07/2024] [Indexed: 09/13/2024]
Abstract
Sirtuins 1 (SIRT1) and Forkhead box protein O1 (FOXO1) expression have been associated with obesity and metabolic dysfunction-associated steatotic liver disease (MASLD). Exercise and/or docosahexaenoic acid (DHA) supplementation have shown beneficial effects on MASLD. The current study aims to assess the relationships between Sirt1, Foxo1 mRNA levels and several MASLD biomarkers, as well as the effects of DHA-rich n-3 PUFA supplementation and/or exercise in the steatotic liver of aged obese female mice, and in peripheral blood mononuclear cells (PBMCs) of postmenopausal women with overweight/obesity. In the liver of 18-month-old mice, Sirt1 levels positively correlated with the expression of genes related to fatty acid oxidation, and negatively correlated with lipogenic and proinflammatory genes. Exercise (long-term treadmill training), especially when combined with DHA, upregulated hepatic Sirt1 mRNA levels. Liver Foxo1 mRNA levels positively associated with hepatic triglycerides (TG) content and the expression of lipogenic and pro-inflammatory genes, while negatively correlated with the lipolytic gene Hsl. In PBMCs of postmenopausal women with overweight/obesity, FOXO1 mRNA expression negatively correlated with the hepatic steatosis index (HSI) and the Zhejiang University index (ZJU). After 16-weeks of DHA-rich PUFA supplementation and/or progressive resistance training (RT), most groups exhibited reduced MASLD biomarkers and risk indexes accompanying with body fat mass reduction, but no significant changes were found between the intervention groups. However, in PBMCs n-3 supplementation upregulated FOXO1 expression, and the RT groups exhibited higher SIRT1 expression. In summary, SIRT1 and FOXO1 could be involved in the beneficial mechanisms of exercise and n-3 PUFA supplementation related to MASLD manifestation.
Collapse
Affiliation(s)
- Jinchunzi Yang
- Center for Nutrition Research and Department of Nutrition, Food Science and Physiology, School of Pharmacy and Nutrition, University of Navarra, 31008, Pamplona, Spain
- Current Address: Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Science, Shenzhen, 518000, China
| | - Elisa Félix-Soriano
- Center for Nutrition Research and Department of Nutrition, Food Science and Physiology, School of Pharmacy and Nutrition, University of Navarra, 31008, Pamplona, Spain
| | - Alejandro Martínez-Gayo
- Center for Nutrition Research and Department of Nutrition, Food Science and Physiology, School of Pharmacy and Nutrition, University of Navarra, 31008, Pamplona, Spain
| | - Javier Ibañez-Santos
- Studies, Research and Sports Medicine Centre (CEIMD), Government of Navarre, 31005, Pamplona, Spain
| | - Neira Sáinz
- Center for Nutrition Research and Department of Nutrition, Food Science and Physiology, School of Pharmacy and Nutrition, University of Navarra, 31008, Pamplona, Spain
| | - J Alfredo Martínez
- Center for Nutrition Research and Department of Nutrition, Food Science and Physiology, School of Pharmacy and Nutrition, University of Navarra, 31008, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029, Madrid, Spain
| | - María J Moreno-Aliaga
- Center for Nutrition Research and Department of Nutrition, Food Science and Physiology, School of Pharmacy and Nutrition, University of Navarra, 31008, Pamplona, Spain.
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029, Madrid, Spain.
- IdISNA, Navarra Institute for Health Research, 31008, Pamplona, Spain.
| |
Collapse
|
26
|
Nie W, Zhu H, Sun X, Zhou J, Xu H, Yu Z, Lu M, Jiang B, Zhou L, Zhou X. Catalpol attenuates hepatic glucose metabolism disorder and oxidative stress in triptolide-induced liver injury by regulating the SIRT1/HIF-1α pathway. Int J Biol Sci 2024; 20:4077-4097. [PMID: 39113710 PMCID: PMC11302874 DOI: 10.7150/ijbs.97362] [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: 04/15/2024] [Accepted: 07/18/2024] [Indexed: 08/10/2024] Open
Abstract
Triptolide (TP), known for its effectiveness in treating various rheumatoid diseases, is also associated with significant hepatotoxicity risks. This study explored Catalpol (CAT), an iridoid glycoside with antioxidative and anti-inflammatory effects, as a potential defense against TP-induced liver damage. In vivo and in vitro models of liver injury were established using TP in combination with different concentrations of CAT. Metabolomics analyses were conducted to assess energy metabolism in mouse livers. Additionally, a Seahorse XF Analyzer was employed to measure glycolysis rate, mitochondrial respiratory functionality, and real-time ATP generation rate in AML12 cells. The study also examined the expression of proteins related to glycogenolysis and gluconeogenesis. Using both in vitro SIRT1 knockout/overexpression and in vivo liver-specific SIRT1 knockout models, we confirmed SIRT1 as a mechanism of action for CAT. Our findings revealed that CAT could alleviate TP-induced liver injury by activating SIRT1, which inhibited lysine acetylation of hypoxia-inducible factor-1α (HIF-1α), thereby restoring the balance between glycolysis and oxidative phosphorylation. This action improved mitochondrial dysfunction and reduced glucose metabolism disorder and oxidative stress caused by TP. Taken together, these insights unveil a hitherto undocumented mechanism by which CAT ameliorates TP-induced liver injury, positioning it as a potential therapeutic agent for managing TP-induced hepatotoxicity.
Collapse
Affiliation(s)
- Weijue Nie
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Hong Zhu
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xin Sun
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jie Zhou
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Heng Xu
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Zhichao Yu
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Minghao Lu
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Baoping Jiang
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Lingling Zhou
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xueping Zhou
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|
27
|
Yahya MA, Alshammari GM, Osman MA, Al-Harbi LN, Yagoub AEA, AlSedairy SA. Liquorice root extract and isoliquiritigenin attenuate high-fat diet-induced hepatic steatosis and damage in rats by regulating AMPK. Arch Physiol Biochem 2024; 130:385-400. [PMID: 36121371 DOI: 10.1080/13813455.2022.2102654] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 07/05/2022] [Accepted: 07/08/2022] [Indexed: 11/11/2022]
Abstract
Objective: This study compared the ability of Liquorice roots aqueous extract (LRE) and its ingredient, isoliquiritigenin (ISL), in alleviating high-fat diet (HFD)-induced hepatic steatosis and examined if this effect involves activation of AMPK.Materials and methods: Control or HFD-fed rats were treated with the vehicle, LRE (200 mg/kg), or ISL (30 mg/kg) for 8 weeks orally.Results: ISL and LRE reduced HFD-induced hyperglycaemia, improved liver structure, lowered serum and hepatic lipids, and attenuated hepatic oxidative stress and inflammation. In the control and HFD-fed rats, ISL and LRE significantly stimulated the muscular and hepatic mRNA and protein levels of AMPK, improved oral glucose tolerance, reduced hepatic mRNA levels of SREBP1/2, and upregulated hepatic levels of PPARα and Bcl2. These effects were comparable for ISL and LRE and were prevented by co-administration of compound C, an AMPK inhibitor.Discussion and conclusion: ISL and LRE provide an effective theory to alleviate hepatic steatosis through activating AMPK.
Collapse
Affiliation(s)
- Mohammed Abdo Yahya
- Department of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Ghedeir M Alshammari
- Department of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Magdi A Osman
- Department of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Laila Naif Al-Harbi
- Department of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Abu ElGasim A Yagoub
- Department of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Sahar Abdulaziz AlSedairy
- Department of Food Science and Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
28
|
Tamimi A, Javid M, Sedighi-Pirsaraei N, Mirdamadi A. Exosome prospects in the diagnosis and treatment of non-alcoholic fatty liver disease. Front Med (Lausanne) 2024; 11:1420281. [PMID: 39144666 PMCID: PMC11322140 DOI: 10.3389/fmed.2024.1420281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 07/16/2024] [Indexed: 08/16/2024] Open
Abstract
The growing prevalence of NAFLD and its global health burden have provoked considerable research on possible diagnostic and therapeutic options for NAFLD. Although various pathophysiological mechanisms and genetic factors have been identified to be associated with NAFLD, its treatment remains challenging. In recent years, exosomes have attracted widespread attention for their role in metabolic dysfunctions and their efficacy as pathological biomarkers. Exosomes have also shown tremendous potential in treating a variety of disorders. With increasing evidence supporting the significant role of exosomes in NAFLD pathogenesis, their theragnostic potential has become a point of interest in NAFLD. Expectedly, exosome-based treatment strategies have shown promise in the prevention and amelioration of NAFLD in preclinical studies. However, there are still serious challenges in preparing, standardizing, and applying exosome-based therapies as a routine clinical option that should be overcome. Due to the great potential of this novel theragnostic agent in NAFLD, further investigations on their safety, clinical efficacy, and application standardization are highly recommended.
Collapse
|
29
|
Wang Q, Zhu K, Zhang A. SIRT1-mediated tunnelling nanotubes may be a potential intervention target for arsenic-induced hepatocyte senescence and liver damage. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 947:174502. [PMID: 38971248 DOI: 10.1016/j.scitotenv.2024.174502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/03/2024] [Accepted: 07/02/2024] [Indexed: 07/08/2024]
Abstract
Arsenic, a widespread environmental poison, can cause significant liver damage upon exposure. Mitochondria are the most sensitive organelles to external factors. Dysfunctional mitochondria play a crucial role in cellular senescence and liver damage. Tunnelling nanotubes (TNTs), membrane structures formed between cells, with fibrous actin (F-actin) serving as the scaffold, facilitate mitochondrial transfer between cells. Notably, TNTs mediate the delivery of healthy mitochondria to damaged cells, thereby mitigating cellular damage. Although limited studies have suggested that F-actin may be modulated by the longevity gene SIRT1, the association between arsenic-induced liver damage and this mechanism remains unexplored. The findings of the current study indicate that arsenic suppresses SIRT1 and F-actin in the rat liver and MIHA cells, impeding the formation of TNTs and mitochondrial transfer between MIHA cells, thereby playing a pivotal role in mitochondrial dysfunction, cellular senescence and liver damage induced by arsenic. Notably, increasing SIRT1 levels effectively mitigated liver mitochondrial dysfunction and cellular senescence triggered by arsenic, highlighting SIRT1's crucial regulatory function. This research provides novel insights into the mechanisms underlying arsenic-induced liver damage, paving the way for the development of targeted preventive and therapeutic drugs to address arsenic-induced liver damage.
Collapse
Affiliation(s)
- Qi Wang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Department of Toxicology, School of Public Health, Guizhou Medical University, Guiyang 561113, Guizhou, People's Republic of China; Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases, Co-Constructed by the Province and Ministry, Guizhou Medical University, Guiyang 561113, Guizhou, People's Republic of China
| | - Kai Zhu
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Department of Toxicology, School of Public Health, Guizhou Medical University, Guiyang 561113, Guizhou, People's Republic of China
| | - Aihua Zhang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Department of Toxicology, School of Public Health, Guizhou Medical University, Guiyang 561113, Guizhou, People's Republic of China; Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases, Co-Constructed by the Province and Ministry, Guizhou Medical University, Guiyang 561113, Guizhou, People's Republic of China.
| |
Collapse
|
30
|
Zhang S, Tuo P, Ji Y, Huang Z, Xiong Z, Li H, Ruan C. Identification of 1-Methylnicotinamide as a specific biomarker for the progression of cirrhosis to hepatocellular carcinoma. J Cancer Res Clin Oncol 2024; 150:310. [PMID: 38890166 PMCID: PMC11189347 DOI: 10.1007/s00432-024-05848-6] [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: 04/15/2024] [Accepted: 06/12/2024] [Indexed: 06/20/2024]
Abstract
PURPOSE Hepatocellular carcinoma (HCC) is a prevalent malignant tumor, often arising from hepatitis induced by the hepatitis B virus (HBV) in China. However, effective biomarkers for early diagnosis are lacking, leading to a 5-year overall survival rate of less than 20% among patients with advanced HCC. This study aims to identify serum biomarkers for early HCC diagnosis to enhance patient survival rates. METHODS We established an independent cohort comprising 27 healthy individuals, 13 patients with HBV-induced cirrhosis, 13 patients with hepatitis B-type HCC, and 8 patients who progressed from cirrhosis to hepatocellular carcinoma during follow-up. Serum metabolic abnormalities during the progression from cirrhosis to HCC were studied using untargeted metabolomics. Liquid chromatography-mass spectrometry-based metabolomics methods characterized the subjects' serum metabolic profiles. Partial least squares discriminant analysis (PLS-DA) was employed to elucidate metabolic profile changes during the progression from cirrhosis to HCC. Differentially expressed metabolites (DEMs) between cirrhosis and HCC groups were identified using the LIMMA package in the R language. Two machine learning algorithms, Least Absolute Shrinkage and Selection Operator (LASSO), and Random Forest Classifier (RF), were used to identify key metabolic biomarkers involved in the progression from cirrhosis to HCC. Key metabolic biomarkers were further validated using targeted metabolomics in a new independent validation cohort comprising 25 healthy individuals and 25 patients with early-stage hepatocellular carcinoma. RESULTS A total of 155 serum metabolites were identified, of which 21/54 metabolites exhibited significant changes in HCC patients compared with cirrhosis patients and healthy individuals, respectively. PLS-DA clustering results demonstrated a significant change trend in the serum metabolic profile of patients with HBV-induced cirrhosis during the progression to HCC. Utilizing LASSO regression and RF algorithms, we confirmed 10 key metabolic biomarkers. Notably, 1-Methylnicotinamide (1-MNAM) exhibited a persistent and significant decrease in healthy individuals, cirrhosis, and HCC patients. Moreover, 1-MNAM levels in developing patients were significantly higher during the cirrhosis stage than in the HCC stage. Targeted metabolomic validation in an external cohort further confirmed the good diagnostic performance of 1-MNAM in early HCC detection. CONCLUSION Our findings imply that 1-MNAM may be a specific biomarker for the progression of cirrhosis to HCC.
Collapse
Affiliation(s)
- Sijia Zhang
- Centre for Medical Research, Ningbo No. 2 Hospital, Ningbo, 315010, China
| | - Ping Tuo
- Centre for Medical Research, Ningbo No. 2 Hospital, Ningbo, 315010, China
| | - Yuanye Ji
- Centre for Medical Research, Ningbo No. 2 Hospital, Ningbo, 315010, China
| | - Zuoan Huang
- Centre for Medical Research, Ningbo No. 2 Hospital, Ningbo, 315010, China
| | - Zi Xiong
- Centre for Medical Research, Ningbo No. 2 Hospital, Ningbo, 315010, China
| | - Hongshan Li
- Liver Disease Department of Integrative Medicine, Ningbo No. 2 Hospital, Ningbo, 315010, China.
| | - Chunyan Ruan
- Centre for Medical Research, Ningbo No. 2 Hospital, Ningbo, 315010, China.
| |
Collapse
|
31
|
Zhuge H, Pan Y, Lai S, Chang K, Ding Q, Cao W, Song Q, Li S, Dou X, Ding B. Penthorum chinense Pursh extract ameliorates alcohol-related fatty liver disease in mice via the SIRT1/AMPK signaling axis. Heliyon 2024; 10:e31195. [PMID: 38832279 PMCID: PMC11145240 DOI: 10.1016/j.heliyon.2024.e31195] [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: 12/08/2023] [Revised: 05/10/2024] [Accepted: 05/13/2024] [Indexed: 06/05/2024] Open
Abstract
Penthorum chinense Pursh (P. chinense), a functional food, has been applied to protect the liver against alcohol-related fatty liver disease (ALD) for a long history in China. This study was designed to evaluate the ameliorative activity of the polyphenolic fraction in P. chinense (PGF) depending on the relief of ALD. The ALD mouse model was established by exposing the mice to a Lieber-DeCarli alcohol liquid diet. We found that PGF administration significantly ameliorated alcohol-induced liver injury, steatosis, oxidative stress, and inflammation in mice. Furthermore, alcohol-increased levels of the critical hepatic lipid synthesis proteins sterol regulatory element binding transcription factor (SREBP-1) and diacylglycerol o-acyltransferase 2 (DGAT2) were attenuated by PGF. Similarly, PGF inhibited the expression of the lipid transport protein very low-density lipoprotein receptor (VLDLR). Interestingly, PGF restored alcohol-inhibited expression of carnitine palmitoyltransferase 1 (CPT1) and peroxisome proliferator-activated receptor alpha (PPARα), essential fatty acid β-oxidation proteins. Mechanistic studies revealed that PGF protects against alcohol-induced hepatocyte injury and lipid deposition via the SIRT1/AMPK signaling pathway. In sum, this research clearly demonstrated the protective effects of PGF against ALD, which was mediated by activating SIRT1/AMPK pathways in hepatocytes. We provide a new theoretical basis for using P. chinense as a functional food in ALD.
Collapse
Affiliation(s)
- Hui Zhuge
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Molecular Medicine Institute, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yan Pan
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Molecular Medicine Institute, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Shanglei Lai
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Molecular Medicine Institute, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Kaixin Chang
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Molecular Medicine Institute, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Qinchao Ding
- College of Public Health, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- College of Animal Science, Zhejiang University, Hangzhou, 310058, China
- Molecular Medicine Institute, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Wenjing Cao
- College of Public Health, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Qing Song
- College of Public Health, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Songtao Li
- College of Public Health, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Xiaobing Dou
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Molecular Medicine Institute, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Bin Ding
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Molecular Medicine Institute, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| |
Collapse
|
32
|
Ding Z, Zhang J, Choudhury M. A High-Fat and High-Fructose Diet Exacerbates Liver Dysfunction by Regulating Sirtuins in a Murine Model. Life (Basel) 2024; 14:729. [PMID: 38929712 PMCID: PMC11205069 DOI: 10.3390/life14060729] [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: 05/10/2024] [Revised: 05/31/2024] [Accepted: 06/01/2024] [Indexed: 06/28/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is rapidly emerging as the most prevalent chronic liver disease, closely linked to the escalating rates of diabesity. The Western diet's abundance of fat and fructose significantly contributes to MASLD, disrupting hepatic glucose metabolism. We previously demonstrated that a high-fat and high-fructose diet (HFHFD) led to increased body and liver weight compared to the low-fat diet (LFD) group, accompanied by glucose intolerance and liver abnormalities, indicating an intermediate state between fatty liver and liver fibrosis in the HFHFD group. Sirtuins are crucial epigenetic regulators associated with energy homeostasis and play a pivotal role in these hepatic dysregulations. Our investigation revealed that HFHFD significantly decreased Sirt1 and Sirt7 gene and protein expression levels, while other sirtuins remained unchanged. Additionally, glucose 6-phosphatase (G6Pase) gene expression was reduced in the HFHFD group, suggesting a potential pathway contributing to fibrosis progression. Chromatin immunoprecipitation analysis demonstrated a significant increase in histone H3 lysine 18 acetylation within the G6Pase promoter in HFHFD livers, potentially inhibiting G6Pase transcription. In summary, HFHFD may inhibit liver gluconeogenesis, potentially promoting liver fibrosis by regulating Sirt7 expression. This study offers an epigenetic perspective on the detrimental impact of fructose on MASLD progression.
Collapse
Affiliation(s)
| | | | - Mahua Choudhury
- Department of Pharmaceutical Sciences, Irma Lerma Rangel School of Pharmacy, Texas A&M University, College Station, TX 77843-1114, USA
| |
Collapse
|
33
|
BinMowyna MN, AlFaris NA, Al-Sanea EA, AlTamimi JZ, Aldayel TS. Resveratrol attenuates against high-fat-diet-promoted non-alcoholic fatty liver disease in rats mainly by targeting the miR-34a/SIRT1 axis. Arch Physiol Biochem 2024; 130:300-315. [PMID: 35254877 DOI: 10.1080/13813455.2022.2046106] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 02/18/2022] [Indexed: 11/09/2022]
Abstract
This study evaluated if miR-34a/SIRT1 signalling mediates the anti-hepatosteatotic effect of resveratrol (RSV) in high-fat-diet (HFD)-fed rats. Rats were divided into seven groups (n = 6/each) as control, control + miR-34a agomir negative control, HFD, HFD + miR-34a, HFD + RSV, HFD + RSV + Ex-527 (a SIRT1 inhibitor), and HFD + RSV + miR-34a agomir. After 8 weeks, RSV suppressed dyslipidemia, lowered fasting glucose and insulin levels, improved insulin sensitivity, and prevented hepatic lipid accumulation. These effects were associated with hepatic downregulation of SREBP1 and SREBP2, upregulation of PPARα, and acetylation of Nrf2 (activation) and NF-κβ p65 (inhibition). Also, RSV reduced the transcription of miR-34a and increased the nuclear localisation of SIRT1 in the livers, muscles, and adipose tissues of HFD-fed rats. All these effects were prevented by EX-527 and miR-34a agmir. In conclusion, RSV prevents HFD-induced insulin resistance and hepatic steatosis by suppressing miR-34a-induced activation of SIRT1.
Collapse
Affiliation(s)
- Mona N BinMowyna
- College of Applied Medical Sciences, Shaqra University, Shaqra, Saudi Arabia
| | - Nora A AlFaris
- Department of Physical Sport Science, College of Education, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Ekram A Al-Sanea
- Department of Biology, College of Sciences, Ibb University, Ibb, Yemen
| | - Jozaa Z AlTamimi
- Department of Physical Sport Science, College of Education, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Tahany S Aldayel
- Department of Physical Sport Science, College of Education, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| |
Collapse
|
34
|
Kung ML, Cheng SM, Wang YH, Cheng KP, Li YL, Hsiao YT, Tan BCM, Chen YW. Deficiency of ADAR2 ameliorates metabolic-associated fatty liver disease via AMPK signaling pathways in obese mice. Commun Biol 2024; 7:594. [PMID: 38760406 PMCID: PMC11101631 DOI: 10.1038/s42003-024-06215-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Accepted: 04/18/2024] [Indexed: 05/19/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a chronic disease caused by hepatic steatosis. Adenosine deaminases acting on RNA (ADARs) catalyze adenosine to inosine RNA editing. However, the functional role of ADAR2 in NAFLD is unclear. ADAR2+/+/GluR-BR/R mice (wild type, WT) and ADAR2-/-/GluR-BR/R mice (ADAR2 KO) mice are fed with standard chow or high-fat diet (HFD) for 12 weeks. ADAR2 KO mice exhibit protection against HFD-induced glucose intolerance, insulin resistance, and dyslipidemia. Moreover, ADAR2 KO mice display reduced liver lipid droplets in concert with decreased hepatic TG content, improved hepatic insulin signaling, better pyruvate tolerance, and increased glycogen synthesis. Mechanistically, ADAR2 KO effectively mitigates excessive lipid production via AMPK/Sirt1 pathway. ADAR2 KO inhibits hepatic gluconeogenesis via the AMPK/CREB pathway and promotes glycogen synthesis by activating the AMPK/GSK3β pathway. These results provide evidence that ADAR2 KO protects against NAFLD progression through the activation of AMPK signaling pathways.
Collapse
Affiliation(s)
- Mei-Lang Kung
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Siao Muk Cheng
- National Institute of Cancer Research, National Health Research Institutes (NHRI), Tainan, Taiwan
| | - Yun-Han Wang
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Kai-Pi Cheng
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Lin Li
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Tsen Hsiao
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Bertrand Chin-Ming Tan
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Neurosurgery, Linkou Medical Center, Chang Gung Memorial Hospital, Linkou, Taiwan
- Research Center for Emerging Viral Infections, Chang Gung University, Taoyuan, Taiwan
| | - Yun-Wen Chen
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
35
|
Méndez-Mancilla A, Turiján-Espinoza E, Vega-Cárdenas M, Hernández-Hernández GE, Uresti-Rivera EE, Vargas-Morales JM, Portales-Pérez DP. miR-21, miR-221, miR-29 and miR-34 are distinguishable molecular features of a metabolically unhealthy phenotype in young adults. PLoS One 2024; 19:e0300420. [PMID: 38662716 PMCID: PMC11045123 DOI: 10.1371/journal.pone.0300420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 02/26/2024] [Indexed: 04/28/2024] Open
Abstract
Discrepancies between the measurement of body mass index (BMI) and metabolic health status have been described for the onset of metabolic diseases. Studying novel biomarkers, some of which are associated with metabolic syndrome, can help us to understand the differences between metabolic health (MetH) and BMI. A group of 1469 young adults with pre-specified anthropometric and blood biochemical parameters were selected. Of these, 80 subjects were included in the downstream analysis that considered their BMI and MetH parameters for selection as follows: norm weight metabolically healthy (MHNW) or metabolically unhealthy (MUNW); overweight/obese metabolically healthy (MHOW) or metabolically unhealthy (MUOW). Our results showed for the first time the differences when the MetH status and the BMI are considered as global MetH statures. First, all the evaluated miRNAs presented a higher expression in the metabolically unhealthy group than the metabolically healthy group. The higher levels of leptin, IL-1b, IL-8, IL-17A, miR-221, miR-21, and miR-29 are directly associated with metabolic unhealthy and OW/OB phenotypes (MUOW group). In contrast, high levels of miR34 were detected only in the MUNW group. We found differences in the SIRT1-PGC1α pathway with increased levels of SIRT1+ cells and diminished mRNA levels of PGCa in the metabolically unhealthy compared to metabolically healthy subjects. Our results demonstrate that even when metabolic diseases are not apparent in young adult populations, MetH and BMI have a distinguishable phenotype print that signals the potential to develop major metabolic diseases.
Collapse
Affiliation(s)
- Alejandro Méndez-Mancilla
- Laboratory of Immunology and Cellular and Molecular Biology, Faculty of Chemical Sciences, Autonomous University of San Luis Potosí, San Luis Potosí, San Luis Potosi, Mexico
- Translational and Molecular Medicine Department, Research Center for Health Sciences and Biomedicine (CICSaB), Autonomous University of San Luis Potosí, San Luis Potosí, San Luis Potosi, Mexico
| | - Eneida Turiján-Espinoza
- Laboratory of Immunology and Cellular and Molecular Biology, Faculty of Chemical Sciences, Autonomous University of San Luis Potosí, San Luis Potosí, San Luis Potosi, Mexico
- Translational and Molecular Medicine Department, Research Center for Health Sciences and Biomedicine (CICSaB), Autonomous University of San Luis Potosí, San Luis Potosí, San Luis Potosi, Mexico
| | - Mariela Vega-Cárdenas
- Translational and Molecular Medicine Department, Research Center for Health Sciences and Biomedicine (CICSaB), Autonomous University of San Luis Potosí, San Luis Potosí, San Luis Potosi, Mexico
| | - Gloria Estela Hernández-Hernández
- Laboratory of Immunology and Cellular and Molecular Biology, Faculty of Chemical Sciences, Autonomous University of San Luis Potosí, San Luis Potosí, San Luis Potosi, Mexico
| | - Edith Elena Uresti-Rivera
- Laboratory of Immunology and Cellular and Molecular Biology, Faculty of Chemical Sciences, Autonomous University of San Luis Potosí, San Luis Potosí, San Luis Potosi, Mexico
- Translational and Molecular Medicine Department, Research Center for Health Sciences and Biomedicine (CICSaB), Autonomous University of San Luis Potosí, San Luis Potosí, San Luis Potosi, Mexico
| | - Juan M. Vargas-Morales
- Laboratory of Immunology and Cellular and Molecular Biology, Faculty of Chemical Sciences, Autonomous University of San Luis Potosí, San Luis Potosí, San Luis Potosi, Mexico
- Laboratory of Clinical Analysis, Faculty of Chemical Sciences, Autonomous University of San Luis Potosí, San Luis Potosí, San Luis Potosi, Mexico
| | - Diana P. Portales-Pérez
- Laboratory of Immunology and Cellular and Molecular Biology, Faculty of Chemical Sciences, Autonomous University of San Luis Potosí, San Luis Potosí, San Luis Potosi, Mexico
- Translational and Molecular Medicine Department, Research Center for Health Sciences and Biomedicine (CICSaB), Autonomous University of San Luis Potosí, San Luis Potosí, San Luis Potosi, Mexico
| |
Collapse
|
36
|
Ko HJ, Park SY, Sim DY, Kim SH, Hur S, Lee JH, Kim Y. Apoptotic Effect of Isoimpertorin via Inhibition of c-Myc and SIRT1 Signaling Axis. Int J Mol Sci 2024; 25:4248. [PMID: 38673833 PMCID: PMC11050721 DOI: 10.3390/ijms25084248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/05/2024] [Accepted: 04/06/2024] [Indexed: 04/28/2024] Open
Abstract
Though Isoimperatorin from Angelicae dahuricae is known to have antiviral, antidiabetic, anti-inflammatory and antitumor effects, its underlying antitumor mechanism remains elusive so far. Hence, the apoptotic mechanism of Isoimperatorin was explored in hepatocellular carcinomas (HCCs). In this study, Isoimperatorin inhibited the viability of Huh7 and Hep3B HCCs and increased the subG1 apoptotic portion and also abrogated the expression of pro-poly-ADP ribose polymerase (pro-PARP) and pro-caspase 3 in Huh7 and Hep3B cells. Also, Isoimperatorin abrogated the expression of cyclin D1, cyclin E1, CDK2, CDK4, CDK6 and increased p21 as G1 phase arrest-related proteins in Huh7 and Hep3B cells. Interestingly, Isoimperatorin reduced the expression and binding of c-Myc and Sirtuin 1 (SIRT1) by Immunoprecipitation (IP), with a binding score of 0.884 in Huh7 cells. Furthermore, Isoimperatorin suppressed the overexpression of c-Myc by the proteasome inhibitor MG132 and also disturbed cycloheximide-treated c-Myc stability in Huh7 cells. Overall, these findings support the novel evidence that the pivotal role of c-Myc and SIRT1 is critically involved in Isoimperatorin-induced apoptosis in HCCs as potent molecular targets in liver cancer therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Youngchul Kim
- Department of Clinical Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (H.-J.K.); (S.-Y.P.); (D.Y.S.); (S.-H.K.); (S.H.); (J.-H.L.)
| |
Collapse
|
37
|
Lu H. Inflammatory liver diseases and susceptibility to sepsis. Clin Sci (Lond) 2024; 138:435-487. [PMID: 38571396 DOI: 10.1042/cs20230522] [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: 09/03/2023] [Revised: 01/09/2024] [Accepted: 03/12/2024] [Indexed: 04/05/2024]
Abstract
Patients with inflammatory liver diseases, particularly alcohol-associated liver disease and metabolic dysfunction-associated fatty liver disease (MAFLD), have higher incidence of infections and mortality rate due to sepsis. The current focus in the development of drugs for MAFLD is the resolution of non-alcoholic steatohepatitis and prevention of progression to cirrhosis. In patients with cirrhosis or alcoholic hepatitis, sepsis is a major cause of death. As the metabolic center and a key immune tissue, liver is the guardian, modifier, and target of sepsis. Septic patients with liver dysfunction have the highest mortality rate compared with other organ dysfunctions. In addition to maintaining metabolic homeostasis, the liver produces and secretes hepatokines and acute phase proteins (APPs) essential in tissue protection, immunomodulation, and coagulation. Inflammatory liver diseases cause profound metabolic disorder and impairment of energy metabolism, liver regeneration, and production/secretion of APPs and hepatokines. Herein, the author reviews the roles of (1) disorders in the metabolism of glucose, fatty acids, ketone bodies, and amino acids as well as the clearance of ammonia and lactate in the pathogenesis of inflammatory liver diseases and sepsis; (2) cytokines/chemokines in inflammatory liver diseases and sepsis; (3) APPs and hepatokines in the protection against tissue injury and infections; and (4) major nuclear receptors/signaling pathways underlying the metabolic disorders and tissue injuries as well as the major drug targets for inflammatory liver diseases and sepsis. Approaches that focus on the liver dysfunction and regeneration will not only treat inflammatory liver diseases but also prevent the development of severe infections and sepsis.
Collapse
Affiliation(s)
- Hong Lu
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY 13210, U.S.A
| |
Collapse
|
38
|
Chiang CH, Zhang TR, Hsu PS, Lin SP, Chen CY. Weight regain, but not weight loss exacerbates hepatic fibrosis during multiple weight cycling events in male mice. Eur J Nutr 2024; 63:965-976. [PMID: 38265751 DOI: 10.1007/s00394-024-03326-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 01/12/2024] [Indexed: 01/25/2024]
Abstract
PURPOSE Weight cycling is a phenomenon characterized by fluctuating body weight that is commonly observed in individuals employing intentional weight loss methods. Despite its prevalence, the impact of weight cycling on health remains equivocal. The current investigation aimed to examine the effects of weight cycling on liver health. METHODS The weight cycling model was established by switching the feeding method of mice between ad libitum (AL) and restricted intake (DR or 60% of AL) of the breeding diet to cause weight gain and weight loss, respectively. The weight cycling model comprised two and a half cycles, with one group terminating the experience during the weight-gain period (S-AL) and the other during the weight-loss period (S-DR). Liver tissue was collected to investigate morphology alterations, apoptosis, lipid metabolism, and mitochondrial homeostasis. RESULTS The results demonstrated that the termination point of weight cycling affected body weight and hepatic steatosis. All parameters examined in the S-DR mice exhibited a comparable trend to those observed in the DR mice. Notably, S-AL mice showed a significant increase in lipid metabolism-related proteins in the liver compared to AL-fed mice, along with reduced lipid droplets. Moreover, hepatic apoptosis and fibrosis were exacerbated in the S-AL mice compared to AL mice, whereas mitochondrial fusion, biogenesis, and mitophagy were decreased in the S-AL mice. CONCLUSION Weight cycling ending in weight gain exacerbated hepatic fibrosis, potentially by inducing apoptosis or disrupting mitochondrial homeostasis. Conversely, weight cycling ending in weight loss demonstrated beneficial effects on hepatic health.
Collapse
Affiliation(s)
- Chun-Hsien Chiang
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Ting-Rui Zhang
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Pu-Sheng Hsu
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Shau-Ping Lin
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Ching-Yi Chen
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
39
|
Chen C, Hu S, Hu HJ, Liu ZX, Wu XT, Zou T, Su H. Dronedarone Attenuates Ang II-Induced Myocardial Hypertrophy Through Regulating SIRT1/FOXO3/PKIA Axis. Korean Circ J 2024; 54:172-186. [PMID: 38654563 PMCID: PMC11040268 DOI: 10.4070/kcj.2023.0197] [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: 07/11/2023] [Revised: 12/21/2023] [Accepted: 01/23/2024] [Indexed: 04/26/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Long-term pathological myocardial hypertrophy (MH) seriously affects the normal function of the heart. Dronedarone was reported to attenuate left ventricular hypertrophy of mice. However, the molecular regulatory mechanism of dronedarone in MH is unclear. METHODS Angiotensin II (Ang II) was used to induce cell hypertrophy of H9C2 cells. Transverse aortic constriction (TAC) surgery was performed to establish a rat model of MH. Cell size was evaluated using crystal violet staining and rhodamine phalloidin staining. Reverse transcription quantitative polymerase chain reaction and western blot were performed to detect the mRNA and protein expressions of genes. JASPAR and luciferase activity were conducted to predict and validate interaction between forkhead box O3 (FOXO3) and protein kinase inhibitor alpha (PKIA) promoter. RESULTS Ang II treatment induced cell hypertrophy and inhibited sirtuin 1 (SIRT1) expression, which were reversed by dronedarone. SIRT1 overexpression or PKIA overexpression enhanced dronedarone-mediated suppression of cell hypertrophy in Ang II-induced H9C2 cells. Mechanistically, SIRT1 elevated FOXO3 expression through SIRT1-mediated deacetylation of FOXO3 and FOXO3 upregulated PKIA expression through interacting with PKIA promoter. Moreover, SIRT1 silencing compromised dronedarone-mediated suppression of cell hypertrophy, while PKIA upregulation abolished the influences of SIRT1 silencing. More importantly, dronedarone improved TAC surgery-induced MH and impairment of cardiac function of rats via affecting SIRT1/FOXO3/PKIA axis. CONCLUSIONS Dronedarone alleviated MH through mediating SIRT1/FOXO3/PKIA axis, which provide more evidences for dronedarone against MH.
Collapse
Affiliation(s)
- Cheng Chen
- Department of Cardiovascular Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Song Hu
- Department of Cardiovascular Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Heng-Jing Hu
- Department of Cardiovascular Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Zhi-Xuan Liu
- Department of Cardiovascular Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Xin-Teng Wu
- Department of Cardiovascular Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Tao Zou
- Department of Cardiovascular Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Hua Su
- Department of Cardiovascular Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China.
| |
Collapse
|
40
|
Sangpairoj K, Pranweerapaiboon K, Saengkhae C, Meemon K, Niamnont N, Tamtin M, Sobhon P, Yisarakun W, Siangcham T. Extracts of tropical green seaweed Caulerpa lentillifera reduce hepatic lipid accumulation by modulating lipid metabolism molecules in HepG2 cells. Heliyon 2024; 10:e27635. [PMID: 38509999 PMCID: PMC10950575 DOI: 10.1016/j.heliyon.2024.e27635] [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: 09/07/2023] [Revised: 03/04/2024] [Accepted: 03/04/2024] [Indexed: 03/22/2024] Open
Abstract
Seaweed has attracted attention as a bioactive source for preventing different chronic diseases, including liver injury and non-alcoholic fatty liver disease, the leading cause of liver-related mortality. Caulerpa lentillifera is characterized as tropical edible seaweed, currently being investigated for health benefits of its extracts and bioactive substances. This study examined the effects of C. lentillifera extract in ethyl acetate fraction (CLEA) on controlling lipid accumulation and lipid metabolism in HepG2 cells induced with oleic acid through the in vitro hepatic steatosis model. Gas chromatography-mass spectrometry (GC-MS) analysis indicated that CLEA contained diverse organic compounds, including hydrocarbons, amino acids, and carboxylic acids. Docked conformation of dl-2-phenyltryptophane and benzoic acid, two major bioactive CLEA components, showed high affinity binding to SIRT1 and AMPK as target molecules of lipid metabolism. CLEA reduced lipid accumulation and intracellular triglyceride levels in HepG2 cells stimulated with oleic acid. The effect of CLEA on regulating expression of lipid metabolism-related molecules was investigated by qPCR and immunoblotting. CLEA promoted expression of the SIRT1 gene in oleic acid-treated HepG2 cells. CLEA also reduced expression levels of SREBF1, FAS, and ACC genes, which might be related to activation of AMPK signaling in lipid-accumulated HepG2 cells. These findings suggest that CLEA contains bioactive compounds potentially reducing triglyceride accumulation in lipid-accumulated HepG2 hepatocytes by controlling lipid metabolism molecules.
Collapse
Affiliation(s)
- Kant Sangpairoj
- Division of Anatomy, Department of Preclinical Science, Faculty of Medicine, Thammasat University, Pathum Thani, Thailand
- Thammasat University Research Unit in Nutraceuticals and Food Safety, Pathum Thani, Thailand
| | - Kanta Pranweerapaiboon
- Chulabhorn International College of Medicine, Thammasat University, Pathum Thani, Thailand
| | | | - Krai Meemon
- Department of Anatomy, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Nakorn Niamnont
- Department of Chemistry, Faculty of Science, King Mongkut's University of Technology Thonburi, Bang Mod, Bangkok, Thailand
| | - Montakan Tamtin
- Department of Fisheries, Kung Krabaen Bay Royal Development Study Centre, Chanthaburi, Thailand
| | - Prasert Sobhon
- Department of Anatomy, Faculty of Science, Mahidol University, Bangkok, Thailand
| | | | - Tanapan Siangcham
- Faculty of Allied Health Sciences, Burapha University, Chonburi, Thailand
| |
Collapse
|
41
|
Zhang R, Zhang Q, Cui Z, Huang B, Ma H. Dimethyl fumarate restores Ca 2+ dyshomeostasis through activation of the SIRT1 signal to treat nonalcoholic fatty liver disease. Life Sci 2024; 341:122505. [PMID: 38364937 DOI: 10.1016/j.lfs.2024.122505] [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: 11/20/2023] [Revised: 02/09/2024] [Accepted: 02/12/2024] [Indexed: 02/18/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is characterized by an excessive lipid accumulation in the liver, with a global prevalence of approximately 25 %. While early-stage steatosis is reversible and can be intervened upon, it has the potential to progress to some serious complications, including cirrhosis and even liver cancer. Dimethyl fumarate (DMF), a derivative of fumaric acid shows promise in intervening in certain diseases. However, the precise effect and underlying mechanism of DMF on hepatic steatosis remain unclear. In this study, we demonstrated that DMF mitigates hepatic steatosis in mice subjected to high-fat/high-cholesterol (HFHC) diets. Meanwhile, our in vivo and in vitro results showed that DMF relieves lipid accumulation, oxidative stress, and endoplasmic reticulum (ER) stress. Mechanically, our findings revealed that the effect of DMF on reducing lipid accumulation is linked to the restoration of Ca2+ homeostasis. Furthermore, we found that activation of the SIRT1 signal by DMF plays an important role in correcting the mishandling of the Ca2+ signal, and knockdown of SIRT1 expression reverses the beneficial role of DMF PA-incubated AML12 cells. In conclusion, our results suggested DMF's amelioration of hepatic steatosis is related to the activation of SIRT1-mediated Ca2+ signaling.
Collapse
Affiliation(s)
- Rui Zhang
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Quanwei Zhang
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - ZiYi Cui
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - BenZeng Huang
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Haitian Ma
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
42
|
Sharma C, Hamza A, Boyle E, Donu D, Cen Y. Post-Translational Modifications and Diabetes. Biomolecules 2024; 14:310. [PMID: 38540730 PMCID: PMC10968569 DOI: 10.3390/biom14030310] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/01/2024] [Accepted: 03/04/2024] [Indexed: 09/22/2024] Open
Abstract
Diabetes and its associated complications have increasingly become major challenges for global healthcare. The current therapeutic strategies involve insulin replacement therapy for type 1 diabetes (T1D) and small-molecule drugs for type 2 diabetes (T2D). Despite these advances, the complex nature of diabetes necessitates innovative clinical interventions for effective treatment and complication prevention. Accumulative evidence suggests that protein post-translational modifications (PTMs), including glycosylation, phosphorylation, acetylation, and SUMOylation, play important roles in diabetes and its pathological consequences. Therefore, the investigation of these PTMs not only sheds important light on the mechanistic regulation of diabetes but also opens new avenues for targeted therapies. Here, we offer a comprehensive overview of the role of several PTMs in diabetes, focusing on the most recent advances in understanding their functions and regulatory mechanisms. Additionally, we summarize the pharmacological interventions targeting PTMs that have advanced into clinical trials for the treatment of diabetes. Current challenges and future perspectives are also provided.
Collapse
Affiliation(s)
- Chiranjeev Sharma
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23219, USA; (C.S.); (A.H.); (E.B.); (D.D.)
| | - Abu Hamza
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23219, USA; (C.S.); (A.H.); (E.B.); (D.D.)
| | - Emily Boyle
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23219, USA; (C.S.); (A.H.); (E.B.); (D.D.)
| | - Dickson Donu
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23219, USA; (C.S.); (A.H.); (E.B.); (D.D.)
| | - Yana Cen
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23219, USA; (C.S.); (A.H.); (E.B.); (D.D.)
- Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, VA 23219, USA
| |
Collapse
|
43
|
Dong H, Guo W, Yue R, Sun X, Zhou Z. Nuclear Nicotinamide Adenine Dinucleotide Deficiency by Nmnat1 Deletion Impaired Hepatic Insulin Signaling, Mitochondrial Function, and Hepatokine Expression in Mice Fed a High-Fat Diet. J Transl Med 2024; 104:100329. [PMID: 38237740 PMCID: PMC10957298 DOI: 10.1016/j.labinv.2024.100329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 12/20/2023] [Accepted: 01/09/2024] [Indexed: 02/12/2024] Open
Abstract
Metabolic syndrome (MetS) is a worldwide challenge that is closely associated with obesity, nonalcoholic liver disease, insulin resistance, and type 2 diabetes. Boosting nicotinamide adenine dinucleotide (NAD+) presents great potential in preventing MetS. However, the function of nuclear NAD+ in the development of MetS remains poorly understood. In this study, hepatocyte-specific Nmnat1 knockout mice were used to determine a possible link between nuclear NAD+ and high-fat diet (HFD)-induced MetS. We found that Nmnat1 knockout significantly reduced hepatic nuclear NAD+ levels but did not exacerbate HFD-induced obesity and hepatic triglycerides accumulation. Interestingly, loss of Nmnat1 caused insulin resistance. Further analysis revealed that Nmnat1 deletion promoted gluconeogenesis but inhibited glycogen synthesis in the liver. Moreover, Nmnat1 deficiency induced mitochondrial dysfunction by decreasing mitochondrial DNA (mtDNA)-encoded complexes Ⅰ and Ⅳ, suppressing mtDNA replication and mtRNA transcription and reducing mtDNA copy number. In addition, Nmnat1 depletion affected the expression of hepatokines in the liver, particularly downregulating the expression of follistatin. These findings highlight the importance of nuclear NAD+ in maintaining insulin sensitivity and provide insights into the mechanisms underlying HFD-induced insulin resistance.
Collapse
Affiliation(s)
- Haibo Dong
- Center for Translational Biomedical Research, University of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, North Carolina
| | - Wei Guo
- Center for Translational Biomedical Research, University of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, North Carolina
| | - Ruichao Yue
- Center for Translational Biomedical Research, University of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, North Carolina
| | - Xinguo Sun
- Center for Translational Biomedical Research, University of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, North Carolina
| | - Zhanxiang Zhou
- Center for Translational Biomedical Research, University of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, North Carolina; Department of Nutrition, University of North Carolina at Greensboro, Greensboro, North Carolina.
| |
Collapse
|
44
|
Soto A, Spongberg C, Martinino A, Giovinazzo F. Exploring the Multifaceted Landscape of MASLD: A Comprehensive Synthesis of Recent Studies, from Pathophysiology to Organoids and Beyond. Biomedicines 2024; 12:397. [PMID: 38397999 PMCID: PMC10886580 DOI: 10.3390/biomedicines12020397] [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: 01/05/2024] [Revised: 02/04/2024] [Accepted: 02/05/2024] [Indexed: 02/25/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a widespread contributor to chronic liver disease globally. A recent consensus on renaming liver disease was established, and metabolic dysfunction-associated steatotic liver disease, MASLD, was chosen as the replacement for NAFLD. The disease's range extends from the less severe MASLD, previously known as non-alcoholic fatty liver (NAFL), to the more intense metabolic dysfunction-associated steatohepatitis (MASH), previously known as non-alcoholic steatohepatitis (NASH), characterized by inflammation and apoptosis. This research project endeavors to comprehensively synthesize the most recent studies on MASLD, encompassing a wide spectrum of topics such as pathophysiology, risk factors, dietary influences, lifestyle management, genetics, epigenetics, therapeutic approaches, and the prospective trajectory of MASLD, particularly exploring its connection with organoids.
Collapse
Affiliation(s)
- Allison Soto
- Department of Surgery, University of Illinois College of Medicine, Chicago, IL 60607, USA;
| | - Colby Spongberg
- Touro College of Osteopathic Medicine, Great Falls, MT 59405, USA
| | | | - Francesco Giovinazzo
- General Surgery and Liver Transplant Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| |
Collapse
|
45
|
Naiini MR, Shahouzehi B, Azizi S, Shafiei B, Nazari-Robati M. Trehalose-induced SIRT1/AMPK activation regulates SREBP-1c/PPAR-α to alleviate lipid accumulation in aged liver. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:1061-1070. [PMID: 37581638 DOI: 10.1007/s00210-023-02644-w] [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: 04/28/2023] [Accepted: 07/24/2023] [Indexed: 08/16/2023]
Abstract
Aging is associated with a disturbance in the regulation of the metabolic function of the liver, which increases the risk of liver and systemic diseases. Trehalose, a natural disaccharide, has been identified to reduce dyslipidemia, hepatic steatosis, and glucose intolerance. However, the roles of trehalose on lipid metabolism in aged liver are unclear which was investigated in this study. Thirty-two male Wistar rats were randomly allocated into four groups (n = 8). Two groups of aged (24 months) and young (4 months) rats were administered 2% trehalose solution orally for 30 days. Control groups of aged and young rats did not receive any treatment. At the end of the treatment period, blood samples and liver tissues were collected. Then the expression of SIRT1, AMPK, SREBP-1c, and PPAR-α and the level of AMPK phosphorylation (p-AMPK) were quantified by real-time polymerase chain reaction and western blotting. Moreover, biochemical parameters and the histopathology of livers were evaluated. Trehalose supplementation increased the level of SIRT1, p-AMPK, and PPAR-α, whereas the level of SREBP-1c was diminished in the liver of old animals. In addition, treatment with trehalose improved histopathological features of senescent livers. Taken together, our results show that old rats developed lipogenesis in the liver which was alleviated with trehalose. Therefore, trehalose may be an effective intervention to reduce the progression of aging-induced liver diseases.
Collapse
Affiliation(s)
- Mahdis Rahimi Naiini
- Department of Clinical Biochemistry, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Beydolah Shahouzehi
- Cardiovascular Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Shahrzad Azizi
- Department of Pathobiology, Faculty of Veterinary Medicine, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Bentolhoda Shafiei
- Department of Clinical Biochemistry, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mahdieh Nazari-Robati
- Department of Clinical Biochemistry, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
46
|
Bahramzadeh A, Samavarchi Tehrani S, Goodarzi G, Seyyedebrahimi S, Meshkani R. Combination therapy of metformin and morin attenuates insulin resistance, inflammation, and oxidative stress in skeletal muscle of high-fat diet-fed mice. Phytother Res 2024; 38:912-924. [PMID: 38091524 DOI: 10.1002/ptr.8086] [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: 06/21/2023] [Revised: 10/25/2023] [Accepted: 11/26/2023] [Indexed: 02/15/2024]
Abstract
Lipid accumulation, inflammation, and oxidative stress are the most important causes of muscle insulin resistance. The aim of this study was to investigate the single and combined treatment effects of metformin (MET) and morin (MOR) on lipid accumulation, inflammation, and oxidative stress in the skeletal muscle of mice fed a high-fat diet. The mice were supplemented with MET (230 mg/kg diet), MOR (100 mg/kg diet), and MET + MOR for 9 weeks. Our results revealed that single treatment with MET or MOR, and with a stronger effect of MET + MOR combined treatment, reduced body weight gain, improved glucose intolerance and enhanced Akt phosphorylation in the muscle tissue. In addition, plasma and muscle triglyceride levels were decreased after treatment with MET and MOR. The expression of genes involved in macrophage infiltration and polarization and pro-inflammatory cytokines showed that MET + MOR combined treatment, significantly reduced inflammation in the muscle. Furthermore, combined treatment of MET + MOR with greater efficacy than the single treatment improved several oxidative stress markers in the muscle. Importantly, combined treatment of MET and MOR could increase the expression of nuclear factor erythroid 2-related factor 2, the master regulator of the antioxidant response. These findings suggest that combination of MET with MOR might ameliorate insulin resistance, inflammation, and oxidative stress in the skeletal muscle of mice fed high-fat diet.
Collapse
Affiliation(s)
- Arash Bahramzadeh
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sadra Samavarchi Tehrani
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Science, Tehran, Iran
| | - Golnaz Goodarzi
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pathobiology and Laboratory Science, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - ShadiSadat Seyyedebrahimi
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Meshkani
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
47
|
Rad NK, Heydari Z, Tamimi AH, Zahmatkesh E, Shpichka A, Barekat M, Timashev P, Hossein-Khannazer N, Hassan M, Vosough M. Review on Kidney-Liver Crosstalk: Pathophysiology of Their Disorders. CELL JOURNAL 2024; 26:98-111. [PMID: 38459727 PMCID: PMC10924833 DOI: 10.22074/cellj.2023.2007757.1376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 12/25/2023] [Accepted: 12/30/2023] [Indexed: 03/10/2024]
Abstract
Kidney-liver crosstalk plays a crucial role in normal and certain pathological conditions. In pathologic states, both renal-induced liver damage and liver-induced kidney diseases may happen through these kidney-liver interactions. This bidirectional crosstalk takes place through the systemic conditions that mutually influence both the liver and kidneys. Ischemia and reperfusion, cytokine release and pro-inflammatory signaling pathways, metabolic acidosis, oxidative stress, and altered enzyme activity and metabolic pathways establish the base of this interaction between the kidneys and liver. In these concomitant kidney-liver diseases, the survival rates strongly correlate with early intervention and treatment of organ dysfunction. Proper care of a nephrologist and hepatologist and the identification of pathological conditions using biomarkers at early stages are necessary to prevent the complications induced by this complex and potentially vicious cycle. Therefore, understanding the characteristics of this crosstalk is essential for better management. In this review, we discussed the available literature concerning the detrimental effects of kidney failure on liver functions and liver-induced kidney diseases.
Collapse
Affiliation(s)
- Niloofar Khoshdel Rad
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Heydari
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov University, Moscow, Russia
| | - Amir Hossein Tamimi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Ensieh Zahmatkesh
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Anastasia Shpichka
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov University, Moscow, Russia
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
- Chemistry Department, Lomonosov Moscow State University, Moscow, Russia
| | - Maryam Barekat
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Peter Timashev
- World-Class Research Center "Digital Biodesign and Personalized Healthcare", Sechenov University, Moscow, Russia
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia.
- Chemistry Department, Lomonosov Moscow State University, Moscow, Russia
| | - Nikoo Hossein-Khannazer
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Moustapha Hassan
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran. ,
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
48
|
Mirrazavi ZS, Behrouz V. Various types of fasting diet and possible benefits in nonalcoholic fatty liver: Mechanism of actions and literature update. Clin Nutr 2024; 43:519-533. [PMID: 38219703 DOI: 10.1016/j.clnu.2024.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 12/28/2023] [Accepted: 01/04/2024] [Indexed: 01/16/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is one of the major causes of chronic liver injury, affecting around one-fourth of the general population across the world. Several important pathophysiological mechanisms underlying NAFLD include oxidative stress, inflammation, liver fibrosis, and apoptosis. Currently, therapeutic approaches are not ideal for managing NAFLD, thus new approaches and treatments are still needed. Over the last two decades, various fasting protocols have been explored to reduce body weight and improve metabolic disorders. In this review, we provide updated literature that supports fasting regimens for subjects with NAFLD and describes underlying mechanisms of action. We suggest that fasting regimens may modulate NAFLD via several mechanisms, including changes in gut microbiota, hepatic arginase, hepatic autophagy, inflammatory responses, liver functional enzymes and hepatic steatosis, fibroblast growth factors signaling, white adipose tissue browning, adipokines, circadian rhythms, lipid profiles, and body composition.
Collapse
Affiliation(s)
| | - Vahideh Behrouz
- Department of Nutrition, Faculty of Public Health, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
49
|
Kayalı A, Bora ES, Acar H, Erbaş O. Evaluation of the Reparative Effect of Sinomenine in an Acetaminophen-Induced Liver Injury Model. Curr Issues Mol Biol 2024; 46:923-933. [PMID: 38275673 PMCID: PMC10814253 DOI: 10.3390/cimb46010059] [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: 12/18/2023] [Revised: 01/09/2024] [Accepted: 01/18/2024] [Indexed: 01/27/2024] Open
Abstract
Due to its rising global prevalence, liver failure treatments are urgently needed. Sinomenine (SIN), an alkaloid from sinomenium acutum, is being studied for its liver-repair properties due to Acetaminophen (APAP) overdose. SIN's effect on APAP-induced hepatotoxicity in rats was examined histologically and biochemically. Three groups of 30 adult male Wistar rats were created: control, APAP-only, and APAP + SIN. Histopathological and biochemical analyses were performed on liver samples after euthanasia. SIN is significantly protected against APAP damage. Compared to APAP-only, SIN reduced cellular injury and preserved hepatocellular architecture. The APAP + SIN Group had significantly lower ALT, MDA, and GSH levels, protecting against hepatocellular damage and oxidative stress. SIN also had dose-dependent antioxidant properties. When examining critical regulatory proteins, SIN partially restored Sirtuin 1 (SIRT1) levels. While BMP-7 levels were unaffected, histopathological evidence and hepatocyte damage percentages supported SIN's liver-restorative effect. SIN protected and repaired rats' livers from APAP-induced liver injury. This study suggests that SIN may treat acute liver damage, warranting further research into its long-term effects, optimal dosage, and clinical applications. These findings aid liver-related emergency department interventions and life-saving treatments.
Collapse
Affiliation(s)
- Ahmet Kayalı
- Department of Emergency Medicine, Faculty of Medicine, Izmir Katip Çelebi University, Izmir 35270, Turkey; (A.K.); (H.A.)
| | - Ejder Saylav Bora
- Department of Emergency Medicine, Izmir Atatürk Research and Training Hospital, Izmir 35360, Turkey
| | - Hüseyin Acar
- Department of Emergency Medicine, Faculty of Medicine, Izmir Katip Çelebi University, Izmir 35270, Turkey; (A.K.); (H.A.)
| | - Oytun Erbaş
- Department of Physiology, Faculty of Medicine, Demiroğlu Bilim University, Istanbul 34395, Turkey;
| |
Collapse
|
50
|
Thayyil Menambath D, Adiga U, Rai T, Adiga S, Shetty V. Identification of the SIRT1 gene's most harmful non-synonymous SNPs and their effects on functional and structural features-an in silico analysis. F1000Res 2024; 12:66. [PMID: 38283900 PMCID: PMC10822041 DOI: 10.12688/f1000research.128706.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/16/2024] [Indexed: 01/30/2024] Open
Abstract
Introduction The sirtuin (Silent mating type information regulation 2 homolog)1(SIRT1) protein plays a vital role in many disorders such as diabetes, cancer, obesity, inflammation, and neurodegenerative and cardiovascular diseases. The objective of this in silico analysis of SIRT1's functional single nucleotide polymorphisms (SNPs) was to gain valuable insight into the harmful effects of non-synonymous SNPs (nsSNPs) on the protein. The objective of the study was to use bioinformatics methods to investigate the genetic variations and modifications that may have an impact on the SIRT1 gene's expression and function. Methods nsSNPs of SIRT1 protein were collected from the dbSNP site, from its three (3) different protein accession IDs. These were then fed to various bioinformatic tools such as SIFT, Provean, and I- Mutant to find the most deleterious ones. Functional and structural effects were examined using the HOPE server and I-Tasser. Gene interactions were predicted by STRING software. The SIFT, Provean, and I-Mutant tools detected the most deleterious three nsSNPs (rs769519031, rs778184510, and rs199983221). Results Out of 252 nsSNPs, SIFT analysis showed that 94 were deleterious, Provean listed 67 dangerous, and I-Mutant found 58 nsSNPs resulting in lowered stability of proteins. HOPE modelling of rs199983221 and rs769519031 suggested reduced hydrophobicity due to Ile 4Thr and Ile223Ser resulting in decreased hydrophobic interactions. In contrast, on modelling rs778184510, the mutant protein had a higher hydrophobicity than the wild type. Conclusions Our study reports that three nsSNPs (D357A, I223S, I4T) are the most damaging mutations of the SIRT1 gene. Mutations may result in altered protein structure and functions. Such altered protein may be the basis for various disorders. Our findings may be a crucial guide in establishing the pathogenesis of various disorders.
Collapse
Affiliation(s)
| | - Usha Adiga
- Biochemistry, KS Hegde Medical Academy, NITTE (DU), Mangalore, Karnataka, 575018, India
| | - Tirthal Rai
- Biochemistry, KS Hegde Medical Academy, NITTE (DU), Mangalore, Karnataka, 575018, India
| | - Sachidananda Adiga
- Pharmacology, KS Hegde Medical Academy, NITTE(DU), Mangalore, Karnataka, 575018, India
| | - Vijith Shetty
- Oncology, KS Hegde Medical Academy, NITTE(DU), Mangalore, Karnataka, 575018, India
| |
Collapse
|