1
|
Rajak S, Shah A, Yadav A, Shahi A, Raza S, Singh MM, Chaturvedi CP, Sinha RA. Inhibition of histone methyltransferase G9a aggravates phenotypic severity of hepatic lipotoxicity in non-alcoholic steatohepatitis (NASH). Biochem Biophys Res Commun 2025; 743:151171. [PMID: 39693938 DOI: 10.1016/j.bbrc.2024.151171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 12/10/2024] [Indexed: 12/20/2024]
Abstract
Lipotoxicity is a key pathological feature in the development of non-alcoholic steatohepatitis (NASH), which is characterized by liver injury, inflammation, and fibrosis. Although lipotoxicity has been shown to induce transcriptomic alterations in liver cells, the specific role of epigenetic regulators in NASH remains elusive. In this study, we demonstrate that pharmacological inhibition of histone methyltransferase G9a significantly worsens NASH progression in mice, as evidenced by increased hepatic cell death, inflammation, and fibrosis. Additionally, at a cellular level both genetic and pharmacological inhibition of G9a in HepG2 cells increased their susceptibility to palmitic acid-induced apoptosis and sub-cellular stress. Furthermore, treatment with G9a inhibitor enhanced TGF-β induced activation of primary human hepatic stellate cells (hHSCs), implicating the role of G9a in NASH pathobiology.
Collapse
Affiliation(s)
- Sangam Rajak
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, 226014, Lucknow, India
| | - Arunim Shah
- Stem Cell Research Centre, Department of Hematology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, 226014, Lucknow, India
| | - Abhishek Yadav
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, 226014, Lucknow, India
| | - Ambuj Shahi
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, 226014, Lucknow, India
| | - Sana Raza
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, 226014, Lucknow, India
| | - Mable M Singh
- Stem Cell Research Centre, Department of Hematology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, 226014, Lucknow, India
| | - Chandra P Chaturvedi
- Stem Cell Research Centre, Department of Hematology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, 226014, Lucknow, India.
| | - Rohit A Sinha
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, 226014, Lucknow, India.
| |
Collapse
|
2
|
Jin S, Li Y, Xia T, Liu Y, Zhang S, Hu H, Chang Q, Yan M. Mechanisms and therapeutic implications of selective autophagy in nonalcoholic fatty liver disease. J Adv Res 2025; 67:317-329. [PMID: 38295876 DOI: 10.1016/j.jare.2024.01.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/08/2024] Open
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) has become the most common chronic liver disease worldwide, whereas there is no approved drug therapy due to its complexity. Studies are emerging to discuss the role of selective autophagy in the pathogenesis of NAFLD, because the specificity among the features of selective autophagy makes it a crucial process in mitigating hepatocyte damage caused by aberrant accumulation of dysfunctional organelles, for which no other pathway can compensate. AIM OF REVIEW This review aims to summarize the types, functions, and dynamics of selective autophagy that are of particular importance in the initiation and progression of NAFLD. And on this basis, the review outlines the therapeutic strategies against NAFLD, in particular the medications and potential natural products that can modulate selective autophagy in the pathogenesis of this disease. KEY SCIENTIFIC CONCEPTS OF REVIEW The critical roles of lipophagy and mitophagy in the pathogenesis of NAFLD are well established, while reticulophagy and pexophagy are still being identified in this disease due to the insufficient understanding of their molecular details. As gradual blockage of autophagic flux reveals the complexity of NAFLD, studies unraveling the underlying mechanisms have made it possible to successfully treat NAFLD with multiple pharmacological compounds that target associated pathways. Overall, it is convinced that the continued research into selective autophagy occurring in NAFLD will further enhance the understanding of the pathogenesis and uncover novel therapeutic targets.
Collapse
Affiliation(s)
- Suwei Jin
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, China
| | - Yujia Li
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Tianji Xia
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, China
| | - Yongguang Liu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, China
| | - Shanshan Zhang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, China
| | - Hongbo Hu
- College of Food Science and Nutritional Engineering, China Agricultural University, China.
| | - Qi Chang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, China.
| | - Mingzhu Yan
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, China.
| |
Collapse
|
3
|
Ahmadizar F, Younossi ZM. Exploring Biomarkers in Nonalcoholic Fatty Liver Disease Among Individuals With Type 2 Diabetes Mellitus. J Clin Gastroenterol 2025; 59:36-46. [PMID: 39352015 PMCID: PMC11630663 DOI: 10.1097/mcg.0000000000002079] [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: 03/27/2024] [Accepted: 09/02/2024] [Indexed: 10/03/2024]
Abstract
Integrating biomarkers into a comprehensive strategy is crucial for precise patient management, especially considering the significant healthcare costs associated with diseases. Current studies emphasize the urgent need for a paradigm shift in conceptualizing nonalcoholic fatty liver disease (NAFLD), now renamed metabolic dysfunction-associated steatotic liver disease (MASLD). Biomarkers are emerging as indispensable tools for accurate diagnosis, risk stratification, and monitoring disease progression. This review classifies biomarkers into conventional and novel categories, such as lipids, insulin resistance, hepatic function, and cutting-edge imaging/omics, and evaluates their potential to transform the approach to MASLD among individuals with type 2 diabetes mellitus (T2D). It focuses on the critical role of biomarkers in early MASLD detection, enhancing predictive accuracy, and discerning responses to interventions (pharmacological or lifestyle modifications). Amid this discussion, the complexities of the relationship between T2D and MASLD are explored, considering factors like age, gender, genetics, ethnicity, and socioeconomic background. Biomarkers enhance the effectiveness of interventions and support global initiatives to reduce the burden of MASLD, thereby improving public health outcomes. This review recognizes the promising potential of biomarkers for diagnostic precision while candidly addressing the challenges in implementing these advancements in clinical practice. The transformative role of biomarkers emerges as a central theme, promising to reshape our understanding of disease trajectories, prognosis, and the customization of personalized therapeutic strategies for improved patient outcomes. From a future perspective, identifying early-stage biomarkers, understanding environmental impact through exposomes, and applying a multiomics approach may reveal additional insight into MASLD development.
Collapse
Affiliation(s)
- Fariba Ahmadizar
- Data Science and Biostatistics Department, Julius Global Health, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
- Beatty Liver and Obesity Research Program Center for Liver Diseases, Inova Health System, Falls Church, VA
| | - Zobair M. Younossi
- The Global NASH Council, Center for Outcomes Research in Liver Disease, Washington, DC
| |
Collapse
|
4
|
Tang C, Peng D, Zong K, Wu Z, Gong M, Li H, Huang Z, Li S. Association between the lymphocyte-to-high-density lipoprotein ratio and metabolic dysfunction-associated steatotic liver disease among US adults: a cross-sectional study from NHANES 2017 to 2020. BMC Gastroenterol 2024; 24:470. [PMID: 39716074 DOI: 10.1186/s12876-024-03565-5] [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: 07/31/2024] [Accepted: 12/13/2024] [Indexed: 12/25/2024] Open
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD) is a liver disease that is strongly associated with chronic low-grade inflammation. Stage 3 of MASLD is characterized by excessive formation of connective tissues, commonly referred to as liver fibrosis. Although numerous inflammatory markers have been identified and extensively studied, including the tumor necrosis factor-α and interleukin-6 have been studied [Byrne CD, Targher G. NAFLD: a multisystem disease. J Hepatol. 2015;62(1 Suppl):S47-64], the lymphocyte-to-high-density lipoprotein ratio (LHR) as a new biomarker that has not been sufficiently studied. This study aims to investigate the relationship between LHR levels and MASLD, determine its potential as a predictive marker for steatosis and fibrosis stages. METHODS This was a population-based study using data from 15,560 participants in the 2017-2020 National Health and Nutrition Examination Survey (NHANES) database. The study aimed to explore the relationship between LHR and MASLD. The disease progression was tracked by continuously measuring CAP and liver stiffness measurements. Participants who exhibited a median Controlled Attenuation Parameter (CAP) of 248 dB/m or higher were deemed to have hepatic steatosis. The LHR was calculated by dividing the lymphocyte count by the high-density lipoprotein cholesterol (HDL-C) level. Multivariate linear regression models were employed to explore the linear association between LHR and MASLD. Fitted smoothing curves and threshold effect analysis were employed to display nonlinear relationships. A two-part linear regression model was employed to estimate threshold effects. Subgroup analyses were conducted to determine the consistency of this association across various demographic groups. RESULTS A total of 6,950 adults aged 18 years and older were enrolled in the study, with an average age of 48.15 ± 17.10 years (49.14% male, 50.86% female). The adjusted multiple logistic regression analysis revealed a significant positive correlation between LHR and MASLD (OR: 1.64, 95% CI: 1.40-1.92). Using the complex two-piece linear regression model, we observed an inverted L-shaped association between LHR and CAP, suggesting a critical inflection point at -2.58. Subgroup analyses indicated a pronounced association of the LHR index with obese individuals (OR: 1.96, 95% CI: 1.66-2.32) and females (OR: 1.76, 95% CI: 1.25-2.46). There was no significant association between LHR and clinically significant fibrosis. CONCLUSION The LHR index is positively correlated with MASLD among US adults. Therefore, LHR may be a robust marker for early screening, diagnosis, and monitoring of treatment efficacy in clinical practice.
Collapse
Affiliation(s)
- Chuanzhi Tang
- Division of Hepatobiliary Pancreatic Surgery, First Affiliated Hospital, Chongqing Medical University, Chongqing, 400000, China
| | - Dadi Peng
- Division of Hepatobiliary Pancreatic Surgery, First Affiliated Hospital, Chongqing Medical University, Chongqing, 400000, China
| | - Kezhen Zong
- Division of Hepatobiliary Pancreatic Surgery, First Affiliated Hospital, Chongqing Medical University, Chongqing, 400000, China
| | - Zhongjun Wu
- Division of Hepatobiliary Pancreatic Surgery, First Affiliated Hospital, Chongqing Medical University, Chongqing, 400000, China
| | - Miao Gong
- Division of Hepatobiliary Pancreatic Surgery, First Affiliated Hospital, Chongqing Medical University, Chongqing, 400000, China
| | - Hui Li
- Chongqing University Cancer Hospital, Chongqing, 400000, China
| | - Zuotian Huang
- Division of Hepatobiliary Pancreatic Surgery, First Affiliated Hospital, Chongqing Medical University, Chongqing, 400000, China.
- Chongqing University Cancer Hospital, Chongqing, 400000, China.
| | - Shanshan Li
- Division of Hepatobiliary Pancreatic Surgery, First Affiliated Hospital, Chongqing Medical University, Chongqing, 400000, China.
| |
Collapse
|
5
|
Zuo R, Wang M, Wang YT, ShenTu Y, Moura AK, Zhou Y, Roudbari K, Hu JZ, Li PL, Hao J, Li X, Zhang Y. Ablation of Hepatic Asah1 Gene Disrupts Hepatic Lipid Homeostasis and Promotes Fibrotic Nonalcoholic Steatohepatitis in Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2024:S0002-9440(24)00444-9. [PMID: 39719015 DOI: 10.1016/j.ajpath.2024.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 10/14/2024] [Accepted: 11/06/2024] [Indexed: 12/26/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD) encompasses a spectrum of chronic liver conditions, ranging from simple steatosis to nonalcoholic steatohepatitis, which may progress to fibrosis/cirrhosis. Here, the GSE163211 data set was analyzed, and Asah1 (encoding acid ceramidase) was identified as a crucial lysosomal gene that positively correlated with NAFLD stages in obese patients. To evaluate the role of Asah1 in the progression of NAFLD, Asah1fl/fl/Albcre mice (hepatocyte-specific deletion of Asah1) and Asah1 floxed (Asah1fl/fl/wild-type) mice were fed with either a normal diet or a high-fat, high-cholesterol paigen diet (PD) for 20 weeks. The results showed that hepatocyte-specific Asah1 ablation markedly aggravated PD-induced hepatic steatosis, hepatitis, and apoptosis, and resulted in marked fibrotic changes. In addition, Asah1 gene ablation exacerbated PD-induced portal venous hemodynamic abnormality. In cultured hepatocytes, Asah1 gene knockdown resulted in increased ceramide and cholesterol levels but did not affect triglyceride level. Knocking down Asah1 gene also exhibited broad impacts on lipid homeostasis pathways, including lipogenesis, fatty acid uptake, fatty acid oxidation, and lipid transport. Furthermore, Asah1 knockdown resulted in increased endoplasmic reticulum stress and lipid droplet biogenesis. Last, Asah1 gene knockdown impaired chaperone-mediated autophagy. In conclusion, these results suggest that Asah1 functions as an important regulator of hepatic lipid homeostasis, and its deficiency exacerbates hepatocyte lipotoxicity and injury, and promotes the development of fibrotic nonalcoholic steatohepatitis.
Collapse
Affiliation(s)
- Rui Zuo
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas
| | - Mi Wang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas; Department of Gastroenterology and Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yun-Ting Wang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas
| | - YangPing ShenTu
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas; Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Alexandra K Moura
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas
| | - Ying Zhou
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas; Department Nephrology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Kiana Roudbari
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas
| | - Jenny Z Hu
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas
| | - Pin-Lan Li
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, School of Medicine, Richmond, Virginia
| | - JiuKuan Hao
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas
| | - Xiang Li
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas
| | - Yang Zhang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas.
| |
Collapse
|
6
|
Shen Y, Qiu A, Huang X, Wen X, Shehzadi S, He Y, Hu Q, Zhang J, Luo D, Yang S. AKR1B10 and digestive tumors development: a review. Front Immunol 2024; 15:1462174. [PMID: 39737179 PMCID: PMC11682995 DOI: 10.3389/fimmu.2024.1462174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 11/26/2024] [Indexed: 01/01/2025] Open
Abstract
Aldo-keto reductase family 1 member B10 (AKR1B10) is a member of the AKR1B subfamily. It is mainly found in cytoplasm, and it is typically expressed in the stomach and intestines. Given that its expression is low or absent in other tissues, AKR1B10 is a potential diagnostic and therapeutic biomarker for various digestive system diseases. Here, we review recent research progress on AKR1B10 in digestive system tumors such as hepatocellular carcinoma, gastric carcinoma, colorectal carcinoma, pancreatic carcinoma, oral squamous cell carcinoma, laryngeal squamous cell carcinoma, cholangiocarcinoma, and nasopharyngeal carcinoma, over the last 5 years. We also discuss the current trends and future research directions for AKR1B10 in both oncological and non-oncological diseases to provide a scientific reference for further exploration of this gene.
Collapse
Affiliation(s)
- Yao Shen
- Medical School, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Ailin Qiu
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xin Huang
- Laboratory Medicine Center, Shenzhen Luohu Hospital Group, the Third Affiliated Hospital (The Affiliated Luohu Hospital) of Shenzhen University, Shenzhen University, Shenzhen, Guangdong, China
- First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Xiaosha Wen
- Laboratory Medicine Center, Shenzhen Luohu Hospital Group, the Third Affiliated Hospital (The Affiliated Luohu Hospital) of Shenzhen University, Shenzhen University, Shenzhen, Guangdong, China
| | - Sundar Shehzadi
- Laboratory Medicine Center, Shenzhen Luohu Hospital Group, the Third Affiliated Hospital (The Affiliated Luohu Hospital) of Shenzhen University, Shenzhen University, Shenzhen, Guangdong, China
| | - Yan He
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Qian Hu
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jian Zhang
- Medical School, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Dixian Luo
- Laboratory Medicine Center, Shenzhen Luohu Hospital Group, the Third Affiliated Hospital (The Affiliated Luohu Hospital) of Shenzhen University, Shenzhen University, Shenzhen, Guangdong, China
| | - Shenghui Yang
- Medical School, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Department of Preventive Medicine, Medical School, Hunan University of Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
7
|
Chhetri A, Park C, Kim H, Manandhar L, Chuluunbaatar C, Hwang J, Wei X, Jang G, Chinbold B, Kwon HM, Lee SW, Park R. TMEM135 deficiency improves hepatic steatosis by suppressing CD36 in a SIRT1-dependent manner. Mol Metab 2024; 92:102080. [PMID: 39647810 DOI: 10.1016/j.molmet.2024.102080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 11/19/2024] [Accepted: 11/28/2024] [Indexed: 12/10/2024] Open
Abstract
OBJECTIVES Dysregulation of lipid homeostasis pathway causes many liver diseases, including hepatic steatosis. One of the primary factors contributing to lipid accumulation is fatty acid uptake by the liver. Transmembrane protein 135 (TMEM135), which exists in mitochondria and peroxisomes, participates in intracellular lipid metabolism. This study aims to investigate the role of TMEM135 on regulating cellular lipid import in the liver. METHODS We used in vivo, ex vivo, and in vitro models of steatosis. TMEM135 knockout (TMEM135KO) and wild type (WT) mice were fed a high-fat diet (HFD) to induce hepatic steatosis. Primary mouse hepatocytes and AML12 cells were treated with free fatty acid (FFA). Additionally, TMEM135-deficient stable cells and overexpressed cells were established using AML12 cells. RESULTS TMEM135 deficiency mitigated lipid accumulation in the liver of HFD-fed TMEM135KO mice. TMEM135-depleted primary hepatocytes and AML12 cells exhibited less lipid accumulation when treated with FFA compared to control cells, as shown as lipid droplets. Consistently, the effect of TMEM135 depletion on lipid accumulation was completely reversed under TMEM135 overexpression conditions. CD36 expression was markedly induced by HFD or FFA, which was reduced by TMEM135 depletion. Among the SIRT family proteins, only SIRT1 expression definitely increased in the liver of HFD-fed TMEM135KO mice along with a significant increase in NAD+/NADH ratio. However, inhibition of SIRT1 in TMEM135-depleted cells using siSIRT1 or the SIRT1 inhibitor EX-527 resulted in an increase of CD36 expression and consequent TG levels. CONCLUSIONS TMEM135 depletion attenuates CD36 expression in a SIRT1-dependent manner, thereby reducing cellular lipid uptake and hepatic steatosis.
Collapse
Affiliation(s)
- Arun Chhetri
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Channy Park
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Hyunsoo Kim
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Laxman Manandhar
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Chagtsalmaa Chuluunbaatar
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Jaetaek Hwang
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Xiaofan Wei
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Gyuho Jang
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Batching Chinbold
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Hyug Moo Kwon
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Sang-Wook Lee
- Department of Radiation Oncology Asan Medical Center, Seoul, Republic of Korea
| | - Raekil Park
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea.
| |
Collapse
|
8
|
Zhang SH, Zhang HJ, Jia YZ, Wang ZY, You ZH, Lian CY, Wang L. Melatonin prevents glyphosate-induced hepatic lipid accumulation in roosters via activating Nrf2 pathway. Int Immunopharmacol 2024; 142:113180. [PMID: 39305889 DOI: 10.1016/j.intimp.2024.113180] [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: 06/14/2024] [Revised: 08/28/2024] [Accepted: 09/12/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND Glyphosate (GLY) is a widely used herbicide with well-defined hepatotoxic effects, in which oxidative stress has been shown to be involved in the pathogenesis of hepatotoxicity. Melatonin (MET), an effective free radical scavenger, has been revealed to alleviate drug-induced liver damage by inhibiting oxidative stress. METHODS In this study, a rooster model with primary chicken embryo hepatocytes was applied to elucidate the therapeutic effects of MET against GLY-induced hepatic damage and the potential mechanism. Histopathological examinations, biochemical tests and immunoblotting analysis were used to monitor the protective effects of MET on GLY-induced hepatic lipid accumulation. Molecular docking analysis was used to reveal the key reason of MET-improved hepatic lipid deposition. RESULTS Data firstly showed that MET administration markedly improved GLY-induced hepatic injury, as evidenced by normalized liver enzymes and alleviated pathological changes of liver tissues. Moreover, MET supplementation alleviated GLY-induced hepatic lipid accumulation, which was correlated with improved serum and hepatic lipid profiles and normalized expression of lipolysis- and lipogenesis-related proteins. Notably, MET significantly inhibited vital enzymes involved in stimulating oxidative stress. Moreover, MET enhanced GLY-inhibited Nrf2 nuclear transcription and increased the expressions of its downstream target genes HO1 and NQO1. Further studies revealed that MET may interact with Nrf2 to enhance nuclear translocation of Nrf2. CONCLUSION Collectively, our results provide the first direct evidence that MET is a novel regulator of Nrf2, highlighting that Nrf2 may be a potential therapeutic target for GLY-induced lipotoxic liver injury.
Collapse
Affiliation(s)
- Shu-Hui Zhang
- College of Veterinary Medicine, Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, 7 Panhe Street, Tai'an City, Shandong Province 271017, China
| | - Hai-Jing Zhang
- New Drug Evaluation Center of Shandong Academy of Pharmaceutical Sciences, Shandong Academy of Pharmaceutical Sciences, 989 Xinluo Street, Ji'nan City 250101, Shandong Province, China
| | - Yan-Zhan Jia
- College of Veterinary Medicine, Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, 7 Panhe Street, Tai'an City, Shandong Province 271017, China
| | - Zhen-Yong Wang
- College of Veterinary Medicine, Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, 7 Panhe Street, Tai'an City, Shandong Province 271017, China
| | - Zhao-Hong You
- Zaozhuang University School of Food Science and Pharmaceutical Engineering, No.1, Beian Road, Shizhong District, Zaozhuang City, Shandong Province 277160, China
| | - Cai-Yu Lian
- College of Veterinary Medicine, Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, 7 Panhe Street, Tai'an City, Shandong Province 271017, China.
| | - Lin Wang
- College of Veterinary Medicine, Shandong Provincial Key Laboratory of Zoonoses, Shandong Agricultural University, 7 Panhe Street, Tai'an City, Shandong Province 271017, China.
| |
Collapse
|
9
|
Heinrich NS, Pedersen RP, Vestergaard MB, Lindberg U, Andersen UB, Haddock B, Fornoni A, Larsson HBW, Rossing P, Hansen TW. Kidney fat by magnetic resonance spectroscopy in type 2 diabetes with chronic kidney disease. J Diabetes Complications 2024; 39:108923. [PMID: 39647261 DOI: 10.1016/j.jdiacomp.2024.108923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 11/05/2024] [Accepted: 11/29/2024] [Indexed: 12/10/2024]
Abstract
BACKGROUND AND HYPOTHESIS The kidneys may be susceptible to ectopic fat and its lipotoxic effects, disposing them to chronic kidney disease (CKD) in type 2 diabetes (T2D). We investigated whether the kidney parenchyma fat content and kidney sinus fat volume would be higher in persons with T2D and CKD. METHODS Cross-sectional study including 29 controls, 27 persons with T2D and no CKD, and 48 persons with T2D and early CKD (urine albumin creatinine ratio (UACR) ≥ 30 mg/g). Kidney parenchyma fat content and kidney sinus fat volume were assessed using magnetic resonance spectroscopy and Dixon scans respectively. RESULTS In the control, T2D without CKD and T2D with CKD groups, respectively, median [1st - 3rd quartile] UACR was 5 [4 - 6], 6 [5 - 10] and 95 [43 - 278] mg/g. and mean ± standard deviation estimated glomerular filtration rate was 89 ± 11, 94 ± 11 and 77 ± 22 ml/min/1.73m2. Kidney parenchyma fat content was, respectively, 1.0 [0.5-2.4], 0.7 [0.2-1.2], 1.0 [0.3-2.0] % (p = 0.26). Kidney sinus fat volume was 2.8 [1.6-7.6], 8.0 [4.7-11.3], 10.3 [5.7-14.0] ml (p < 0.01). Around 90 % of T2D participants received a sodium-glucose cotransporter-2 inhibitor or glucagon-like peptide-1 receptor agonist. CONCLUSIONS In a setting of modern, multifactorial T2D management, kidney parenchyma fat content, evaluated with magnetic resonance spectroscopy, was similar among healthy controls and persons with T2D irrespective of CKD status. Still, kidney sinus fat volume was higher in the presence of T2D and higher still with CKD.
Collapse
Affiliation(s)
| | - Rune Ploegstra Pedersen
- Department of Clinical Physiology and Nuclear Medicine at Rigshospitalet Glostrup, Glostrup, Denmark
| | - Mark Bitsch Vestergaard
- Department of Clinical Physiology and Nuclear Medicine at Rigshospitalet Glostrup, Glostrup, Denmark
| | - Ulrich Lindberg
- Department of Clinical Physiology and Nuclear Medicine at Rigshospitalet Glostrup, Glostrup, Denmark
| | - Ulrik Bjørn Andersen
- Department of Clinical Physiology and Nuclear Medicine at Rigshospitalet Glostrup, Glostrup, Denmark
| | - Bryan Haddock
- Department of Clinical Physiology and Nuclear Medicine at Rigshospitalet Glostrup, Glostrup, Denmark
| | - Alessia Fornoni
- Katz Family Division of Nephrology and Hypertension and Peggy and Harold Katz Drug Discovery Center, Miami, USA
| | - Henrik Bo Wiberg Larsson
- Department of Clinical Physiology and Nuclear Medicine at Rigshospitalet Glostrup, Glostrup, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Peter Rossing
- Steno Diabetes Center Copenhagen, Herlev, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | | |
Collapse
|
10
|
Cui Y, Yang K, Guo C, Xia Z, Jiang B, Xue Y, Song B, Hu W, Zhang M, Wei Y, Zhang C, Zhang S, Fang J. Carbon monoxide as a negative feedback mechanism on HIF-1α in the progression of metabolic-associated fatty liver disease. Nitric Oxide 2024; 153:1-12. [PMID: 39369813 DOI: 10.1016/j.niox.2024.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/01/2024] [Accepted: 10/03/2024] [Indexed: 10/08/2024]
Abstract
Metabolic-associated fatty liver disease (MAFLD) encompasses various chronic liver conditions, yet lacks approved drugs. Hypoxia-inducible factor-1α (HIF-1α) is pivotal in MAFLD development. Our prior research highlighted the efficacy of the nano-designed carbon monoxide (CO) donor, targeting HIF-1α in a mouse hepatic steatosis model. Given heme oxygenase-1 (HO-1, a major downstream molecule of HIF-1α) as the primary source of intrinsic CO, we hypothesized that upregulation of HO-1/CO, responsive to HIF-1α, forms a negative feedback loop regulating MAFLD progression. In this study, we explored the potential negative feedback mechanism of CO on HIF-1α and its downstream effects on MAFLD advancement. HIF-1α emerges early in hepatic steatosis induced by a high-fat (HF) diet, triggering increased HO-1 and inflammation. SMA/CORM2 effectively suppresses HIF-1α and steatosis progression when administered within the initial week of HF diet initiation but loses impact later. In adipose tissues, concurrent metabolic dysfunction and inflammation with HIF-1α activation suggest adipose tissue expansion initiates HF-induced steatosis, triggering hypoxia and liver inflammation. Notably, in an in vitro study using mouse hepatocytes treated with fatty acids, downregulating HO-1 intensified HIF-1α induction at moderate fatty acid concentrations. However, this effect diminished at high concentrations. These results suggest the HIF-1α-HO-1-CO axis as a feedback loop under physiological and mild pathological conditions. Excessive HIF-1α upregulation in pathological conditions overwhelms the CO feedback loop. Additional CO application effectively suppresses HIF-1α and disease progression, indicating potential application for MAFLD control.
Collapse
Affiliation(s)
- Yingying Cui
- Peking University First Hospital Ningxia Women and Children's Hospital (Ningxia Hui Autonomous Region Maternal and Child Health Hospital), Yinchuan, 750000, China; Department of Toxicology, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China
| | - Kai Yang
- Department of Medical Technology, Anhui Medical College, No.632, Furong Road, Hefei, Anhui Province, China
| | - Chunyu Guo
- Department of Toxicology, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China
| | - Zhengmei Xia
- Department of Toxicology, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China
| | - Benchun Jiang
- Department of Gastricintestinal Surgery, Shengjing Hospital of China Medical University, No.36 Sanhao Street, Shenyang, 110004, Liaoning, China
| | - Yanni Xue
- Department of Toxicology, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China
| | - Bingdong Song
- Department of Toxicology, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China
| | - Weirong Hu
- Department of Toxicology, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China
| | - Mingjie Zhang
- Department of General Surgery, Shengjing Hospital of China Medical University, No.36 Sanhao Street, Shenyang, 110004, Liaoning, China
| | - Yanyan Wei
- Department of Infectious Disease, the First Affiliated Hospital of Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Cheng Zhang
- Department of Toxicology, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China
| | - Shichen Zhang
- Anhui Provincial Center for Maternal and Child Health Genetics, School of Public Health and Health Management, Anhui Medical College, No 632 Furong Road, Hefei, 230601, Anhui, China.
| | - Jun Fang
- Department of Toxicology, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China; Anhui Provincial Center for Maternal and Child Health Genetics, School of Public Health and Health Management, Anhui Medical College, No 632 Furong Road, Hefei, 230601, Anhui, China; Faculty of Pharmaceutical Sciences, Sojo University, Ikeda 4-22-1, Kumamoto, 860-0082, Japan.
| |
Collapse
|
11
|
Li W, Yu L. Role and therapeutic perspectives of extracellular vesicles derived from liver and adipose tissue in metabolic dysfunction-associated steatotic liver disease. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2024; 52:355-369. [PMID: 38833340 DOI: 10.1080/21691401.2024.2360008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 05/22/2024] [Indexed: 06/06/2024]
Abstract
The global epidemic of metabolic diseases has led to the emergence of metabolic dysfunction-associated steatotic liver disease (MASLD) and metabolic dysfunction-associated steatohepatitis (MASH), which pose a significant threat to human health. Despite recent advances in research on the pathogenesis and treatment of MASLD/MASH, there is still a lack of more effective and targeted therapies. Extracellular vesicles (EVs) discovered in a wide range of tissues and body fluids encapsulate different activated biomolecules and mediate intercellular communication. Recent studies have shown that EVs derived from the liver and adipose tissue (AT) play vital roles in MASLD/MASH pathogenesis and therapeutics, depending on their sources and intervention types. Besides, adipose-derived stem cell (ADSC)-derived EVs appear to be more effective in mitigating MASLD/MASH. This review presents an overview of the definition, extraction strategies, and characterisation of EVs, with a particular focus on the biogenesis and release of exosomes. It also reviews the effects and potential molecular mechanisms of liver- and AT-derived EVs on MASLD/MASH, and emphasises the contribution and clinical therapeutic potential of ADSC-derived EVs. Furthermore, the future perspective of EV therapy in a clinical setting is discussed.
Collapse
Affiliation(s)
- Wandi Li
- Senior Department of Burns and Plastic Surgery, the Fourth Medical Center of PLA General Hospital, Haidian District, Beijing, P.R. China
| | - Lili Yu
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, P.R. China
- Endocrine Department, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang Medical University, Henan, P.R. China
| |
Collapse
|
12
|
Moliterni C, Vari F, Schifano E, Tacconi S, Stanca E, Friuli M, Longo S, Conte M, Salvioli S, Gnocchi D, Mazzocca A, Uccelletti D, Vergara D, Dini L, Giudetti AM. Lipotoxicity of palmitic acid is associated with DGAT1 downregulation and abolished by PPARα activation in liver cells. J Lipid Res 2024; 65:100692. [PMID: 39505261 DOI: 10.1016/j.jlr.2024.100692] [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: 04/26/2024] [Revised: 10/21/2024] [Accepted: 10/31/2024] [Indexed: 11/08/2024] Open
Abstract
Lipotoxicity refers to the harmful effects of excess fatty acids on metabolic health, and it can vary depending on the type of fatty acids involved. Saturated and unsaturated fatty acids exhibit distinct effects, though the precise mechanisms behind these differences remain unclear. Here, we investigated the lipotoxicity of palmitic acid (PA), a saturated fatty acid, compared with oleic acid (OA), a monounsaturated fatty acid, in the hepatic cell line HuH7. Our results demonstrated that PA, unlike OA, induces lipotoxicity, endoplasmic reticulum (ER) stress, and autophagy inhibition. Compared with OA, PA treatment leads to less lipid droplet (LD) accumulation and a significant reduction in the mRNA and protein level of diacylglycerol acyltransferase 1 (DGAT1), a key enzyme of triacylglycerol synthesis. Using modulators of ER stress and autophagy, we established that DGAT1 downregulation by PA is closely linked to these cellular pathways. Notably, the ER stress inhibitor 4-phenylbutyrate can suppress PA-induced DGAT1 downregulation. Furthermore, knockdown of DGAT1 by siRNA or with A922500, a specific DGAT1 inhibitor, resulted in cell death, even with OA. Both PA and OA increased the oxygen consumption rate; however, the increase associated with PA was only partially coupled to ATP synthesis. Importantly, treatment with GW7647 a specific PPARα agonist mitigated the lipotoxic effects of PA, restoring PA-induced ER stress, autophagy block, and DGAT1 suppression. In conclusion, our study highlights the crucial role of DGAT1 in PA-induced lipotoxicity, broadening the knowledge of the mechanisms underlying hepatic lipotoxicity and providing the basis for potential therapeutic interventions.
Collapse
Affiliation(s)
- Camilla Moliterni
- Department of Biology and Biotechnology "C. Darwin", Sapienza University of Rome, Rome, Italy
| | - Francesco Vari
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy; Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome, Italy
| | - Emily Schifano
- Department of Biology and Biotechnology "C. Darwin", Sapienza University of Rome, Rome, Italy
| | - Stefano Tacconi
- Department of Biology and Biotechnology "C. Darwin", Sapienza University of Rome, Rome, Italy
| | - Eleonora Stanca
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Marzia Friuli
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Rome, Italy
| | - Serena Longo
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Maria Conte
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Stefano Salvioli
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy; IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Davide Gnocchi
- Interdisciplinary Department of Medicine, University of Bari School of Medicine, Bari, Italy
| | - Antonio Mazzocca
- Interdisciplinary Department of Medicine, University of Bari School of Medicine, Bari, Italy
| | - Daniela Uccelletti
- Department of Biology and Biotechnology "C. Darwin", Sapienza University of Rome, Rome, Italy
| | - Daniele Vergara
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Luciana Dini
- Department of Biology and Biotechnology "C. Darwin", Sapienza University of Rome, Rome, Italy.
| | - Anna Maria Giudetti
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy.
| |
Collapse
|
13
|
Wang J, Li Q, Huo Y, Liu X, Shi Y, Xie B. ASPP2 deficiency promotes the progression of metabolic dysfunction-associated steatohepatitis via ACSL4 upregulation. Sci Rep 2024; 14:29177. [PMID: 39587161 PMCID: PMC11589572 DOI: 10.1038/s41598-024-80415-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 11/18/2024] [Indexed: 11/27/2024] Open
Abstract
As a member of the p53-binding protein family, apoptosis-stimulating protein p53 2 (ASPP2) is closely related to autophagy and apoptosis. However, the mechanistic role of ASPP2 in the development of metabolic dysfunction-associated steatohepatitis (MASH) remains elusive. Therefore, we investigated the role and underlying mechanisms of ASPP2 in MASH progression in a mouse model of MASH and a cellular model of metabolic dysfunction-associated fatty liver disease. ASPP2 deficiency significantly promoted the inflammatory response, steatosis, and MASH progression in mice. Through transcriptomic analysis, increased ACSL4 expression was identified as a potential key factor. Further elucidation of the underlying mechanisms demonstrated that ASPP2 deficiency increased lipid accumulation and inhibited mitochondrial respiration capacity in HepG2 cells induced by oleic acid. However, silencing of ACSL4 reversed these effects. Thus, our study indicates that ASPP2 is an important regulator of MASH progression through ACSL4 upregulation, highlighting its potential as an alternative approach to MASH treatment.
Collapse
Affiliation(s)
- Jinming Wang
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Quanwei Li
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Yunfei Huo
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Xiaoni Liu
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Ying Shi
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China.
| | - Bangxiang Xie
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
14
|
Shou JW, Ma J, Wang X, Li XX, Chen SC, Kang BH, Shaw PC. Free Cholesterol-Induced Liver Injury in Non-Alcoholic Fatty Liver Disease: Mechanisms and a Therapeutic Intervention Using Dihydrotanshinone I. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2406191. [PMID: 39558866 DOI: 10.1002/advs.202406191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 08/28/2024] [Indexed: 11/20/2024]
Abstract
Build-up of free cholesterol (FC) substantially contributes to the development and severity of non-alcoholic fatty liver disease (NAFLD). Here, we investigate the specific mechanism by which FC induces liver injury in NAFLD and propose a novel therapeutic approach using dihydrotanshinone I (DhT). Rather than cholesterol ester (CE), we observed elevated levels of total cholesterol, FC, and alanine transaminase (ALT) in NAFLD patients and high-cholesterol diet-induced NAFLD mice compared to those in healthy controls. The FC level demonstrated a positive correlation with the ALT level in both patients and mice. Mechanistic studies revealed that FC elevated reactive oxygen species level, impaired the function of lysosomes, and disrupted lipophagy process, consequently inducing cell apoptosis. We then found that DhT protected mice on an HCD diet, independent of gut microbiota. DhT functioned as a potent ligand for peroxisome proliferator-activated receptor α (PPARα), stimulating its transcriptional function and enhancing catalase expression to lower reactive oxygen species (ROS) level. Notably, the protective effect of DhT was nullified in mice with hepatic PPARα knockdown. Thus, these findings are the first to report the detrimental role of FC in NAFLD, which could lead to the development of new treatment strategies for NAFLD by leveraging the therapeutic potential of DhT and PPARα pathway.
Collapse
Affiliation(s)
- Jia-Wen Shou
- Li Dak Sum Yip Yio Chin R&D Centre for Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, 852852, China
| | - Juncai Ma
- Centre for Cell and Developmental Biology, State Key Laboratory for Agrobiotechnology, School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, 852852, China
| | - Xuchu Wang
- Department of Laboratory Medicine, the Second Affiliated Hospital of Zhejiang University, Hangzhou, 310000, China
| | - Xiao-Xiao Li
- Li Dak Sum Yip Yio Chin R&D Centre for Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, 852852, China
- Research Center for Chinese Medicine Innovation, The Hong Kong Polytechnic University, Hong Kong, 852852, China
| | - Shu-Cheng Chen
- School of Nursing, The Hong Kong Polytechnic University, Hong Kong, 852852, China
| | - Byung-Ho Kang
- Centre for Cell and Developmental Biology, State Key Laboratory for Agrobiotechnology, School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, 852852, China
| | - Pang-Chui Shaw
- Li Dak Sum Yip Yio Chin R&D Centre for Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, 852852, China
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, 852852, China
- State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants and Institute of Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, 852852, China
| |
Collapse
|
15
|
Soluyanova P, Quintás G, Pérez-Rubio Á, Rienda I, Moro E, van Herwijnen M, Verheijen M, Caiment F, Pérez-Rojas J, Trullenque-Juan R, Pareja E, Jover R. The Development of a Non-Invasive Screening Method Based on Serum microRNAs to Quantify the Percentage of Liver Steatosis. Biomolecules 2024; 14:1423. [PMID: 39595599 PMCID: PMC11592063 DOI: 10.3390/biom14111423] [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: 10/08/2024] [Revised: 10/28/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is often asymptomatic and underdiagnosed; consequently, there is a demand for simple, non-invasive diagnostic tools. In this study, we developed a method to quantify liver steatosis based on miRNAs, present in liver and serum, that correlate with liver fat. The miRNAs were analyzed by miRNAseq in liver samples from two cohorts of patients with a precise quantification of liver steatosis. Common miRNAs showing correlation with liver steatosis were validated by RT-qPCR in paired liver and serum samples. Multivariate models were built using partial least squares (PLS) regression to predict the percentage of liver steatosis from serum miRNA levels. Leave-one-out cross validation and external validation were used for model selection and to estimate predictive performance. The miRNAseq results disclosed (a) 144 miRNAs correlating with triglycerides in a set of liver biobank samples (n = 20); and (b) 124 and 102 miRNAs correlating with steatosis by biopsy digital image and MRI analyses, respectively, in liver samples from morbidly obese patients (n = 24). However, only 35 miRNAs were common in both sets of samples. RT-qPCR allowed to validate the correlation of 10 miRNAs in paired liver and serum samples. The development of PLS models to quantitatively predict steatosis demonstrated that the combination of serum miR-145-3p, 122-5p, 143-3p, 500a-5p, and 182-5p provided the lowest root mean square error of cross validation (RMSECV = 1.1, p-value = 0.005). External validation of this model with a cohort of mixed MASLD patients (n = 25) showed a root mean squared error of prediction (RMSEP) of 5.3. In conclusion, it is possible to predict the percentage of hepatic steatosis with a low error rate by quantifying the serum level of five miRNAs using a cost-effective and easy-to-implement RT-qPCR method.
Collapse
Affiliation(s)
- Polina Soluyanova
- Unidad Mixta de Investigación en Hepatología Experimental, IIS Hospital La Fe, 46026 Valencia, Spain; (P.S.); (E.M.)
- Departamento de Bioquímica y Biología Molecular, Universidad de Valencia, 46010 Valencia, Spain
| | - Guillermo Quintás
- Health and Biomedicine, LEITAT Technological Center, 08225 Terrassa, Spain;
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), ISCIII, 28029 Madrid, Spain
| | - Álvaro Pérez-Rubio
- Servicio de Cirugía General y Aparato Digestivo, Hospital Universitario Dr. Peset, 46017 Valencia, Spain; (Á.P.-R.); (E.P.)
| | - Iván Rienda
- Pathology Department, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain; (I.R.); (J.P.-R.)
| | - Erika Moro
- Unidad Mixta de Investigación en Hepatología Experimental, IIS Hospital La Fe, 46026 Valencia, Spain; (P.S.); (E.M.)
- Departamento de Bioquímica y Biología Molecular, Universidad de Valencia, 46010 Valencia, Spain
| | - Marcel van Herwijnen
- Department of Translational Genomics, Research Institute of Oncology and Developmental Biology (GROW), Maastricht University, 6229-ER Maastricht, The Netherlands; (M.v.H.); (M.V.); (F.C.)
| | - Marcha Verheijen
- Department of Translational Genomics, Research Institute of Oncology and Developmental Biology (GROW), Maastricht University, 6229-ER Maastricht, The Netherlands; (M.v.H.); (M.V.); (F.C.)
| | - Florian Caiment
- Department of Translational Genomics, Research Institute of Oncology and Developmental Biology (GROW), Maastricht University, 6229-ER Maastricht, The Netherlands; (M.v.H.); (M.V.); (F.C.)
| | - Judith Pérez-Rojas
- Pathology Department, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain; (I.R.); (J.P.-R.)
| | - Ramón Trullenque-Juan
- Servicio de Cirugía General y Aparato Digestivo, Hospital Universitario Dr. Peset, 46017 Valencia, Spain; (Á.P.-R.); (E.P.)
| | - Eugenia Pareja
- Servicio de Cirugía General y Aparato Digestivo, Hospital Universitario Dr. Peset, 46017 Valencia, Spain; (Á.P.-R.); (E.P.)
| | - Ramiro Jover
- Unidad Mixta de Investigación en Hepatología Experimental, IIS Hospital La Fe, 46026 Valencia, Spain; (P.S.); (E.M.)
- Departamento de Bioquímica y Biología Molecular, Universidad de Valencia, 46010 Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), ISCIII, 28029 Madrid, Spain
| |
Collapse
|
16
|
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
|
17
|
Amorim R, Soares P, Chavarria D, Benfeito S, Cagide F, Teixeira J, Oliveira PJ, Borges F. Decreasing the burden of non-alcoholic fatty liver disease: From therapeutic targets to drug discovery opportunities. Eur J Med Chem 2024; 277:116723. [PMID: 39163775 DOI: 10.1016/j.ejmech.2024.116723] [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: 05/30/2024] [Revised: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 08/22/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD) presents a pervasive global pandemic, affecting approximately 25 % of the world's population. This grave health issue not only demands urgent attention but also stands as a significant economic concern on a global scale. The genesis of NAFLD can be primarily attributed to unhealthy dietary habits and a sedentary lifestyle, albeit certain genetic factors have also been recorded to contribute to its occurrence. NAFLD is characterized by fat accumulation in more than 5 % of hepatocytes according to histological analysis, or >5.6 % of lipid volume fraction in total liver weight in patients. The pathophysiology of NAFLD/non-alcoholic steatohepatitis (NASH) is multifactorial and the mechanisms underlying the progression to advanced forms remain unclear, thereby representing a challenge to disease therapy. Despite the substantial efforts from the scientific community and the large number of pre-clinical and clinical trials performed so far, only one drug was approved by the Food and Drug Administration (FDA) to treat NAFLD/NASH specifically. This review provides an overview of available information concerning emerging molecular targets and drug candidates tested in clinical studies for the treatment of NAFLD/NASH. Improving our understanding of NAFLD pathophysiology and pharmacotherapy is crucial not only to explore new molecular targets, but also to potentiate drug discovery programs to develop new therapeutic strategies. This knowledge endeavours scientific efforts to reduce the time for achieving a specific and effective drug for NAFLD or NASH management and improve patients' quality of life.
Collapse
Affiliation(s)
- Ricardo Amorim
- CNC-UC, Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; CIBB, Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Portugal
| | - Pedro Soares
- CIQUP-IMS/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal
| | - Daniel Chavarria
- CIQUP-IMS/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal
| | - Sofia Benfeito
- CIQUP-IMS/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal
| | - Fernando Cagide
- CIQUP-IMS/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal
| | - José Teixeira
- CNC-UC, Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; CIBB, Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Portugal
| | - Paulo J Oliveira
- CNC-UC, Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; CIBB, Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Portugal.
| | - Fernanda Borges
- CIQUP-IMS/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal.
| |
Collapse
|
18
|
Anwar S, Siddique R, Ahmad S, Haider MZ, Ali H, Sami A, Lucas RS, Shafiq M, Nisa BU, Javed B, Akram J, Tabassum J, Javed MA. Genome wide identification and characterization of Bax inhibitor-1 gene family in cucumber (Cucumis sativus) under biotic and abiotic stress. BMC Genomics 2024; 25:1032. [PMID: 39497028 PMCID: PMC11536926 DOI: 10.1186/s12864-024-10704-5] [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: 01/24/2024] [Accepted: 08/12/2024] [Indexed: 11/06/2024] Open
Abstract
In plants, the BAX inhibitor-1 (BI-1) gene plays a crucial part in controlling cell death under stress conditions. This mechanism of Programmed Cell Death (PCD) is genetically regulated and is crucial for the elimination of unwanted or damaged cells in a controlled manner, which is essential for normal development and tissue maintenance. A study on cucumber identified and characterized five BI-1 genes: CsBI1, CsBI2, CsBI3, CsBI4, and CsBI5. These genes share conserved domains, indicating common evolutionary history and function. Physicochemical analysis revealed their molecular weights and isoelectric points, while subcellular localization showed their presence in different cellular compartments. The phylogenetic analysis highlighted evolutionary relationships with related crops. Chromosomal distribution and synteny analysis suggested segmental or tandem duplications within the gene family. Protein-protein interaction analysis revealed extensive interactions with other cucumber proteins. Cis-regulatory elements in the promoter regions provided insights into potential functions and transcriptional regulation. miRNAs showed diverse regulatory mechanisms, including mRNA cleavage and translational inhibition. The CsBI3, CsBI4 and CsBI5 genes exhibit elevated expression levels during cold stress, suggesting their vital involvement in cucumber plant defense mechanisms. The application of chitosan oligosaccharides externally confirms their distinct expression patterns. The qRT-PCR confirms the upregulation of CsBI genes in ToLCNDV-infected plants, indicating their potential to mitigate biotic and abiotic stresses. The comprehensive genome-wide exploration provides opportunities for the development of cold-tolerant and virus-resistant cucumber variants by traditional breeding or gene.
Collapse
Affiliation(s)
- Samia Anwar
- Department of Botany, Lahore College for Women University, P.O BOX. 54000, Lahore, Pakistan
| | - Riffat Siddique
- Department of Botany, Lahore College for Women University, P.O BOX. 54000, Lahore, Pakistan
| | - Shakeel Ahmad
- Seed Center and Plant Genetic Resources Bank, Ministry of Environment, Water and Agriculture, Riyadh, 14712, Kingdom of Saudi Arabia
| | - Muhammad Zeshan Haider
- Department of Plant Breeding and Genetics, Faculty of Agricultural Sciences, University of the Punjab, P.O BOX. 54590, Lahore, Pakistan
| | - Haider Ali
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Adnan Sami
- Department of Plant Breeding and Genetics, Faculty of Agricultural Sciences, University of the Punjab, P.O BOX. 54590, Lahore, Pakistan
| | - Rosa Sanchez Lucas
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Muhammad Shafiq
- Department of Horticulture, Faculty of Agricultural Sciences, University of the Punjab, P.O BOX. 54590, Lahore, Pakistan.
| | - Bader Un Nisa
- Department of Botany, Lahore College for Women University, P.O BOX. 54000, Lahore, Pakistan
| | - Bilal Javed
- Department of Horticulture, Faculty of Agricultural Sciences, University of the Punjab, P.O BOX. 54590, Lahore, Pakistan
| | - Jannat Akram
- Department of Botany, Lahore College for Women University, P.O BOX. 54000, Lahore, Pakistan
| | - Javaria Tabassum
- Department of Plant Breeding and Genetics, Faculty of Agricultural Sciences, University of the Punjab, P.O BOX. 54590, Lahore, Pakistan.
| | - Muhammad Arshad Javed
- Department of Plant Breeding and Genetics, Faculty of Agricultural Sciences, University of the Punjab, P.O BOX. 54590, Lahore, Pakistan.
| |
Collapse
|
19
|
Li N, Hao L, Li S, Deng J, Yu F, Zhang J, Nie A, Hu X. The NRF-2/HO-1 Signaling Pathway: A Promising Therapeutic Target for Metabolic Dysfunction-Associated Steatotic Liver Disease. J Inflamm Res 2024; 17:8061-8083. [PMID: 39512865 PMCID: PMC11542495 DOI: 10.2147/jir.s490418] [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: 08/06/2024] [Accepted: 10/18/2024] [Indexed: 11/15/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a progressive liver disorder with a rising prevalence. It begins with lipid accumulation in hepatocytes and gradually progresses to Metabolic-associated steatohepatitis (MASH), fibrosis, cirrhosis, and potentially hepatocellular carcinoma (HCC). The pathophysiology of MASLD is complex and involves multiple factors, with oxidative stress playing a crucial role. Oxidative stress drives the progression of MASLD by causing cellular damage, inflammatory responses, and fibrosis, making it a key pathogenic mechanism. The Nuclear Factor Erythroid 2-Related Factor 2 / Heme Oxygenase-1 (Nrf2/HO-1) signaling axis provides robust multi-organ protection against a spectrum of endogenous and exogenous insults, particularly oxidative stress. It plays a pivotal role in mediating antioxidant, anti-inflammatory, and anti-apoptotic responses. Many studies indicate that activating the Nrf2/HO-1 signaling pathway can significantly mitigate the progression of MASLD. This article examines the role of the Nrf2/HO-1 signaling pathway in MASLD and highlights natural compounds that protect against MASLD by targeting Nrf2/HO-1 activation. The findings indicate that the Nrf2/HO-1 signaling pathway holds great promise as a therapeutic target for MASLD.
Collapse
Affiliation(s)
- Na Li
- Department of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| | - Liyuan Hao
- Department of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| | - Shenghao Li
- Department of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| | - Jiali Deng
- Department of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| | - Fei Yu
- Department of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| | - Junli Zhang
- Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing, People's Republic of China
| | - Aiyu Nie
- Department of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| | - Xiaoyu Hu
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| |
Collapse
|
20
|
Tan W, Zhang J, Chen L, Wang Y, Chen R, Zhang H, Liang F. Copper homeostasis and cuproptosis-related genes: Therapeutic perspectives in non-alcoholic fatty liver disease. Diabetes Obes Metab 2024; 26:4830-4845. [PMID: 39233500 DOI: 10.1111/dom.15846] [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: 07/06/2024] [Revised: 07/16/2024] [Accepted: 07/17/2024] [Indexed: 09/06/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD), a metabolic-associated fatty liver disease, has become the most common chronic liver disease worldwide. Recently, the discovery of cuproptosis, a newly identified mode of cell death, further highlighted the importance of copper in maintaining metabolic homeostasis. An increasing number of studies have confirmed that liver copper metabolism is closely related to the pathogenesis of NAFLD. However, the relationship between NAFLD and copper metabolism, especially cuproptosis, remains unclear. In this review, we aim to summarize the current understanding of copper metabolism and its dysregulation, particularly the role of copper metabolism dysregulation in the pathogenesis of NAFLD. More importantly, this review emphasizes potential gene-targeted therapeutic strategies, challenges and the future of cuproptosis-related genes in the treatment of NAFLD. This review aims to provide innovative therapeutic strategies for NAFLD.
Collapse
Affiliation(s)
- Wangjing Tan
- Department of Acupuncture and Moxibustion, College of Acupuncture-Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
| | - Junli Zhang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Chen
- Department of Acupuncture and Moxibustion, College of Acupuncture-Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
- Hubei Provincial Collaborative Innovation Center of Preventive Treatment by Acupuncture and Moxibustion, Wuhan, China
- Hubei Shizhen Laboratory, Wuhan, China
| | - Yayuan Wang
- Department of Acupuncture and Moxibustion, College of Acupuncture-Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
- Hubei Provincial Collaborative Innovation Center of Preventive Treatment by Acupuncture and Moxibustion, Wuhan, China
- Hubei Shizhen Laboratory, Wuhan, China
| | - Rui Chen
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haiming Zhang
- Department of Oncology, Integrated Traditional Chinese and Western Medicine, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fengxia Liang
- Department of Acupuncture and Moxibustion, College of Acupuncture-Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
- Hubei Provincial Collaborative Innovation Center of Preventive Treatment by Acupuncture and Moxibustion, Wuhan, China
- Hubei Shizhen Laboratory, Wuhan, China
- Acupuncture and Moxibustion Department, Affiliated Hospital of Hubei University of Chinese Medicine(Hubei Provincial Hospital of Traditional Chinese Medicine), Wuhan, China
| |
Collapse
|
21
|
Noh HR, Sui G, Lee JW, Wang F, Park JS, Ma Y, Ma H, Jeong JW, Shin DS, Wu X, Hwang BY, Roh YS. Jolkinolide B Ameliorates Liver Inflammation and Lipogenesis by Regulating JAK/STAT3 Pathway. Biomol Ther (Seoul) 2024; 32:793-800. [PMID: 39370730 PMCID: PMC11535294 DOI: 10.4062/biomolther.2024.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 06/19/2024] [Accepted: 07/02/2024] [Indexed: 10/08/2024] Open
Abstract
Hepatic dysregulation of lipid metabolism exacerbates inflammation and enhances the progression of metabolic dysfunction-associated steatotic liver disease (MASLD). STAT3 has been linked to lipid metabolism and inflammation. Jolkinolide B (JB), derived from Euphorbia fischeriana, is known for its pharmacological anti-inflammatory and anti-tumor properties. Therefore, this study investigated whether JB affects MASLD prevention by regulating STAT3 signaling. JB attenuated steatosis and inflammatory responses in palmitic acid (PA)-treated hepatocytes. Additionally, JB treatment reduced the mRNA expression of de-novo lipogenic genes, such as acetyl-CoA carboxylase and stearoyl-CoA desaturase 1. Interestingly, JB-mediated reduction in inflammation and lipogenesis was dependent on STAT3 signaling. JB consistently modulated mitochondrial dysfunction and the mRNA expression of inflammatory cytokines by inhibiting PA-induced JAK/STAT3 activation. This study suggests that JB is a potential therapeutic agent to prevent major stages of MASLD through inhibition of JAK/STAT3 signaling in hepatocytes.
Collapse
Affiliation(s)
- Hye-Rin Noh
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - Guoyan Sui
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - Jin Woo Lee
- College of Pharmacy, Duksung Women’s University, Seoul 01369, Republic of Korea
| | - Feng Wang
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - Jeong-Su Park
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - Yuanqiang Ma
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - Hwan Ma
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - Ji-Won Jeong
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - Dong-Su Shin
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - Xuefeng Wu
- Shanghai Jiao Tong University School of Medicine, Shanghai Institute of Immunology, Department of Immunology and Microbiology, Hongqiao International Institute of Medicine, Shanghai Ton-gren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Bang-Yeon Hwang
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju 28160, Republic of Korea
| | - Yoon Seok Roh
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju 28160, Republic of Korea
| |
Collapse
|
22
|
Zhao W, Wang X, Nie W, Jiang M, Zhao Y, Zhang T, Ding Y. Zhimu-Huangbai herb-pair ameliorates hepatic steatosis in mice by regulating IRE1α/XBP1s pathway to inhibit SREBP-1c. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 134:156017. [PMID: 39265443 DOI: 10.1016/j.phymed.2024.156017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 05/14/2024] [Accepted: 08/31/2024] [Indexed: 09/14/2024]
Abstract
BACKGROUND Currently, there is a lack of validated pharmacological interventions for non-alcoholic fatty liver disease (NAFLD), which is characterized by the accumulation of hepatic triglyceride. Zhimu-Huangbai (ZH) herb-pair is a traditional Chinese medicine that regulates glucose and lipid metabolism disorders. However, the precise mechanisms underlying the preventive effects of hepatic triglyceride induced by high-fat diet (HFD) remain elusive. PURPOSE The study aimed to examine the impact of ZH herb-pair on NAFLD in mice and explore the underlying mechanisms, particularly its effects on endoplasmic reticulum (ER) stress and lipid metabolism. METHODS NAFLD was induced in mice using HFD, and the treated mice were orally administered ZH, metformin (Glucophage) or lovastatin. The lipid metabolism factors, ER stress markers, and the unfolded protein response (UPR) branch factors were measured using immunohistochemistry, western blotting or qRT-PCR. Co-Immunoprecipitation (CoIP) was performed to reveal the connection between SCAP and SREBP-1c. Tunicamycin (TM) and plasmid delivery were used to induce acute ER stress or crease XBP1 gain function models. The main compounds in ZH binding to IRE1α protein were studied by molecular docking and cellular thermal shift assay (CETSA). RESULTS Treatment with ZH significantly ameliorated hepatic steatosis and reduced lipid synthesis process mainly inhibiting the expression of mature active form of SREBP-1c through relieving ER stress. The expression of IRE1α and XBP1s was inhibited after treatment with ZH. In addition, ZH improved the fatty liver phenotype caused by XBP1 overexpression via decreasing srebp1c transcription. In vitro experimental results suggested that the main compounds in ZH decreased cellular TG contents. Mechanistically, ZH targeted IRE1α and inhibited XBP1s mRNA expression to relieve ER stress and inhibit SREBP-1c production. CONCLUSIONS ZH herb-pair can protect against NAFLD by reducing the expression of SREBP-1c, in part, via regulating IRE1α/XBP1s pathway.
Collapse
Affiliation(s)
- Wenjun Zhao
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, PR China
| | - Xiaoying Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, PR China
| | - Wenlong Nie
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, PR China
| | - Min Jiang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, PR China
| | - Yuan Zhao
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, PR China
| | - Tong Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, PR China.
| | - Yue Ding
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, PR China; National Innovation Platform for Medical Industry-education Integration, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, PR China.
| |
Collapse
|
23
|
Ma H, Sui GY, Park JS, Wang F, Ma Y, Shin DS, Rustamov N, Jang JS, Chang SI, Lee J, Roh YS. Blockade of 11β-hydroxysteroid dehydrogenase type 1 ameliorates metabolic dysfunction-associated steatotic liver disease and fibrosis. Heliyon 2024; 10:e39534. [PMID: 39498052 PMCID: PMC11534184 DOI: 10.1016/j.heliyon.2024.e39534] [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: 06/01/2024] [Revised: 10/16/2024] [Accepted: 10/16/2024] [Indexed: 11/07/2024] Open
Abstract
11β-Hydroxysteroid dehydrogenase type 1 (11β-HSD1) is a key enzyme involved in the conversion of cortisone to active cortisol in the liver. Elevated cortisol levels can trigger oxidative stress, inflammation, and hepatocyte damage, highlighting the importance of 11β-HSD1 inhibition as a potential therapeutic approach. This study aimed to explore the effects of INU-101, an inhibitor of 11β-HSD1, on the development of metabolic dysfunction-associated steatotic liver disease (MASLD) and fibrosis. Our findings demonstrated that INU-101 effectively mitigated cortisol-induced lipid accumulation, reactive oxygen species generation, and hepatocyte apoptosis. Furthermore, 11β-HSD1 inhibition suppressed hepatic stellate cell activation by modulating β-catenin and phosphorylated SMAD2/3. INU-101 administration significantly reduced hepatic lipid accumulation and liver fibrosis in mice fed fast-food diet. This study suggests that INU-101 holds promise as a clinical candidate for treating MASLD and fibrosis, offering potential therapeutic benefits by targeting the intricate processes involving 11β-HSD1 and cortisol regulation in the liver.
Collapse
Affiliation(s)
- Hwan Ma
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, 28160, South Korea
| | - Guo-Yan Sui
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, 28160, South Korea
| | - Jeong-Su Park
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, 28160, South Korea
| | - Feng Wang
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, 28160, South Korea
| | - Yuanqiang Ma
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, 28160, South Korea
| | - Dong-Su Shin
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, 28160, South Korea
| | - Nodir Rustamov
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, 28160, South Korea
| | | | | | - Jin Lee
- Department of Pathology, School of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Yoon Seok Roh
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, 28160, South Korea
| |
Collapse
|
24
|
Li Y, Kong H, Li C, Gu Z, Ban X, Li Z. Cooperative action of non-digestible oligosaccharides improves lipid metabolism of high-fat diet-induced mice. Food Funct 2024; 15:10434-10446. [PMID: 39324226 DOI: 10.1039/d4fo03183k] [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: 09/27/2024]
Abstract
Non-digestible oligosaccharides are known to exert health-promoting effects. However, the specific mechanisms by which they regulate host physiology remain unclear. Understanding these mechanisms will facilitate the development of non-digestible oligosaccharide compositions that can achieve synergistic effects. This study selected three representative non-digestible oligosaccharides, namely xylo-oligosaccharides (XOS), galacto-oligosaccharides (GOS), and isomalto-oligosaccharides (IMO), to investigate their effects as dietary interventions on mice fed a high-fat diet. The results demonstrated that XOS and IMO synergistically mitigated weight gain and ectopic lipid deposition. Further analysis revealed that XOS significantly altered the composition of the gut microbiota, while IMO significantly enhanced insulin sensitivity via the PI3K/Akt pathway. Moreover, the combination of XOS and IMO synergistically promoted the oxidation and breakdown of fatty acids and increased the abundance of acetate and propionate-producing bacteria, such as Lactobacillus. These findings suggest a novel strategy for obesity management based on dietary intervention with XOS and IMO.
Collapse
Affiliation(s)
- Yiwen Li
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China.
- Institute of Future Food Technology, JITRI, Yixing 214200, China
| | - Haocun Kong
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China.
- Institute of Future Food Technology, JITRI, Yixing 214200, China
| | - Caiming Li
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China.
- Institute of Future Food Technology, JITRI, Yixing 214200, China
| | - Zhengbiao Gu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China.
| | - Xiaofeng Ban
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China.
| | - Zhaofeng Li
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China.
- Institute of Future Food Technology, JITRI, Yixing 214200, China
| |
Collapse
|
25
|
Brito ML, Coutinho-Wolino KS, Almeida PP, Trigueira PDC, Alves APDP, Magliano DC, Stockler-Pinto MB. Unstressing the Reticulum: Nutritional Strategies for Modulating Endoplasmic Reticulum Stress in Obesity. Mol Nutr Food Res 2024; 68:e2400361. [PMID: 39363792 DOI: 10.1002/mnfr.202400361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 09/03/2024] [Indexed: 10/05/2024]
Abstract
The progression of obesity involves several molecular mechanisms that are closely associated with the pathophysiological response of the disease. Endoplasmic reticulum (ER) stress is one such factor. Lipotoxicity disrupts endoplasmic reticulum homeostasis in the context of obesity. Furthermore, it induces ER stress by activating several signaling pathways via inflammatory responses and oxidative stress. ER performs crucial functions in protein synthesis and lipid metabolism; thus, triggers such as lipotoxicity can promote the accumulation of misfolded proteins in the organelle. The accumulation of these proteins can lead to metabolic disorders and chronic inflammation, resulting in cell death. Thus, alternatives, such as flavonoids, amino acids, and polyphenols that are associated with antioxidant and anti-inflammatory responses have been proposed to attenuate this response by modulating ER stress via the administration of nutrients and bioactive compounds. Decreasing inflammation and oxidative stress can reduce the expression of several ER stress markers and improve clinical outcomes through the management of obesity, including the control of body weight, visceral fat, and lipid accumulation. This review explores the metabolic changes resulting from ER stress and discusses the role of nutritional interventions in modulating the ER stress pathway in obesity.
Collapse
Affiliation(s)
- Michele Lima Brito
- Pathology Post Graduate Program, Fluminense Federal University (UFF), Niterói, RJ, 24070-090, Brazil
| | - Karen Salve Coutinho-Wolino
- Cardiovascular Sciences Post Graduate Program, Fluminense Federal University (UFF), Niterói, RJ, 24070-090, Brazil
| | - Patricia Pereira Almeida
- Pathology Post Graduate Program, Fluminense Federal University (UFF), Niterói, RJ, 24070-090, Brazil
| | | | - Ana Paula de Paula Alves
- Endocrinology Post Graduate Program, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, 24210-201, Brazil
| | - D'Angelo Carlo Magliano
- Pathology Post Graduate Program, Fluminense Federal University (UFF), Niterói, RJ, 24070-090, Brazil
- Cardiovascular Sciences Post Graduate Program, Fluminense Federal University (UFF), Niterói, RJ, 24070-090, Brazil
- Endocrinology Post Graduate Program, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, 24210-201, Brazil
- Morphology Department, Biomedical Institute, Fluminense Federal University (UFF), Niterói, RJ, 24020-150, Brazil
| | - Milena Barcza Stockler-Pinto
- Pathology Post Graduate Program, Fluminense Federal University (UFF), Niterói, RJ, 24070-090, Brazil
- Cardiovascular Sciences Post Graduate Program, Fluminense Federal University (UFF), Niterói, RJ, 24070-090, Brazil
- Nutrition Sciences Postgraduate Program, Fluminense Federal University (UFF), Niterói, RJ, 24020-140, Brazil
| |
Collapse
|
26
|
Hu B, Sui J, Wang Y, Li L, Gong D, Zhu Z, Liao W, Sun G, Xia H. A systematic review of dietary and circulating carotenoids and liver disease. Food Funct 2024; 15:9813-9832. [PMID: 39229651 DOI: 10.1039/d4fo03082f] [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: 09/05/2024]
Abstract
Background: due to the high incidence of liver disease and the severity of adverse outcomes, liver disease has become a serious public health problem, bringing a huge disease burden to individuals, families, and society. Most studies have shown significant differences in serum carotenoid content and dietary carotenoid intake between liver disease patients and non-liver disease patients, but some studies have reported contrary results. This paper aimed to systematically review and analyze all published epidemiological studies on carotenoids and liver disease to quantitatively assess the relationship between serum and dietary carotenoid concentrations and liver disease. Methods: by systematically searching PubMed, Web of Science, Scopus, Embase, and Cochrane databases according to pre-combined search terms from inception to July 23, 2024, 30 studies were found to meet the exclusion criteria. Finally, 3 RCT studies, 6 cohort studies, 11 case-control studies, 9 cross-sectional studies, and 1 RCT-combined cross-sectional study were included in the further analysis. Two reviewers independently scored the literature quality and extracted data, and the results were represented by the standard mean difference (SMD) with a 95% confidence interval. Cochran Q statistics and I2 statistics were used to evaluate statistical heterogeneity (defined as significant when P < 0.05 or I2 > 50%). When there was insignificant heterogeneity, a fixed effects model was selected; otherwise a random effects model was used. Publication bias was assessed by the Egger test. Results: pooled meta-analysis showed that serum α-carotene (SMD = -0.58, 95% CI (-0.83, -0.32), P < 0.001), β-carotene (SMD = -0.81, 95% CI (-1.13, -0.49), P < 0.001), and lycopene (SMD = -1.06, 95% CI (-1.74, -0.38), P < 0.001) were negatively correlated with the risk and severity of liver disease. However, no significant difference was observed between serum β-cryptoxanthin (SMD = 0.02, 95% CI (-0.41, 0.45), P = 0.92) and lutein/zeaxanthin (SMD = 0.62, 95% CI (-1.20, 2.45), P = 0.502). Dietary β-carotene intake (SMD = -0.22, 95% CI (-0.31, -0.13), P < 0.001) was negatively associated with the risk of liver disease. The Egger test showed no publication bias (P > 0.05). An intake of more than 6 mg of carotenoids on an energy-restricted diet can effectively alleviate the symptoms of NAFLD. Conclusion: lower serum concentrations of α-carotene, β-carotene, and lycopene were associated with a higher risk of liver disease. Meanwhile, dietary intake of β-carotene could reduce the incidence of liver disease. However, for malignant diseases such as liver cancer, it did not show the significant effects of carotenoid supplementation.
Collapse
Affiliation(s)
- Bihuan Hu
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing 210009, PR China.
| | - Jing Sui
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing 210009, PR China.
- Research Institute for Environment and Health, Nanjing University of Information Science and Technology, Nanjing, Jiangsu, 210044, China
| | - Ying Wang
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing 210009, PR China.
| | - Lihua Li
- Lianshui People's Hospital Affiliated to Kangda College of Nanjing Medical University, Huai'an, Jiangsu, 223400, China
| | - Daochen Gong
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing 210009, PR China.
| | - Zixuan Zhu
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing 210009, PR China.
| | - Wang Liao
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing 210009, PR China.
| | - Guiju Sun
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing 210009, PR China.
| | - Hui Xia
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing 210009, PR China.
| |
Collapse
|
27
|
Pinton G, Perucca M, Gigliotti V, Mantovani E, Clemente N, Malecka J, Chrostek G, Dematteis G, Lim D, Moro L, Chiazza F. EZH2-Mediated H3K27 Trimethylation in the Liver of Mice Is an Early Epigenetic Event Induced by High-Fat Diet Exposure. Nutrients 2024; 16:3260. [PMID: 39408226 PMCID: PMC11479199 DOI: 10.3390/nu16193260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/17/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND/OBJECTIVES Methyltransferase EZH2-mediated H3K27me3 is involved in liver inflammation and fibrosis, but its role in hepatic metabolic derangements is not yet clearly defined. We investigated if a high-fat diet (HFD) induced early changes in EZH2 expression and H3K27 me3 in the liver of mice. METHODS Five-week-old mice were fed an HFD or a low-fat diet (Control) for 2 weeks (2 W) or 8 weeks (8 W). Body weight was recorded weekly. Glycemia and oral glucose tolerance were assessed at baseline and after 2 W-8 W. Finally, livers were collected for further analysis. RESULTS As expected, mice that received 8 W HFD showed an increase in body weight, glycemia, and liver steatosis and an impairment in glucose tolerance; no alterations were observed in 2 W HFD mice. Eight weeks of HFD caused hepatic EZH2 nuclear localization and increased H3 K27me3; surprisingly, the same alterations occurred in 2 W HFD mice livers, even before overweight onset. We demonstrated that selective EZH2 inhibition reduced H3K27me3 and counteracted lipid accumulation in HUH-7 cells upon palmitic acid treatment. CONCLUSIONS In conclusion, we point to EZH2/H3K27me3 as an early epigenetic event occurring in fatty-acid-challenged livers both in vivo and in vitro, thus establishing EZH2 as a potential pharmacological target for metabolic derangements.
Collapse
Affiliation(s)
- Giulia Pinton
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale (UPO), Largo Donegani 2, 28100 Novara, Italy; (G.P.); (V.G.); (G.C.); (G.D.)
| | - Mattia Perucca
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale (UPO), Largo Donegani 2, 28100 Novara, Italy; (G.P.); (V.G.); (G.C.); (G.D.)
| | - Valentina Gigliotti
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale (UPO), Largo Donegani 2, 28100 Novara, Italy; (G.P.); (V.G.); (G.C.); (G.D.)
| | - Elena Mantovani
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale (UPO), Largo Donegani 2, 28100 Novara, Italy; (G.P.); (V.G.); (G.C.); (G.D.)
| | - Nausicaa Clemente
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), Università del Piemonte Orientale (UPO), Via Solaroli 17, 28100 Novara, Italy
| | - Justyna Malecka
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale (UPO), Largo Donegani 2, 28100 Novara, Italy; (G.P.); (V.G.); (G.C.); (G.D.)
| | - Gabriela Chrostek
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale (UPO), Largo Donegani 2, 28100 Novara, Italy; (G.P.); (V.G.); (G.C.); (G.D.)
| | - Giulia Dematteis
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale (UPO), Largo Donegani 2, 28100 Novara, Italy; (G.P.); (V.G.); (G.C.); (G.D.)
| | - Dmitry Lim
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale (UPO), Largo Donegani 2, 28100 Novara, Italy; (G.P.); (V.G.); (G.C.); (G.D.)
| | - Laura Moro
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale (UPO), Largo Donegani 2, 28100 Novara, Italy; (G.P.); (V.G.); (G.C.); (G.D.)
| | - Fausto Chiazza
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale (UPO), Largo Donegani 2, 28100 Novara, Italy; (G.P.); (V.G.); (G.C.); (G.D.)
| |
Collapse
|
28
|
Kineman RD, Del Rio-Moreno M, Waxman DJ. Liver-specific actions of GH and IGF1 that protect against MASLD. Nat Rev Endocrinol 2024:10.1038/s41574-024-01037-0. [PMID: 39322791 DOI: 10.1038/s41574-024-01037-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/29/2024] [Indexed: 09/27/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD; also known as nonalcoholic fatty liver disease) is a chronic condition associated with metabolic syndrome, a group of conditions that includes obesity, insulin resistance, hyperlipidaemia and cardiovascular disease. Primary growth hormone (GH) deficiency is associated with MASLD, and the decline in circulating levels of GH with weight gain might contribute to the development of MASLD. Raising endogenous GH secretion or administering GH replacement therapy in the context of MASLD enhances insulin-like growth factor 1 (IGF1) production and reduces steatosis and the severity of liver injury. GH and IGF1 indirectly control MASLD progression by regulating systemic metabolic function. Evidence supports the proposal that GH and IGF1 also have a direct role in regulating liver metabolism and health. This Review focuses on how GH acts on the hepatocyte in a sex-dependent manner to limit lipid accumulation, reduce stress, and promote survival and regeneration. In addition, we discuss how GH and IGF1 might regulate non-parenchymal cells of the liver to control inflammation and fibrosis, which have a major effect on hepatocyte survival and regeneration. Development of a better understanding of how GH and IGF1 coordinate the functions of specific, individual liver cell types might provide insight into the aetiology of MASLD initiation and progression and suggest novel approaches for the treatment of MASLD.
Collapse
Affiliation(s)
- Rhonda D Kineman
- Department of Medicine, Section of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago, Chicago, IL, USA.
- Jesse Brown VA Medical Center, Research and Development Division, Chicago, IL, USA.
| | - Mercedes Del Rio-Moreno
- Department of Medicine, Section of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago, Chicago, IL, USA
- Jesse Brown VA Medical Center, Research and Development Division, Chicago, IL, USA
| | - David J Waxman
- Department of Biology and Bioinformatics Program, Boston University, Boston, MA, USA
| |
Collapse
|
29
|
Zi Y, Qin Y, Ma C, Qiao Y, Xu X, Yang Y, He Q, Li M, Liu Y, Gao F. Transcriptome analysis reveals hepatic disordered lipid metabolism, lipotoxic injury, and abnormal development in IUGR sheep fetuses due to maternal undernutrition during late pregnancy. Theriogenology 2024; 226:350-362. [PMID: 38968678 DOI: 10.1016/j.theriogenology.2024.06.020] [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: 10/13/2023] [Revised: 06/19/2024] [Accepted: 06/23/2024] [Indexed: 07/07/2024]
Abstract
Although lipid metabolism in fetal livers under intrauterine growth restriction (IUGR) conditions has been widely studied, the implications of maternal undernutrition on fetal hepatic lipid metabolism, lipotoxic injury, and abnormal development remain largely unknown. Therefore, this study investigated the effects of maternal undernutrition on disordered hepatic lipid metabolism, lipotoxic injury, and abnormal development in IUGR sheep fetuses using transcriptome analysis. Seventeen singleton ewes were randomly divided into three groups on day 90 of pregnancy: a control group (CG; 0.63 MJ metabolic energy/body weight (ME/BW)0.75/day, n = 5), maternal undernutrition group 1 (MU1; 0.33 MJ ME/BW0.75/day, n = 6), and maternal undernutrition group 2 (MU2; 0.20 MJ ME/BW0.75/day, n = 6). The fetuses were euthanized and recovered on day 130 of pregnancy. The levels of free fatty acids (FFA) in maternal blood (P < 0.01), fetal blood (P < 0.01), and fetal livers (P < 0.05) were increased in the MU1 and MU2 groups, but fetal hepatic triglyceride (TG) levels in the MU2 group (P < 0.01) and β-hydroxybutyrate levels in the MU1 and MU2 groups (P < 0.01) were decreased compared to the CG. Severe inflammatory cell infiltration and increased non-alcoholic fatty liver disease activity scores were observed in MU1 and MU2 fetuses (P < 0.01). Progressive deposition of fetal hepatic reticular fibers and collagen fibers in the fetal livers of the MU1 and MU2 groups and significant hepatic fibrosis were observed in the MU2 fetuses (P < 0.05). Gene set enrichment analysis showed that genes involved in lipid accumulation and FFA beta oxidation were downregulated in both MU groups compared to those in the controls. The fetal liver mRNA expression of the β-oxidation regulator, acetyl-CoA acetyltransferase 1, and the TCA regulator, isocitrate dehydrogenase were reduced in MU1 (P < 0.05) and MU2 (P < 0.01) fetuses, and downregulated mRNA expression of long chain fatty acid CoA ligase 1 (P < 0.05) and glycerol-3-phosphate acyltransferase (P < 0.01) was observed in MU2 fetuses. Differentially expressed genes (DEGs) in MU1 versus CG (360 DEGs) and MU2 versus CG (746 DEGs) were identified using RNA sequencing. Bioinformatics analyses of the 231 intersecting DEGs between MU1 versus CG and MU2 versus CG indicated that neutrophil extracellular traps (NETs) were induced and played a central role in fetal hepatic injury in IUGR sheep. Increased maternal blood myeloperoxidase (MPO) levels (P < 0.01), NE (Elane)-positive areas in fetal liver sections (P < 0.05), and fetal liver MPO protein expression (P < 0.01) were found in the MU1 and MU2 groups; however, MPO levels were reduced in the fetal membrane (P < 0.01) and fetal blood (P < 0.05) in the MU1 group, and in the maternal-fetal placenta and fetal blood in the MU2 group (P < 0.01). Analysis of gene expression trends in the intersecting DEGs between MU1 versus CG (129 DEGs) and MU2 versus CG (515 DEGs) further revealed that 30 hub genes were essential regulators of the G2/M cell cycle, all of which were associated with hepatocellular carcinoma. G0/G1 phase cells of the fetal liver were reduced in the MU1 (P < 0.05) and MU2 (P < 0.01) groups, whereas G2/M phase cells were elevated in the MU1 and MU2 groups (P < 0.01). The representatives of upregulated hub genes and fetal liver protein expression of maternal embryonic leucine zipper kinase and protein regulator of cytokinesis 1 were progressively enhanced in the MU1 and MU2 groups (P < 0.01), and topoisomerase II alpha protein expression in the MU2 group (P < 0.05), as expected. These results indicate that FFA overload, severe lipotoxic injury, and NETs were induced, and disease-promoting regulators of the G2/M cell cycle were upregulated in the fetal liver of IUGR sheep. These findings provide new insights into the pathogenesis of impaired hepatic lipid metabolism and abnormal development and the molecular origin of post-natal liver disease in IUGR due to maternal undernutrition. This information can support the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Yang Zi
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, 010018, China; Shenzheng Institute of Advanced Technology, Chinese Academy of Sciences, Shenzheng, China
| | - Yulong Qin
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Chi Ma
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Yina Qiao
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Xiaoyi Xu
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Yilin Yang
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Qiuyue He
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Mingyue Li
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Yingchun Liu
- College of Life Science, Inner Mongolia Key Laboratory of Biomanufacturing, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Feng Gao
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, 010018, China.
| |
Collapse
|
30
|
Yan Y, Yuan H, Yang F, Na H, Yu X, Liu J, Wang Y. Seabuckthorn polysaccharides mitigate hepatic steatosis by modulating the Nrf-2/HO-1 pathway and gut microbiota. AMB Express 2024; 14:100. [PMID: 39251509 PMCID: PMC11383914 DOI: 10.1186/s13568-024-01756-7] [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: 05/25/2024] [Accepted: 08/14/2024] [Indexed: 09/11/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is becoming a significant global public health threat. Seabuckthorn (Hippophae rhamnoides L.) has been used in traditional Chinese medicine (TCM). The hypolipidemic effects of Seabuckthorn polysaccharides (SP) against high-fat diets (HFD)-induced NAFLD were systematically explored and compared with that of Bifidobacterium lactis V9 (B. Lactis V9). Results showed that HFD-induced alanine transaminase (ALT) and aspartate aminotransferase (AST) levels decreased by 2.8-fold and 4.5-fold, respectively, after SP supplementation. Moreover, the alleviating effect on hepatic lipid accumulation is better than that of B. Lactis V9. The ACC and FASN mRNA levels were significantly reduced by 1.8 fold (P < 0.05) and 2.3 folds (P < 0.05), respectively, while the CPT1α and PPARα mRNA levels was significantly increased by 2.3 fold (P < 0.05) and 1.6 fold (P < 0.05), respectively, after SP administration. SP activated phosphorylated-AMPK and inhibited PPARγ protein expression, improved serum oxidative stress and inflammation (P < 0.05). SP supplementation leads to increased hepatic expression of nuclear factor erythroid 2-related factor 2 (Nrf-2), heme oxygenase-1 (HO-1) and Superoxide dismutase-2 (SOD-2). Furthermore, SP treatment improved HFD-induced intestinal dysbiosis. Lentisphaerae, Firmicutes, Tenericutes and Peptococcus sp., RC9_gut_group sp., and Parabacteroides sp. of the gut microbiota were significantly associated with hepatic steatosis and indicators related to oxidative stress and inflammation. Therefore, SP can mitigate hepatic lipid accumulation by regulating Nrf-2/HO-1 signaling pathways and gut microbiota. This study offers new evidence supporting the use of SP as a prebiotic treatment for NAFLD.
Collapse
Affiliation(s)
- Yan Yan
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Haisheng Yuan
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Fan Yang
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Heiya Na
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Xiuling Yu
- Inner Mongolia Tianqi Biotechnology Co., Ltd, Chifeng, 024000, China
| | - Jingran Liu
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot, 010018, China.
| | - Yuzhen Wang
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot, 010018, China.
| |
Collapse
|
31
|
Tripathi M, Gauthier K, Sandireddy R, Zhou J, Guptta P, Sakthivel S, Teo WW, Naing YT, Arul K, Tikno K, Park SH, Wu Y, Wang L, Bay BH, Sun L, Giguere V, Chow PKH, Ghosh S, McDonnell DP, Yen PM, Singh BK. Esrra regulates Rplp1-mediated translation of lysosome proteins suppressed in metabolic dysfunction-associated steatohepatitis and reversed by alternate day fasting. Mol Metab 2024; 87:101997. [PMID: 39032642 PMCID: PMC11327444 DOI: 10.1016/j.molmet.2024.101997] [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: 05/15/2024] [Revised: 07/03/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024] Open
Abstract
OBJECTIVE Currently, little is known about the mechanism(s) regulating global and specific protein translation during metabolic dysfunction-associated steatohepatitis (MASH; previously known as non-alcoholic steatohepatitis, NASH). METHODS Unbiased label-free quantitative proteome, puromycin-labelling and polysome profiling were used to understand protein translation activity in vitro and in vivo. RESULTS We observed a global decrease in protein translation during lipotoxicity in human primary hepatocytes, mouse hepatic AML12 cells, and livers from a dietary mouse model of MASH. Interestingly, proteomic analysis showed that Rplp1, which regulates ribosome and translation pathways, was one of the most downregulated proteins. Moreover, decreased Esrra expression and binding to the Rplp1 promoter, diminished Rplp1 gene expression during lipotoxicity. This, in turn, reduced global protein translation and Esrra/Rplp1-dependent translation of lysosome (Lamp2, Ctsd) and autophagy (sqstm1, Map1lc3b) proteins. Of note, Esrra did not increase its binding to these gene promoters or their gene transcription, confirming its regulation of their translation during lipotoxicity. Notably, hepatic Esrra-Rplp1-dependent translation of lysosomal and autophagy proteins also was impaired in MASH patients and liver-specific Esrra knockout mice. Remarkably, alternate day fasting induced Esrra-Rplp1-dependent expression of lysosomal proteins, restored autophagy, and reduced lipotoxicity, inflammation, and fibrosis in hepatic cell culture and in vivo models of MASH. CONCLUSIONS Esrra regulation of Rplp1-mediated translation of lysosome/autolysosome proteins was downregulated during MASH. Alternate day fasting activated this novel pathway and improved MASH, suggesting that Esrra and Rplp1 may serve as therapeutic targets for MASH. Our findings also provided the first example of a nuclear hormone receptor, Esrra, to not only regulate transcription but also protein translation, via induction of Rplp1.
Collapse
Affiliation(s)
- Madhulika Tripathi
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Karine Gauthier
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS, Ecole Normale Supérieure de Lyon, 46 Allée d'Italie 69364 Lyon Cedex 07, France
| | - Reddemma Sandireddy
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Jin Zhou
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Priyanka Guptta
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Suganya Sakthivel
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Wei Wen Teo
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Yadanar Than Naing
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Kabilesh Arul
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Keziah Tikno
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Sung-Hee Park
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, C238A Levine Science Research Center, Durham, NC 27710, USA
| | - Yajun Wu
- Department of Anatomy, Yong Loo Lin School of Medicine, NUS 117594, Singapore
| | - Lijin Wang
- Centre for Computational Biology, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore; Pennington Biomedical Research Center, Laboratory of Bioinformatics and Computational Biology, Baton Rouge, LA 70808, USA
| | - Boon-Huat Bay
- Department of Anatomy, Yong Loo Lin School of Medicine, NUS 117594, Singapore
| | - Lei Sun
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Vincent Giguere
- Goodman Cancer Research Centre, McGill University, 1160 Pine Avenue West, Montreal, Québec H3A 1A3, Canada
| | - Pierce K H Chow
- Dept of Surgery, Singapore General Hospital and Dept. of Surgical Oncology, National Cancer Centre 169608, Singapore
| | - Sujoy Ghosh
- Centre for Computational Biology, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore; Pennington Biomedical Research Center, Laboratory of Bioinformatics and Computational Biology, Baton Rouge, LA 70808, USA
| | - Donald P McDonnell
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, C238A Levine Science Research Center, Durham, NC 27710, USA
| | - Paul M Yen
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore; Duke Molecular Physiology Institute and Dept. of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Brijesh K Singh
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore.
| |
Collapse
|
32
|
Liu YN, Zhu HX, Li TY, Yang X, Li XJ, Zhang WK. Lipid nanoparticle encapsulated oleic acid induced lipotoxicity to hepatocytes via ROS overload and the DDIT3/BCL2/BAX/Caspases signaling in vitro and in vivo. Free Radic Biol Med 2024; 222:361-370. [PMID: 38945456 DOI: 10.1016/j.freeradbiomed.2024.06.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 06/24/2024] [Accepted: 06/28/2024] [Indexed: 07/02/2024]
Abstract
BACKGROUND To date, Non-alcoholic fatty liver disease (NAFLD) is one of the most common liver disease associated with clinical complications. Dietary fatty acids have been suggested to be involved in preventing or reversing the accumulation of hepatic fat. However, contradicting roles of monounsaturated fatty acids to the liver have been implicated in various human and murine models, mainly due to the insolubility nature of fatty acids. METHODS High pressure homogenization methods were used to fabricate oleic acid embedded lipid nanoparticles (OALNs). The in vitro and in vivo models were used to validate the physiological effect of this OALNs via various cellular and molecular approaches including cell viability essay, fluorescent staining, electron microscope, RNAseq, qPCR, Western blots, and IHC staining. RESULTS We successfully fabricated OALNs with enhanced stability and solubility. More importantly, lipid accumulation was successfully induced in hepatocytes via the application of OALNs in a dose-dependent manner. Overload of OALNs resulted in ROS accumulation and apoptosis of hepatocytes dose-dependently. With the help of transcriptome sequencing and traditional experimental approaches, we demonstrated that the lipotoxic effect induced by OALNs was exerted via the DDIT3/BCL2/BAX/Caspases signaling. Moreover, we also verified that OALNs induced steatosis and subsequent apoptosis in the liver of mice via the activation of DDIT3 in vivo. CONCLUSIONS In all, our results established a potential pathogenic model of NAFLD for further studies and indicated the possible involvement of DDIT3 signaling in abnormal steatosis process of the liver.
Collapse
Affiliation(s)
- Ya-Nan Liu
- School of Pharmaceutical Sciences, South-Central Minzu University, No 182 Minzu Avenue, Wuhan, 430064, China.
| | - Hong-Xia Zhu
- Guangzhou National Laboratory, No. 9 XingDaoHuanBei Road, Guangzhou International Bio Island, Guangzhou, 510005, Guangdong Province, China.
| | - Tao-Yu Li
- Guangzhou National Laboratory, No. 9 XingDaoHuanBei Road, Guangzhou International Bio Island, Guangzhou, 510005, Guangdong Province, China.
| | - Xinzhou Yang
- School of Pharmaceutical Sciences, South-Central Minzu University, No 182 Minzu Avenue, Wuhan, 430064, China.
| | - Xiao-Jun Li
- School of Pharmaceutical Sciences, South-Central Minzu University, No 182 Minzu Avenue, Wuhan, 430064, China.
| | - Wei Kevin Zhang
- Guangzhou National Laboratory, No. 9 XingDaoHuanBei Road, Guangzhou International Bio Island, Guangzhou, 510005, Guangdong Province, China.
| |
Collapse
|
33
|
Zhao X, Zhu Y, Yao Q, Zhao B, Lin G, Zhang M, Guo C, Li Y. Lipidomics Investigation Reveals the Reversibility of Hepatic Injury by Silica Nanoparticles in Rats After a 6-Week Recovery Duration. SMALL METHODS 2024; 8:e2301430. [PMID: 38191992 DOI: 10.1002/smtd.202301430] [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: 10/17/2023] [Revised: 12/19/2023] [Indexed: 01/10/2024]
Abstract
Given the inevitable human exposure owing to its increasing production and utilization, the comprehensive safety evaluation of silica nanoparticles (SiNPs) has sparked concerns. Substantial evidence indicated liver damage by inhaled SiNPs. Notwithstanding, few reports focused on the persistence or reversibility of hepatic injuries, and the intricate molecular mechanisms involved remain limited. Here, rats are intratracheally instilled with SiNPs in two regimens (a 3-month exposure and a subsequent 6-week recovery after terminating SiNPs administration) to assess the hepatic effects. Nontargeted lipidomics revealed alterations in lipid metabolites as a contributor to the hepatic response and recovery effects of SiNPs. In line with the functional analysis of differential lipid metabolites, SiNPs activated oxidative stress, and induced lipid peroxidation and lipid deposition in the liver, as evidenced by the elevated hepatic levels of ROS, MDA, TC, and TG. Of note, these indicators showed great improvements after a 6-week recovery, even returning to the control levels. According to the correlation, ROC curve, and SEM analysis, 11 lipids identified as potential regulatory molecules for ameliorating liver injury by SiNPs. Collectively, the work first revealed the reversibility of SiNP-elicited hepatotoxicity from the perspective of lipidomics and offered valuable laboratory evidence and therapeutic strategy to facilitate nanosafety.
Collapse
Affiliation(s)
- Xinying Zhao
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Yawen Zhu
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China
| | - Qing Yao
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Bosen Zhao
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Guimiao Lin
- School of Public Health, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518060, China
| | - Min Zhang
- Department of Nephrology, Affiliated Beijing Chaoyang Hospital of Capital Medical University, Beijing, 100020, China
| | - Caixia Guo
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China
| | - Yanbo Li
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| |
Collapse
|
34
|
L'homme L, Sermikli BP, Haas JT, Fleury S, Quemener S, Guinot V, Barreby E, Esser N, Caiazzo R, Verkindt H, Legendre B, Raverdy V, Cheval L, Paquot N, Piette J, Legrand-Poels S, Aouadi M, Pattou F, Staels B, Dombrowicz D. Adipose tissue macrophage infiltration and hepatocyte stress increase GDF-15 throughout development of obesity to MASH. Nat Commun 2024; 15:7173. [PMID: 39169003 PMCID: PMC11339436 DOI: 10.1038/s41467-024-51078-2] [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: 10/19/2023] [Accepted: 07/29/2024] [Indexed: 08/23/2024] Open
Abstract
Plasma growth differentiation factor-15 (GDF-15) levels increase with obesity and metabolic dysfunction-associated steatotic liver disease (MASLD) but the underlying mechanism remains poorly defined. Using male mouse models of obesity and MASLD, and biopsies from carefully-characterized patients regarding obesity, type 2 diabetes (T2D) and MASLD status, we identify adipose tissue (AT) as the key source of GDF-15 at onset of obesity and T2D, followed by liver during the progression towards metabolic dysfunction-associated steatohepatitis (MASH). Obesity and T2D increase GDF15 expression in AT through the accumulation of macrophages, which are the main immune cells expressing GDF15. Inactivation of Gdf15 in macrophages reduces plasma GDF-15 concentrations and exacerbates obesity in mice. During MASH development, Gdf15 expression additionally increases in hepatocytes through stress-induced TFEB and DDIT3 signaling. Together, these results demonstrate a dual contribution of AT and liver to GDF-15 production in metabolic diseases and identify potential therapeutic targets to raise endogenous GDF-15 levels.
Collapse
Affiliation(s)
- Laurent L'homme
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France.
| | - Benan Pelin Sermikli
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Joel T Haas
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Sébastien Fleury
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Sandrine Quemener
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Valentine Guinot
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Emelie Barreby
- Center for Infectious Medicine (CIM), Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Nathalie Esser
- Laboratory of Immunometabolism and Nutrition, GIGA-I3, University of Liège, Liège, Belgium
- Division of Diabetes, Nutrition and Metabolic Disorders, Department of Medicine, CHU Liège, Liège, Belgium
| | - Robert Caiazzo
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1190-EGID (Translational research in Diabetes), Lille, France
| | - Hélène Verkindt
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1190-EGID (Translational research in Diabetes), Lille, France
| | - Benjamin Legendre
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1190-EGID (Translational research in Diabetes), Lille, France
| | - Violeta Raverdy
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1190-EGID (Translational research in Diabetes), Lille, France
| | - Lydie Cheval
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, Laboratoire de Physiologie Rénale et Tubulopathies, Paris, France
- CNRS EMR 8228-Unité Métabolisme et Physiologie Rénale, Paris, France
| | - Nicolas Paquot
- Laboratory of Immunometabolism and Nutrition, GIGA-I3, University of Liège, Liège, Belgium
- Division of Diabetes, Nutrition and Metabolic Disorders, Department of Medicine, CHU Liège, Liège, Belgium
| | - Jacques Piette
- Laboratory of Virology and Immunology, GIGA-Signal Transduction, University of Liège, Liège, Belgium
| | - Sylvie Legrand-Poels
- Laboratory of Immunometabolism and Nutrition, GIGA-I3, University of Liège, Liège, Belgium
| | - Myriam Aouadi
- Center for Infectious Medicine (CIM), Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - François Pattou
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1190-EGID (Translational research in Diabetes), Lille, France
| | - Bart Staels
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - David Dombrowicz
- Univ. Lille, INSERM, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France.
| |
Collapse
|
35
|
Vu HT, Nguyen VD, Ikenaga H, Matsubara T. Application of PPAR Ligands and Nanoparticle Technology in Metabolic Steatohepatitis Treatment. Biomedicines 2024; 12:1876. [PMID: 39200340 PMCID: PMC11351628 DOI: 10.3390/biomedicines12081876] [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: 07/29/2024] [Revised: 08/10/2024] [Accepted: 08/13/2024] [Indexed: 09/02/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease/steatohepatitis (MASLD/MASH) is a major disease worldwide whose effective treatment is challenging. Peroxisome proliferator-activated receptors (PPARs) belong to the nuclear receptor superfamily and function as ligand-activated transcription factors. To date, three distinct subtypes of PPARs have been characterized: PPARα, PPARβ/δ, and PPARγ. PPARα and PPARγ are crucial regulators of lipid metabolism that modulate the transcription of genes involved in fatty acid (FA), bile acid, and cholesterol metabolism. Many PPAR agonists, including natural (FAs, eicosanoids, and phospholipids) and synthetic (fibrate, thiazolidinedione, glitazar, and elafibranor) agonists, have been developed. Furthermore, recent advancements in nanoparticles (NPs) have led to the development of new strategies for MASLD/MASH therapy. This review discusses the applications of specific cell-targeted NPs and highlights the potential of PPARα- and PPARγ-targeted NP drug delivery systems for MASLD/MASH treatment.
Collapse
Affiliation(s)
- Hung Thai Vu
- Department of Anatomy and Regenerative Biology, Graduate School of Medicine, Osaka Metropolitan University, Osaka 545-8585, Osaka, Japan; (H.T.V.); (V.D.N.)
| | - Vien Duc Nguyen
- Department of Anatomy and Regenerative Biology, Graduate School of Medicine, Osaka Metropolitan University, Osaka 545-8585, Osaka, Japan; (H.T.V.); (V.D.N.)
| | - Hiroko Ikenaga
- Department of Hepatology, Graduate School of Medicine, Osaka Metropolitan University, Osaka 545-8585, Osaka, Japan
| | - Tsutomu Matsubara
- Department of Anatomy and Regenerative Biology, Graduate School of Medicine, Osaka Metropolitan University, Osaka 545-8585, Osaka, Japan; (H.T.V.); (V.D.N.)
- Research Institute for Light-induced Acceleration System (RILACS), Osaka Metropolitan University, Sakai 599-8570, Osaka, Japan
| |
Collapse
|
36
|
Niu MY, Dong GT, Li Y, Luo Q, Cao L, Wang XM, Wang QW, Wang YT, Zhang Z, Zhong XW, Dai WB, Li LY. Fanlian Huazhuo Formula alleviates high-fat diet-induced non-alcoholic fatty liver disease by modulating autophagy and lipid synthesis signaling pathway. World J Gastroenterol 2024; 30:3584-3608. [PMID: 39193572 PMCID: PMC11346146 DOI: 10.3748/wjg.v30.i30.3584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/15/2024] [Accepted: 07/22/2024] [Indexed: 08/08/2024] Open
Abstract
BACKGROUND Fanlian Huazhuo Formula (FLHZF) has the functions of invigorating spleen and resolving phlegm, clearing heat and purging turbidity. It has been identified to have therapeutic effects on type 2 diabetes mellitus (T2DM) in clinical application. Non-alcoholic fatty liver disease (NAFLD) is frequently diagnosed in patients with T2DM. However, the therapeutic potential of FLHZF on NAFLD and the underlying mechanisms need further investigation. AIM To elucidate the effects of FLHZF on NAFLD and explore the underlying hepatoprotective mechanisms in vivo and in vitro. METHODS HepG2 cells were treated with free fatty acid for 24 hours to induce lipid accumulation cell model. Subsequently, experiments were conducted with the different concentrations of freeze-dried powder of FLHZF for 24 hours. C57BL/6 mice were fed a high-fat diet for 8-week to establish a mouse model of NAFLD, and then treated with the different concentrations of FLHZF for 10 weeks. RESULTS FLHZF had therapeutic potential against lipid accumulation and abnormal changes in biochemical indicators in vivo and in vitro. Further experiments verified that FLHZF alleviated abnormal lipid metabolism might by reducing oxidative stress, regulating the AMPKα/SREBP-1C signaling pathway, activating autophagy, and inhibiting hepatocyte apoptosis. CONCLUSION FLHZF alleviates abnormal lipid metabolism in NAFLD models by regulating reactive oxygen species, autophagy, apoptosis, and lipid synthesis signaling pathways, indicating its potential for clinical application in NAFLD.
Collapse
Affiliation(s)
- Meng-Yuan Niu
- Pharmacology Laboratory, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine, Zhongshan 528400, Guangdong Province, China
| | - Geng-Ting Dong
- Pharmacology Laboratory, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine, Zhongshan 528400, Guangdong Province, China
| | - Yi Li
- Pharmacology Laboratory, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine, Zhongshan 528400, Guangdong Province, China
| | - Qing Luo
- Pharmacology Laboratory, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine, Zhongshan 528400, Guangdong Province, China
| | - Liu Cao
- Pharmacology Laboratory, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine, Zhongshan 528400, Guangdong Province, China
| | - Xi-Min Wang
- Pharmacology Laboratory, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine, Zhongshan 528400, Guangdong Province, China
| | - Qi-Wen Wang
- Pharmacology Laboratory, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine, Zhongshan 528400, Guangdong Province, China
| | - Yi-Ting Wang
- Pharmacology Laboratory, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine, Zhongshan 528400, Guangdong Province, China
| | - Zhe Zhang
- Pharmacology Laboratory, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine, Zhongshan 528400, Guangdong Province, China
| | - Xi-Wen Zhong
- Pharmacology Laboratory, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine, Zhongshan 528400, Guangdong Province, China
| | - Wei-Bo Dai
- Pharmacology Laboratory, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine, Zhongshan 528400, Guangdong Province, China
| | - Le-Yu Li
- Department of Endocrinology, Zhongshan Hospital of Traditional Chinese Medicine Affiliated to Guangzhou University of Traditional Chinese Medicine, Zhongshan 528400, Guangdong Province, China
| |
Collapse
|
37
|
Walls KM, Joh JY, Hong KU, Hein DW. Heterocyclic Amines Disrupt Lipid Homeostasis in Cryopreserved Human Hepatocytes. Cardiovasc Toxicol 2024; 24:747-756. [PMID: 38851663 PMCID: PMC11300155 DOI: 10.1007/s12012-024-09874-1] [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: 02/23/2024] [Accepted: 05/23/2024] [Indexed: 06/10/2024]
Abstract
Metabolic dysfunction associated-steatotic liver disease (MASLD)/metabolic dysfunction-associated steatohepatitis (MASH) is the liver manifestation of metabolic syndrome, which is characterized by insulin resistance, hyperglycemia, hypertension, dyslipidemia, and/or obesity. Environmental pollutant exposure has been recently identified as a risk factor for developing MASH. Heterocyclic amines (HCAs) are mutagens generated when cooking meat at high temperatures or until well-done. Recent epidemiological studies reported that dietary HCA exposure may be linked to insulin resistance and type II diabetes, and we recently reported that HCAs induce insulin resistance and glucose production in human hepatocytes. However, no previous studies have examined the effects of HCAs on hepatic lipid homeostasis. In the present study, we assessed the effects of two common HCAs, MeIQx (2-amino-3, 8-dimethylimidazo [4, 5-f] quinoxaline) and PhIP (2-amino-1-methyl-6-phenylimidazo[4, 5-b] pyridine), on lipid homeostasis in cryopreserved human hepatocytes. Exposure to a single concentration of 25 μM MeIQx or PhIP in human hepatocytes led to dysregulation of lipid homeostasis, typified by significant increases in lipid droplets and triglycerides. PhIP significantly increased expression of lipid droplet-associated genes, PNPLA3 and HSD17B13, and both HCAs significantly increased PLIN2. Exposure to MeIQx or PhIP also significantly increased expression of several key genes involved in lipid synthesis, transport and metabolism, including FASN, DGAT2, CPT1A, SCD, and CD36. Furthermore, both MeIQx and PhIP significantly increased intracellular cholesterol and decreased expression of PON1 which is involved in cholesterol efflux. Taken together, these results suggest that HCAs dysregulate lipid production, metabolism, and storage. The current study demonstrates, for the first time, that HCA exposure may lead to fat accumulation in hepatocytes, which may contribute to hepatic insulin resistance and MASH.
Collapse
Affiliation(s)
- Kennedy M Walls
- Department of Pharmacology and Toxicology and Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY, 40202, USA
- Environmental Justice, Community Health and Environmental Review Division, US Environmental Protection Agency, Chicago, USA
| | - Jonathan Y Joh
- Department of Pharmacology and Toxicology and Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Kyung U Hong
- Department of Pharmacology and Toxicology and Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY, 40202, USA
- Department of Pharmaceutical and Administrative Sciences, Western New England University, Springfield, USA
| | - David W Hein
- Department of Pharmacology and Toxicology and Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY, 40202, USA.
| |
Collapse
|
38
|
Yu Z, Fan X, Zhao X, He T, Li X, Du H, Zhao M, Zhu R, Li M, Zhang Z, Han F. Polystyrene Nanoplastics Induce Lipid Metabolism Disorder by Activating the PERK-ATF4 Signaling Pathway in Mice. ACS APPLIED MATERIALS & INTERFACES 2024; 16:34524-34537. [PMID: 38926154 DOI: 10.1021/acsami.4c04416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/28/2024]
Abstract
In recent years, the study of microplastics (MPs) and nanoplastics (NPs) and their effects on human health has gained significant attention. The impacts of NPs on lipid metabolism and the specific mechanisms involved remain poorly understood. To address this, we utilized high-throughput sequencing and molecular biology techniques to investigate how endoplasmic reticulum (ER) stress might affect hepatic lipid metabolism in the presence of polystyrene nanoplastics (PS-NPs). Our findings suggest that PS-NPs activate the PERK-ATF4 signaling pathway, which in turn upregulates the expression of genes related to lipid synthesis via the ATF4-PPARγ/SREBP-1 pathway. This activation leads to an abnormal accumulation of lipid droplets in the liver. 4-PBA, a known ER stress inhibitor, was found to mitigate the PS-NPs-induced lipid metabolism disorder. These results demonstrate the hepatotoxic effects of PS-NPs and clarify the mechanisms of abnormal lipid metabolism induced by PS-NPs.
Collapse
Affiliation(s)
- Ziteng Yu
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Xingpei Fan
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Xinyi Zhao
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Tianyue He
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Xiaoyan Li
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Haining Du
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Meimei Zhao
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Ruijiao Zhu
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Mengcong Li
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Ziyi Zhang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Fang Han
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| |
Collapse
|
39
|
Wang D, Zhou BY, Xiang L, Chen XY, Feng JX. Alanine aminotransferase as a risk marker for new-onset metabolic dysfunction-associated fatty liver disease. World J Gastroenterol 2024; 30:3132-3139. [PMID: 39006380 PMCID: PMC11238669 DOI: 10.3748/wjg.v30.i25.3132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 05/07/2024] [Accepted: 06/13/2024] [Indexed: 07/01/2024] Open
Abstract
In this editorial, we comment on the article by Chen et al. Metabolic dysfunction-associated fatty liver disease (MAFLD) is a global public health burden whose incidence has risen concurrently with overweight and obesity. Given its detrimental health impact, early identification of at-risk individuals is crucial. MAFLD diagnosis is based on evidence of hepatic steatosis indicated by liver biopsy, imaging, or blood biomarkers, and one of the following conditions: Overweight/ obesity, type 2 diabetes mellitus, or metabolic dysregulation. However, in large-scale epidemiological studies, liver biopsies are not feasible. The application of techniques such as ultrasonography, computed tomography, magnetic resonance imaging, and magnetic resonance spectroscopy is restricted by their limited sensitivity, low effectiveness, high costs, and need for specialized software. Blood biomarkers offer several advantages, particularly in large-scale epidemiological studies or clinical scenarios where traditional imaging techniques are impractical. Analysis of cumulative effects of excess high-normal blood alanine aminotransferase (ALT) levels of blood ALT levels could facilitate identification of at-risk patients who might not be detected through conventional imaging methods. Accordingly, investigating the utility of blood biomarkers in MAFLD should enhance early detection and monitoring, enabling timely intervention and management and improving patient outcomes.
Collapse
Affiliation(s)
- Di Wang
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Bing-Yan Zhou
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Lei Xiang
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Xu-Yong Chen
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Jie-Xiong Feng
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| |
Collapse
|
40
|
Zhang C, Tong Q, Liu K, Mao T, Song Y, Qu Y, Chen X, Qiu Z. Morroniside delays the progression of non-alcoholic steatohepatitis by promoting AMPK-mediated lipophagy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 129:155703. [PMID: 38723527 DOI: 10.1016/j.phymed.2024.155703] [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: 01/24/2024] [Revised: 04/23/2024] [Accepted: 04/30/2024] [Indexed: 05/30/2024]
Abstract
BACKGROUND Non-alcoholic steatohepatitis (NASH), the inflammatory subtype in the progression of non-alcoholic fatty liver disease, is becoming a serious burden threatening human health, but no approved medication is available to date. Mononoside is a natural active substance derived from Cornus officinalis and has been confirmed to have great potential in regulating lipid metabolism in our previous studies. However, its effect and mechanism to inhibit the progression of NASH remains unclear. PURPOSE Our work aimed to explore the action of mononoside in delaying the progression of NASH and its regulatory mechanisms from the perspective of regulating lipophagy. METHODS AND RESULTS Male C57BL/6 mice were fed with a high-fat and high-fructose diet for 16 weeks to establish a NASH mouse model. After 8 weeks of high-fat and high-fructose feeding, these mice were administrated with different doses of morroniside. H&E staining, ORO staining, Masson staining, RNA-seq, immunoblotting, and immunofluorescence were performed to determine the effects and molecular mechanisms of morroniside in delaying the progression of NASH. In this study, we found that morroniside is effective in attenuating hepatic lipid metabolism disorders and inflammatory response activation, thereby limiting the progression from simple fatty liver to NASH in high-fat and high-fructose diet-fed mice. Mechanistically, we identified AMPK signaling as the key molecular pathway for the positive efficacy of morroniside by transcriptome sequencing. Our results revealed that morroniside maintained hepatic lipid metabolism homeostasis and inhibited NLRP3 inflammasome activation by promoting AMPKα phosphorylation-mediated lipophagy and fatty acid oxidation. Consistent results were observed in palmitic acid-treated cell models. Of particular note, silencing AMPKα both in vivo and in vitro reversed morroniside-induced lipophagy flux enhancement and NLRP3 inflammasome inhibition, emphasizing the critical role of AMPKα activation in the effect of morroniside in inhibiting NASH progression. CONCLUSION In summary, the present study provides strong evidence for the first time that morroniside inhibits NASH progression by promoting AMPK-dependent lipophagy and inhibiting NLRP3 inflammasome activation, suggesting that morroniside is expected to be a potential molecular entity for the development of therapeutic drugs for NASH.
Collapse
Affiliation(s)
- Cong Zhang
- College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, China.
| | - Qiao Tong
- Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou 310023, China
| | - Kexin Liu
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, Wuhan 430072, China
| | - Tongyun Mao
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Yingying Song
- Hubei Key Laboratory of Resources and Chemistry of Chinese Medicine, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Yaqin Qu
- Hubei Key Laboratory of Resources and Chemistry of Chinese Medicine, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Xin Chen
- Hubei Key Laboratory of Resources and Chemistry of Chinese Medicine, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China; Hubei Shizhen Laboratory, Wuhan 430061, China
| | - Zhenpeng Qiu
- Hubei Key Laboratory of Resources and Chemistry of Chinese Medicine, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China; Hubei Shizhen Laboratory, Wuhan 430061, China; Center of Traditional Chinese Medicine Modernization for Liver Diseases, Hubei University of Chinese Medicine, Wuhan, 430065, China.
| |
Collapse
|
41
|
Xie L, Hao X, Xie J, Mo J, Yuan C, Chen W. Acetylated pelargonidin-3- O-glucoside alleviates hepatocyte lipid deposition through activating the AMPK-mediated lysosome-autophagy pathway and redox state. Food Funct 2024; 15:6929-6942. [PMID: 38659316 DOI: 10.1039/d4fo00185k] [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: 04/26/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a worldwide public health issue, but a widely accepted therapy is still lacking until now. Anthocyanins are natural flavonoid compounds that possess various bioactivities, but their applications are limited due to their low bioavailability and stability. Acylated anthocyanins are reported to show higher stability, whereas their effects on NAFLD are still unclear. Herein, pelargonidin-3-O-(6''-acetyl)-glucoside (Ace Pg3G) was found to dose-dependently reduce intracellular lipid droplets and triglycerides, and improve cellular oxidative stress that accompanied lipid deposition. Besides, Ace Pg3G was proved to activate AMPK phosphorylation, thus stimulating AMPK-mediated lysosome-autophagy pathway to eliminate overloaded lipid. Further study unveiled that Ace Pg3G regulated genes related to lipid metabolism downstream of AMPK to inhibit lipid synthesis and accelerate lipid oxidation. Overall, this study provided the first evidence, to our best knowledge, that Ace Pg3G ameliorated free fatty acid-induced lipid deposition in hepatocytes through regulating AMPK-mediated autophagy pathways and redox state.
Collapse
Affiliation(s)
- Lianghua Xie
- Department of Traditional Chinese Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China.
- Department of Food Science and Nutrition, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, China
| | - Xin Hao
- Department of Food Science and Nutrition, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, China
| | - Jiahong Xie
- Department of Food Science and Nutrition, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, China
| | - Jianling Mo
- Department of Traditional Chinese Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China.
| | - Changzheng Yuan
- School of Public Health, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - Wei Chen
- Department of Traditional Chinese Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China.
- Department of Food Science and Nutrition, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, China
| |
Collapse
|
42
|
Hirata A, Harada S, Iida M, Kurihara A, Fukai K, Kuwabara K, Kato S, Matsumoto M, Sata M, Miyagawa N, Toki R, Edagawa S, Sugiyama D, Sato A, Hirayama A, Sugimoto M, Soga T, Tomita M, Okamura T, Takebayashi T. Association of Nonalcoholic Fatty Liver Disease with Arterial Stiffness and its Metabolomic Profiling in Japanese Community-Dwellers. J Atheroscler Thromb 2024; 31:1031-1047. [PMID: 38311416 PMCID: PMC11224684 DOI: 10.5551/jat.64616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 11/28/2023] [Indexed: 02/10/2024] Open
Abstract
AIMS Nonalcoholic fatty liver disease (NAFLD) is known to be associated with atherosclerosis. This study focused on upstream changes in the process by which NAFLD leads to atherosclerosis. The study aimed to confirm the association between NAFLD and the cardio-ankle vascular index (CAVI), an indicator of subclinical atherosclerosis, and explore metabolites involved in both by assessing 94 plasma polar metabolites. METHODS A total of 928 Japanese community-dwellers (306 men and 622 women) were included in this study. The association between NAFLD and CAVI was examined using a multivariable regression model adjusted for confounders. Metabolites commonly associated with NAFLD and CAVI were investigated using linear mixed-effects models in which batch numbers of metabolite measurements were used as a random-effects variable, and false discovery rate-adjusted p-values were calculated. To determine the extent to which these metabolites mediated the association between NAFLD and CAVI, mediation analysis was conducted. RESULTS NAFLD was positively associated with CAVI (coefficients [95% Confidence intervals (CI)]=0.23 [0.09-0.37]; p=0.001). A total of 10 metabolites were involved in NAFLD and CAVI, namely, branched-chain amino acids (BCAAs; valine, leucine, and isoleucine), aromatic amino acids (AAAs; tyrosine and tryptophan), alanine, proline, glutamic acid, glycerophosphorylcholine, and 4-methyl-2-oxopentanoate. Mediation analysis showed that BCAAs mediated more than 20% of the total effect in the association between NAFLD and CAVI. CONCLUSIONS NAFLD was associated with a marker of atherosclerosis, and several metabolites related to insulin resistance, including BCAAs and AAAs, could be involved in the process by which NAFLD leads to atherosclerosis.
Collapse
Affiliation(s)
- Aya Hirata
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, Tokyo, Japan
| | - Sei Harada
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, Tokyo, Japan
- Institute for Advanced Biosciences, Keio University, Yamagata, Japan
| | - Miho Iida
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, Tokyo, Japan
| | - Ayako Kurihara
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, Tokyo, Japan
| | - Kota Fukai
- Department of Preventive Medicine, Tokai University School of Medicine, Kanagawa, Japan
| | - Kazuyo Kuwabara
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, Tokyo, Japan
| | - Suzuka Kato
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, Tokyo, Japan
| | - Minako Matsumoto
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, Tokyo, Japan
| | - Mizuki Sata
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, Tokyo, Japan
| | - Naoko Miyagawa
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, Tokyo, Japan
| | - Ryota Toki
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, Tokyo, Japan
| | - Shun Edagawa
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, Tokyo, Japan
| | - Daisuke Sugiyama
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, Tokyo, Japan
- Faculty of Nursing and Medical Care, Keio University, Kanagawa, Japan
| | - Asako Sato
- Institute for Advanced Biosciences, Keio University, Yamagata, Japan
| | - Akiyoshi Hirayama
- Institute for Advanced Biosciences, Keio University, Yamagata, Japan
| | - Masahiro Sugimoto
- Institute for Advanced Biosciences, Keio University, Yamagata, Japan
- Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Tomoyoshi Soga
- Institute for Advanced Biosciences, Keio University, Yamagata, Japan
- Faculty of Environment and Information Studies, Keio University, Kanagawa, Japan
| | - Masaru Tomita
- Institute for Advanced Biosciences, Keio University, Yamagata, Japan
- Faculty of Environment and Information Studies, Keio University, Kanagawa, Japan
| | - Tomonori Okamura
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, Tokyo, Japan
| | - Toru Takebayashi
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, Tokyo, Japan
- Institute for Advanced Biosciences, Keio University, Yamagata, Japan
| |
Collapse
|
43
|
Atallah E, Trehiou S, Alquier-Bacquie V, Lasserre F, Arroyo J, Molette C, Remignon H. Development of hepatic steatosis in male and female mule ducks after respective force-feeding programs. Front Physiol 2024; 15:1392968. [PMID: 38974520 PMCID: PMC11224645 DOI: 10.3389/fphys.2024.1392968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 05/24/2024] [Indexed: 07/09/2024] Open
Abstract
Male and female mule ducks were subjected to a force-feeding diet to induce liver steatosis as it is generally done only with male ducks for the production of foie gras. The different biochemical measurements indicated that the course of hepatic steatosis development was present in both sexes and associated with a huge increase in liver weight mainly due to the synthesis and accumulation of lipids in hepatocytes. In livers of male and female ducks, this lipid accumulation was associated with oxidative stress and hypoxia. However, certain specific modifications (kinetics of lipid droplet development and hepatic inflammation) indicate that female ducks may tolerate force-feeding less well, at least at the hepatic level. This is in contradiction with what is generally reported concerning hepatic steatosis induced by dietary disturbances in mammals but could be explained by the very specific conditions imposed by force-feeding. Despite this, force-feeding female ducks seems entirely feasible, provided that the final quality of the product is as good as that of the male ducks, which will remain to be demonstrated in future studies.
Collapse
Affiliation(s)
- Elham Atallah
- Toxalim (Research Centre in Food Toxicology), INRAE, ENVT, UPS, Université de Toulouse, Toulouse, France
| | - Sabrina Trehiou
- Toxalim (Research Centre in Food Toxicology), INRAE, ENVT, UPS, Université de Toulouse, Toulouse, France
| | - Valérie Alquier-Bacquie
- Toxalim (Research Centre in Food Toxicology), INRAE, ENVT, UPS, Université de Toulouse, Toulouse, France
| | - Frédéric Lasserre
- Toxalim (Research Centre in Food Toxicology), INRAE, ENVT, UPS, Université de Toulouse, Toulouse, France
| | | | | | - Hervé Remignon
- Toxalim (Research Centre in Food Toxicology), INRAE, ENVT, UPS, Université de Toulouse, Toulouse, France
- INP-ENSAT, Université de Toulouse, Castanet-Tolosan, France
| |
Collapse
|
44
|
Ramos-Molina B, Rossell J, Pérez-Montes de Oca A, Pardina E, Genua I, Rojo-López MI, Julián MT, Alonso N, Julve J, Mauricio D. Therapeutic implications for sphingolipid metabolism in metabolic dysfunction-associated steatohepatitis. Front Endocrinol (Lausanne) 2024; 15:1400961. [PMID: 38962680 PMCID: PMC11220194 DOI: 10.3389/fendo.2024.1400961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 06/03/2024] [Indexed: 07/05/2024] Open
Abstract
The prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD), a leading cause of chronic liver disease, has increased worldwide along with the epidemics of obesity and related dysmetabolic conditions characterized by impaired glucose metabolism and insulin signaling, such as type 2 diabetes mellitus (T2D). MASLD can be defined as an excessive accumulation of lipid droplets in hepatocytes that occurs when the hepatic lipid metabolism is totally surpassed. This metabolic lipid inflexibility constitutes a central node in the pathogenesis of MASLD and is frequently linked to the overproduction of lipotoxic species, increased cellular stress, and mitochondrial dysfunction. A compelling body of evidence suggests that the accumulation of lipid species derived from sphingolipid metabolism, such as ceramides, contributes significantly to the structural and functional tissue damage observed in more severe grades of MASLD by triggering inflammatory and fibrogenic mechanisms. In this context, MASLD can further progress to metabolic dysfunction-associated steatohepatitis (MASH), which represents the advanced form of MASLD, and hepatic fibrosis. In this review, we discuss the role of sphingolipid species as drivers of MASH and the mechanisms involved in the disease. In addition, given the absence of approved therapies and the limited options for treating MASH, we discuss the feasibility of therapeutic strategies to protect against MASH and other severe manifestations by modulating sphingolipid metabolism.
Collapse
Affiliation(s)
- Bruno Ramos-Molina
- Group of Obesity, Diabetes & Metabolism, Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, Spain
| | - Joana Rossell
- Group of Endocrinology, Diabetes & Nutrition, Institut de Recerca SANT PAU, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Madrid, Spain
| | | | - Eva Pardina
- Department de Biochemistry & Molecular Biology, Facultat de Biologia, Universitat de Barcelona (UB), Barcelona, Spain
| | - Idoia Genua
- Department of Endocrinology & Nutrition, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Marina I. Rojo-López
- Group of Endocrinology, Diabetes & Nutrition, Institut de Recerca SANT PAU, Barcelona, Spain
| | - María Teresa Julián
- Department of Endocrinology & Nutrition, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Núria Alonso
- Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Madrid, Spain
- Department of Endocrinology & Nutrition, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Josep Julve
- Group of Endocrinology, Diabetes & Nutrition, Institut de Recerca SANT PAU, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Madrid, Spain
| | - Didac Mauricio
- Group of Endocrinology, Diabetes & Nutrition, Institut de Recerca SANT PAU, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Madrid, Spain
- Department of Endocrinology & Nutrition, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
- Department of Endocrinology & Nutrition, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Faculty of Medicine, University of Vic/Central University of Catalonia (UVIC/UCC), Vic, Spain
| |
Collapse
|
45
|
Shi Y, Taherifard E, Saeed A, Saeed A. MASLD-Related HCC: A Comprehensive Review of the Trends, Pathophysiology, Tumor Microenvironment, Surveillance, and Treatment Options. Curr Issues Mol Biol 2024; 46:5965-5983. [PMID: 38921027 PMCID: PMC11202630 DOI: 10.3390/cimb46060356] [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: 05/11/2024] [Revised: 06/07/2024] [Accepted: 06/11/2024] [Indexed: 06/27/2024] Open
Abstract
Hepatocellular carcinoma (HCC) represents a significant burden on global healthcare systems due to its considerable incidence and mortality rates. Recent trends indicate an increase in the worldwide incidence of metabolic dysfunction-associated steatotic liver disease (MASLD) and a shift in the etiology of HCC, with MASLD replacing the hepatitis B virus as the primary contributor to new cases of HCC. MASLD-related HCC exhibits distinct characteristics compared to viral HCC, including unique immune cell profiles resulting in an overall more immunosuppressive or exhausted tumor microenvironment. Furthermore, MASLD-related HCC is frequently identified in older age groups and among individuals with cardiometabolic comorbidities. Additionally, a greater percentage of MASLD-related HCC cases occur in noncirrhotic patients compared to those with viral etiologies, hindering early detection. However, the current clinical practice guidelines lack specific recommendations for the screening of HCC in MASLD patients. The evolving landscape of HCC management offers a spectrum of therapeutic options, ranging from surgical interventions and locoregional therapies to systemic treatments, for patients across various stages of the disease. Despite ongoing debates, the current evidence does not support differences in optimal treatment modalities based on etiology. In this study, we aimed to provide a comprehensive overview of the current literature on the trends, characteristics, clinical implications, and treatment modalities for MASLD-related HCC.
Collapse
Affiliation(s)
- Yuming Shi
- Department of Medicine, Division of Hematology & Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA 15232, USA; (Y.S.); (E.T.)
| | - Erfan Taherifard
- Department of Medicine, Division of Hematology & Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA 15232, USA; (Y.S.); (E.T.)
| | - Ali Saeed
- Department of Medicine, Ochsner Lafayette General Medical Center, Lafayette, LA 70503, USA;
| | - Anwaar Saeed
- Department of Medicine, Division of Hematology & Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA 15232, USA; (Y.S.); (E.T.)
- UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| |
Collapse
|
46
|
Bitterer F, Kupke P, Adenugba A, Evert K, Glehr G, Riquelme P, Scheibert L, Preverin G, Böhm C, Hornung M, Schlitt HJ, Wenzel JJ, Geissler EK, Safinia N, Hutchinson JA, Werner JM. Soluble CD46 as a diagnostic marker of hepatic steatosis. EBioMedicine 2024; 104:105184. [PMID: 38838471 PMCID: PMC11179574 DOI: 10.1016/j.ebiom.2024.105184] [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: 01/05/2024] [Revised: 05/21/2024] [Accepted: 05/21/2024] [Indexed: 06/07/2024] Open
Abstract
BACKGROUND The increasing prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD) incurs substantial morbidity, mortality and healthcare costs. Detection and clinical intervention at early stages of disease improves prognosis; however, we are currently limited by a lack of reliable diagnostic tests for population screening and monitoring responses to therapy. To address this unmet need, we investigated human invariant Natural Killer T cell (iNKT) activation by fat-loaded hepatocytes, leading to the discovery that circulating soluble CD46 (sCD46) levels accurately predict hepatic steatosis. METHODS sCD46 in plasma was measured using a newly developed immuno-competition assay in two independent cohorts: Prospective living liver donors (n = 156; male = 66, female = 90) and patients with liver tumours (n = 91; male = 58, female = 33). sCD46 levels were statistically evaluated as a predictor of hepatic steatosis. FINDINGS Interleukin-4-secreting (IL-4+) iNKT cells were over-represented amongst intrahepatic lymphocytes isolated from resected human liver samples. IL-4+ iNKT cells preferentially developed in cocultures with a fat-loaded, hepatocyte-like cell line, HepaRG. This was attributed to induction of matrix metalloproteases (MMP) in fat-loaded HepaRG cells and primary human liver organoids, which led to indiscriminate cleavage of immune receptors. Loss of cell-surface CD46 resulted in unrepressed differentiation of IL-4+ iNKT cells. sCD46 levels were elevated in patients with hepatic steatosis. Discriminatory cut-off values for plasma sCD46 were found that accurately classified patients according to histological steatosis grade. INTERPRETATION sCD46 is a reliable clinical marker of hepatic steatosis, which can be conveniently and non-invasively measured in serum and plasma samples, raising the possibility of using sCD46 levels as a diagnostic method for detecting or grading hepatic steatosis. FUNDING F.B. was supported by the Else Kröner Foundation (Award 2016_kolleg.14). G.G. was supported by the Bristol Myers Squibb Foundation for Immuno-Oncology (Award FA-19-009). N.S. was supported by a Wellcome Trust Fellowship (211113/A/18/Z). J.A.H. received funding from the European Union's Horizon 2020 research and innovation programme (Award 860003). J.M.W. received funding from the Else Kröner Foundation (Award 2015_A10).
Collapse
Affiliation(s)
- Florian Bitterer
- Department of Surgery, University Hospital Regensburg, Regensburg 93053, Germany
| | - Paul Kupke
- Department of Surgery, University Hospital Regensburg, Regensburg 93053, Germany
| | - Akinbami Adenugba
- Department of Surgery, University Hospital Regensburg, Regensburg 93053, Germany
| | - Katja Evert
- Institute of Pathology, University of Regensburg, Regensburg 93053, Germany
| | - Gunther Glehr
- Department of Surgery, University Hospital Regensburg, Regensburg 93053, Germany
| | - Paloma Riquelme
- Department of Surgery, University Hospital Regensburg, Regensburg 93053, Germany
| | - Lena Scheibert
- Department of Surgery, University Hospital Regensburg, Regensburg 93053, Germany
| | - Giulia Preverin
- Department of Surgery, University Hospital Regensburg, Regensburg 93053, Germany
| | - Christina Böhm
- Oxford Nanopore Technologies PLC, Oxford Science Park, Oxford OX4 4DQ, United Kingdom
| | - Matthias Hornung
- Department of Surgery, University Hospital Regensburg, Regensburg 93053, Germany
| | - Hans J Schlitt
- Department of Surgery, University Hospital Regensburg, Regensburg 93053, Germany
| | - Jürgen J Wenzel
- Institute of Clinical Microbiology and Hygiene, University of Regensburg, Regensburg 93053, Germany
| | - Edward K Geissler
- Department of Surgery, University Hospital Regensburg, Regensburg 93053, Germany
| | - Niloufar Safinia
- Department of Hepatology, King's College London, London SE5 8AF, United Kingdom
| | - James A Hutchinson
- Department of Surgery, University Hospital Regensburg, Regensburg 93053, Germany.
| | - Jens M Werner
- Department of Surgery, University Hospital Regensburg, Regensburg 93053, Germany.
| |
Collapse
|
47
|
Li X, He W, Chen X, Zhang Y, Zhang J, Liu F, Li J, Zhao D, Xia P, Ma W, Wu T, Wang H, Yuan Y. TRIM45 facilitates NASH-progressed HCC by promoting fatty acid synthesis via catalyzing FABP5 ubiquitylation. Oncogene 2024; 43:2063-2077. [PMID: 38755308 DOI: 10.1038/s41388-024-03056-7] [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: 12/04/2023] [Revised: 04/23/2024] [Accepted: 04/29/2024] [Indexed: 05/18/2024]
Abstract
Non-alcoholic steatohepatitis (NASH) is rapidly surpassing viral hepatitis as the primary cause of hepatocellular carcinoma (HCC). However, understanding of NASH-progressed HCC remains poor, which might impede HCC diagnosis and therapy. In this study, we aim to identify shared transcriptional changes between NASH and HCC, of which we focused on E3 ligase TRIM45. We found TRIM45 exacerbates HCC cells proliferation and metastasis in vitro and in vivo. Further transcriptome analysis revealed TRIM45 predominantly affects fatty acid metabolism and oleic acid restored impaired proliferation and metastasis of TRIM45-deficient HCC cells. IP-tandem mass spectrum and FABP5 depriving experiment indicated that TRIM45 enhance fatty acid synthesis depending on FABP5 presence. Interestingly, we found TRIM45 directly added K33-type and K63-type poly-ubiquitin chains to FABP5 NLS domain, which ultimately promoted FABP5 nuclear translocation. Nuclear FABP5 interacted with PPARγ to facilitate downstream lipid synthesis gene expression. We observed TRIM45 accelerated NASH-to-HCC transition and exacerbated both NASH and NASH-HCC with the enhanced fatty acid production in vivo. Moreover, high concentration of fatty acid increased TRIM45 expression. The established mechanism was substantiated by gene expression correlation in TCGA-LIHC. Collectively, our research revealed a common lipid reprograming process in NASH and HCC and identified the cyclical amplification of the TRIM45-FABP5-PPARγ-fatty acid axis. This signaling pathway offers potential therapeutic targets for therapeutic intervention in NASH and NASH-progressed HCC.
Collapse
Affiliation(s)
- Xiaomian Li
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, China
| | - Wenzhi He
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, China
- College of Life Sciences, Hubei Key Laboratory of Cell Homeostasis, Wuhan University, Wuhan, China
| | - Xi Chen
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, China
| | - Yangwenqing Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, China
| | - Jia Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, China
| | - Fusheng Liu
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, China
| | - Jinghua Li
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, China
| | - Dongli Zhao
- College of Life Sciences, Hubei Key Laboratory of Cell Homeostasis, Wuhan University, Wuhan, China
| | - Peng Xia
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, China
| | - Weijie Ma
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, China
| | - Tiangen Wu
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China.
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, China.
| | - Haitao Wang
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China.
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, China.
| | - Yufeng Yuan
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China.
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, China.
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
48
|
Małkowska P. Positive Effects of Physical Activity on Insulin Signaling. Curr Issues Mol Biol 2024; 46:5467-5487. [PMID: 38920999 PMCID: PMC11202552 DOI: 10.3390/cimb46060327] [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: 04/23/2024] [Revised: 05/24/2024] [Accepted: 05/28/2024] [Indexed: 06/27/2024] Open
Abstract
Physical activity is integral to metabolic health, particularly in addressing insulin resistance and related disorders such as type 2 diabetes mellitus (T2DM). Studies consistently demonstrate a strong association between physical activity levels and insulin sensitivity. Regular exercise interventions were shown to significantly improve glycemic control, highlighting exercise as a recommended therapeutic strategy for reducing insulin resistance. Physical inactivity is closely linked to islet cell insufficiency, exacerbating insulin resistance through various pathways including ER stress, mitochondrial dysfunction, oxidative stress, and inflammation. Conversely, physical training and exercise preserve and restore islet function, enhancing peripheral insulin sensitivity. Exercise interventions stimulate β-cell proliferation through increased circulating levels of growth factors, further emphasizing its role in maintaining pancreatic health and glucose metabolism. Furthermore, sedentary lifestyles contribute to elevated oxidative stress levels and ceramide production, impairing insulin signaling and glucose metabolism. Regular exercise induces anti-inflammatory responses, enhances antioxidant defenses, and promotes mitochondrial function, thereby improving insulin sensitivity and metabolic efficiency. Encouraging individuals to adopt active lifestyles and engage in regular exercise is crucial for preventing and managing insulin resistance and related metabolic disorders, ultimately promoting overall health and well-being.
Collapse
Affiliation(s)
- Paulina Małkowska
- Institute of Physical Culture Sciences, University of Szczecin, 71-065 Szczecin, Poland
| |
Collapse
|
49
|
Jia W, Zhong L, Ren Q, Teng D, Gong L, Dong H, Li J, Wang C, He YX, Yang J. Microcystin-RR promote lipid accumulation through CD36 mediated signal pathway and fatty acid uptake in HepG2 cells. ENVIRONMENTAL RESEARCH 2024; 249:118402. [PMID: 38309560 DOI: 10.1016/j.envres.2024.118402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 01/19/2024] [Accepted: 02/01/2024] [Indexed: 02/05/2024]
Abstract
Microcystins (MC)-RR is a significant analogue of MC-LR, which has been identified as a hepatotoxin capable of influencing lipid metabolism and promoting the progression of liver-related metabolic diseases. However, the toxicity and biological function of MC-RR are still not well understood. In this study, the toxic effects and its role in lipid metabolism of MC-RR were investigated in hepatoblastoma cells (HepG2cells). The results demonstrated that MC-RR dose-dependently reduced cell viability and induced apoptosis. Additionally, even at low concentrations, MC-RR promoted lipid accumulation through up-regulating levels of triglyceride, total cholesterol, phosphatidylcholines and phosphatidylethaolamine in HepG2 cells, with no impact on cell viability. Proteomics and transcriptomics analysis further revealed significant alterations in the protein and gene expression profiles in HepG2 cells treated with MC-RR. Bioinformatic analysis, along with subsequent validation, indicated the upregulation of CD36 and activation of the AMPK and PI3K/AKT/mTOR in response to MC-RR exposure. Finally, knockdown of CD36 markedly ameliorated MC-RR-induced lipid accumulation in HepG2 cells. These findings collectively suggest that MC-RR promotes lipid accumulation in HepG2 cells through CD36-mediated signal pathway and fatty acid uptake. Our findings provide new insights into the hepatotoxic mechanism of MC-RR.
Collapse
Affiliation(s)
- Wenjuan Jia
- School of Basic Medical Sciences, Qingdao University, Qingdao, 266071, China; Department of Cardiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, China.
| | - Lin Zhong
- School of Basic Medical Sciences, Qingdao University, Qingdao, 266071, China; Department of Cardiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, China
| | - Qingmiao Ren
- The Precision Medicine Laboratory, The First Hospital of Lanzhou University, Lanzhou, 730000, China
| | - Da Teng
- School of Basic Medical Sciences, Qingdao University, Qingdao, 266071, China; Department of Cardiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, China
| | - Lei Gong
- Department of Cardiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, China
| | - Haibin Dong
- Department of Cardiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, China
| | - Jun Li
- School of Basic Medical Sciences, Qingdao University, Qingdao, 266071, China; Department of Cardiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, China
| | - Chunxiao Wang
- Department of Cardiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, China
| | - Yong-Xing He
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, 730000, China.
| | - Jun Yang
- Department of Cardiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, China.
| |
Collapse
|
50
|
Pozo-Morales M, Cobham AE, Centola C, McKinney MC, Liu P, Perazzolo C, Lefort A, Libert F, Bai H, Rohner N, Singh SP. Starvation-resistant cavefish reveal conserved mechanisms of starvation-induced hepatic lipotoxicity. Life Sci Alliance 2024; 7:e202302458. [PMID: 38467419 PMCID: PMC10927358 DOI: 10.26508/lsa.202302458] [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: 10/26/2023] [Revised: 02/14/2024] [Accepted: 02/19/2024] [Indexed: 03/13/2024] Open
Abstract
Starvation causes the accumulation of lipid droplets in the liver, a somewhat counterintuitive phenomenon that is nevertheless conserved from flies to humans. Much like fatty liver resulting from overfeeding, hepatic lipid accumulation (steatosis) during undernourishment can lead to lipotoxicity and atrophy of the liver. Here, we found that although surface populations of Astyanax mexicanus undergo this evolutionarily conserved response to starvation, the starvation-resistant cavefish larvae of the same species do not display an accumulation of lipid droplets upon starvation. Moreover, cavefish are resistant to liver atrophy during starvation, providing a unique system to explore strategies for liver protection. Using comparative transcriptomics between zebrafish, surface fish, and cavefish, we identified the fatty acid transporter slc27a2a/fatp2 to be correlated with the development of fatty liver. Pharmacological inhibition of slc27a2a in zebrafish rescues steatosis and atrophy of the liver upon starvation. Furthermore, down-regulation of FATP2 in Drosophila larvae inhibits the development of starvation-induced steatosis, suggesting the evolutionarily conserved importance of the gene in regulating fatty liver upon nutrition deprivation. Overall, our study identifies a conserved, druggable target to protect the liver from atrophy during starvation.
Collapse
Affiliation(s)
| | - Ansa E Cobham
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Cielo Centola
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | | | - Peiduo Liu
- Department of Genetics, Development, and Cell Biology, Iowa State University, Ames, IA, USA
| | | | - Anne Lefort
- IRIBHM, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | | | - Hua Bai
- Department of Genetics, Development, and Cell Biology, Iowa State University, Ames, IA, USA
| | - Nicolas Rohner
- Stowers Institute for Medical Research, Kansas City, MO, USA
- Department of Cell Biology & Physiology, University of Kansas Medical Center, Kansas City, KS, USA
| | | |
Collapse
|