1
|
Jian J, Zhang R, Dong Y, Zheng H, Liao X. Association between NAFLD and liver fibrosis with nutritional risk index based on the NHANES 2017-2018. Lipids Health Dis 2025; 24:6. [PMID: 39773264 PMCID: PMC11705686 DOI: 10.1186/s12944-024-02427-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 12/30/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Nutrition and its associated inflammation have been acknowledged as vital factors in the etiopathogenesis of non-alcoholic fatty liver disease (NAFLD) and liver fibrosis. The nutritional risk index (NRI) has been widely recognized as a valid indicator of nutritional status in several diseases, including osteoporosis and cardiovascular disease. However, the role of NRI in NAFLD and liver fibrosis remains unclear. METHODS Participants were selected from the National Health and Nutrition Examination Survey data for the 2017-2018 cycle. Association between NRI and both NAFLD and liver fibrosis was evaluated using multiple logistic regression and restricted cubic spline (RCS) analysis. Mediation analysis was employed to assess the influence of inflammation on the association between NRI and both NAFLD and liver fibrosis. RESULTS Compared to their respective control groups, individuals with NAFLD and liver fibrosis exhibited higher NRI levels. Multiple logistic regression analyses indicated that NRI was positively associated with the odds of NAFLD and liver fibrosis across both continuous scales and quantile groups, with adjustments for relevant covariables. The RCS model demonstrated a dose-response effect between NRI and the odds of NAFLD, but not with liver fibrosis. Receiver operating characteristic (ROC) analysis revealed the area under the ROC curves of 0.798 and 0.775 for NAFLD and liver fibrosis, respectively. Mediation analysis showed that inflammation accounted for 3.139% of the effect of NRI on the odds of NAFLD, suggesting inflammation might partially mediate the impact of NRI on NAFLD. CONCLUSIONS Our findings indicate that NRI may serve as a potential associated marker for these liver diseases, underscoring the importance of nutritional status in their etiopathogenesis.
Collapse
Affiliation(s)
- Jieming Jian
- Department of Endocrinology, Translational Research of Diabetes Key Laboratory of Chongqing Education Commission of China, the Second Affiliated Hospital of Army Medical University, Chongqing, 400037, China
| | - Rui Zhang
- Department of Endocrinology, Translational Research of Diabetes Key Laboratory of Chongqing Education Commission of China, the Second Affiliated Hospital of Army Medical University, Chongqing, 400037, China
| | - Yuan Dong
- Department of Endocrinology, Translational Research of Diabetes Key Laboratory of Chongqing Education Commission of China, the Second Affiliated Hospital of Army Medical University, Chongqing, 400037, China
| | - Hongting Zheng
- Department of Endocrinology, Translational Research of Diabetes Key Laboratory of Chongqing Education Commission of China, the Second Affiliated Hospital of Army Medical University, Chongqing, 400037, China.
| | - Xiaoyu Liao
- Department of Endocrinology, Translational Research of Diabetes Key Laboratory of Chongqing Education Commission of China, the Second Affiliated Hospital of Army Medical University, Chongqing, 400037, China.
| |
Collapse
|
2
|
Li J, Zhang Y, Ma X, Liu R, Xu C, He Q, Dong M. Identification and validation of cuproptosis-related genes for diagnosis and therapy in nonalcoholic fatty liver disease. Mol Cell Biochem 2025; 480:473-489. [PMID: 38512536 DOI: 10.1007/s11010-024-04957-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 02/03/2024] [Indexed: 03/23/2024]
Abstract
In recent years, nonalcoholic fatty liver disease (NAFLD) has become a more serious public health issue worldwide. This study strived to investigate the molecular mechanism of pathogenesis of NAFLD and explore promising diagnostic and therapeutic targets for NAFLD. Raw data from GSE130970 were downloaded from the Gene Expression Omnibus database. We used the dataset to analyze the expression levels of cuproptosis-related genes in NAFLD patients and healthy controls to identify the differentially expressed cuproptosis-related genes (DECRGs). The relationship and potential mechanism between DECRGs and clinicopathological factors were examined by enrichment analysis and two consensus clustering methods. We screened key DECRGs based on Random Forest (RF), and then verified the key DECRGs in NAFLD patients, high-fat diet (HFD)-fed mice, and palmitic acid-induced AML12 cells. ROC analysis showed good diagnostic function of DECRGs in normal and NAFLD liver tissue. Two consensus clusters indicated the important role of cuproptosis in the development of NAFLD. We screened for key DECRGs (DLD, DLAT) based on RF and found a close relationship between the DECRGs and clinicopathological factors. We collected clinical blood samples to verify the differences in gene expression levels by qPCR. In addition, we further verified the expression levels of DLD and DLAT in HFD mice and AML12 cells, which showed the same results. This study provides a novel perspective on the pathogenesis of NAFLD. We identified two cuproptosis-related genes that are closely related to NAFLD. These genes may play a significant role in the molecular pathogenesis of NAFLD, which may be useful to make progress in the diagnosis and treatment of NAFLD.
Collapse
Affiliation(s)
- Jinquan Li
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Yi Zhang
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Xiaohan Ma
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Ruiqi Liu
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Cuicui Xu
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Qin He
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, Shandong, China.
- Institute of Endocrine and Metabolic Diseases of Shandong University, Jinan, Shandong, China.
- Jinan Clinical Research Center for Endocrine and Metabolic Disease, Jinan, Shandong, China.
- Key Laboratory of Endocrine and Metabolic Diseases, Shandong Province Medicine & Health, Jinan, Shandong, China.
| | - Ming Dong
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, Shandong, China.
- Institute of Endocrine and Metabolic Diseases of Shandong University, Jinan, Shandong, China.
- Jinan Clinical Research Center for Endocrine and Metabolic Disease, Jinan, Shandong, China.
- Key Laboratory of Endocrine and Metabolic Diseases, Shandong Province Medicine & Health, Jinan, Shandong, China.
| |
Collapse
|
3
|
Islam MA, Khairnar R, Fleishman J, Reznik SE, Ragolia L, Gobbooru S, Kumar S. Female C57BL/6 mice exhibit protection against nonalcoholic fatty liver disease and diabesity accompanied by differential regulation of hepatic lipocalin prostaglandin D 2 synthase. Mol Cell Endocrinol 2025; 595:112404. [PMID: 39505230 DOI: 10.1016/j.mce.2024.112404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 10/21/2024] [Accepted: 10/28/2024] [Indexed: 11/08/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD) and its development into nonalcoholic steatohepatitis (NASH) are challenging health concerns globally. Clinically, the prevalence and severity of NAFLD/NASH are higher in men than in premenopausal women. NAFLD is strongly correlated with obesity, both of which are tied to high-fat/fructose-rich western diets. Therefore, we aimed to investigate sexual dimorphism in NAFLD pathogenesis in male and female C57BL/6 mice fed different diets. Male and female C57BL/67 mice were divided into four groups and kept on a chow (C), chow plus high fructose (CF), high fat (HF), and high fat plus high fructose (HFF) diet for 22 weeks. Liver tissues were collected at the end of the study and processed for NAFLD/NASH-related histology (H&E and trichrome staining), protein expression (SREBP1, SCAP, FABP4, α-SMA, TGF-β and L-PGDS), and biochemical parameters measurement. Our results displayed that female mice exhibited protection against NAFLD and diabesity on HF and HFF diets compared to male mice fed similar diets. Additionally, female mice showed protection from fibrosis compared to male mice. Both male and female mice fed HF and HFF diet groups displayed the cytosol-to-nuclear translocation of Lipocalin Prostaglandin D2 Synthase (L-PGDS). Cytoplasmic levels of L-PGDS were absent in females compared to low levels in males, revealing a possible sex-specific mechanism tied to fructose and fat metabolism. Collectively, female mice showed protection against NAFLD and diabesity relative to male mice, accompanied by differential regulation of hepatic lipocalin prostaglandin D2 synthase.
Collapse
Affiliation(s)
- Md Asrarul Islam
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Rhema Khairnar
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Joshua Fleishman
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Sandra E Reznik
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Louis Ragolia
- NYU Grossman Long Island School of Medicine, Mineola, NY, 11501, USA
| | - Shruthi Gobbooru
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Sunil Kumar
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA.
| |
Collapse
|
4
|
Park JS, Ma YQ, Wang F, Ma H, Sui G, Rustamov N, Han M, Son Y, Park CW, Han SB, Hong JT, Jeong LS, Lee J, Roh YS. A3AR antagonism mitigates metabolic dysfunction-associated steatotic liver disease by exploiting monocyte-derived Kupffer cell necroptosis and inflammation resolution. Metabolism 2024; 164:156114. [PMID: 39732364 DOI: 10.1016/j.metabol.2024.156114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/17/2024] [Accepted: 12/20/2024] [Indexed: 12/30/2024]
Abstract
BACKGROUND & AIMS Metabolic dysfunction-associated steatotic liver (MASLD) progression is driven by chronic inflammation and fibrosis, largely influenced by Kupffer cell (KC) dynamics, particularly replenishment of pro-inflammatory monocyte-derived KCs (MoKCs) due to increased death of embryo-derived KCs. Adenosine A3 receptor (A3AR) plays a key role in regulating metabolism and immune responses, making it a promising therapeutic target. This study aimed to investigate the impact of selective A3AR antagonism for regulation of replenished MoKCs, thereby improving MASLD. APPROACH & RESULTS A3AR expression was significantly elevated in KCs from both patients with MASLD and fast-food diet (FFD)-fed mice. A3AR knockout (KO) mice displayed marked improvements in hepatic inflammation and fibrosis along with a reduction in CLEC4F-positive KCs. The spatial transcriptomics of these KCs revealed disrupted mitochondrial integrity, increased oxidative stress, and enhanced cell death due to A3AR deletion. Similarly, in vivo FM101 treatment, a highly potent and selective antagonist of A3AR with a truncated 4'-thioadenosine structure, mitigated FFD-induced MASLD in mice. Mechanistically, FM101 induces β-arrestin2-mediated A3AR degradation, leading to mitochondrial dysfunction-mediated necroptosis in KCs. Consistently, A3AR was highly expressed in monocyte-derived macrophages in MASLD patients, with strong correlations with macrophage activation and monocyte chemoattractant gene sets. Thus, FM101 induced necroptosis in pro-inflammatory MoKCs, facilitating anti-inflammatory effects. CONCLUSIONS This study demonstrated that inhibiting A3AR via FM101 or genetic deletion alleviates MASLD by inducing mitochondrial dysfunction and subsequent necroptosis in MoKCs, establishing FM101 as a promising therapeutic strategy for MASLD.
Collapse
Affiliation(s)
- Jeong-Su Park
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, South Korea
| | - Yuan-Qiang Ma
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, South Korea
| | - Feng Wang
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, South Korea
| | - Hwan Ma
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, South Korea
| | - Guoyan Sui
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, South Korea
| | - Nodir Rustamov
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, South Korea
| | - Minyeong Han
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, South Korea
| | - Yejin Son
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, South Korea
| | - Chun-Woong Park
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, South Korea
| | - Sang-Bae Han
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, South Korea
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, South Korea
| | - Lak Shin Jeong
- Research and Development Center, Future Medicine Co., Ltd, Seongnam, South Korea; College of Pharmacy, Seoul National University, Seoul, 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, Chungbuk, South Korea.
| |
Collapse
|
5
|
Duan S, Tu Z, Duan L, Tu R. Differential effects of systemic immune inflammation indices on hepatic steatosis and hepatic fibrosis: evidence from NHANES 1999-2018. BMC Gastroenterol 2024; 24:463. [PMID: 39695411 DOI: 10.1186/s12876-024-03557-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 12/10/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Several studies have demonstrated that systemic immune inflammation index (SII) has a positive relationship with hepatic steatosis. However, it is lack of system evidence for the correlation between SII and hepatic fibrosis. The objective of this study was to evaluate the relationships between SII and hepatic steatosis or hepatic fibrosis. METHODS A cross-sectional analysis was performed from the National Health and Nutrition Examination Survey (NHANES). Fibrosis-4 index (FIB-4), NAFLD fibrosis score (NFS) and hepamet fibrosis score (HFS) were the indicators for hepatic fibrosis; fatty liver index (FLI), NAFLD liver fat score (LFS) and Framingham steatosis index (FSI) were the indicators for hepatic steatosis. Pearson's test, generalized linear model (GLM) and restricted cubic splines (RCS) were used to analyze associations of SII with hepatic fibrosis and hepatic steatosis. RESULTS A total of 21,833 participants were enrolled in the study. Pearson's test and GLM revealed that there were negative relationships between SII and hepatic fibrosis (FIB-4, NFS and HFS), while positive relationships between SII and hepatic steatosis (FLI, LFS and FSI). The corresponding β (95%CI) of SII and hepatic fibrosis were - 0.35(-0.46, -0.24), -0.67(-0.71, -0.63) and - 0.10(-0.12, -0.09), respectively. The corresponding β (95%CI) of SII and hepatic steatosis were 6.12(4.75, 7.50), 0.22(0.12, 0.31) and 0.27(0.20, 0.34), respectively. Statistically significant non-linear association were found in SII with hepatic fibrosis and hepatic steatosis in RCS model (all P < 0.001). CONCLUSION There was a negative significant association between SII and hepatic fibrosis, while a positive significant association between SII and hepatic steatosis.
Collapse
Affiliation(s)
- Shuyin Duan
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Zhanwen Tu
- Suzhou Medical College of Soochow University, 199 Renai Road, Suzhou, Jiangsu, China
| | - Lijuan Duan
- College of Medicine, Huanghe University of Science and Technology, 666 Zijingshan South Road, Zhengzhou, Henan, 450061, China
| | - Runqi Tu
- College of Medicine, Huanghe University of Science and Technology, 666 Zijingshan South Road, Zhengzhou, Henan, 450061, China.
| |
Collapse
|
6
|
Tan EY, Muthiah MD, Sanyal AJ. Metabolomics at the cutting edge of risk prediction of MASLD. Cell Rep Med 2024; 5:101853. [PMID: 39657668 PMCID: PMC11722125 DOI: 10.1016/j.xcrm.2024.101853] [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: 07/25/2024] [Revised: 10/12/2024] [Accepted: 11/14/2024] [Indexed: 12/12/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a major public health threat globally. Management of patients afflicted with MASLD and research in this domain are limited by the lack of robust well-established non-invasive biomarkers for diagnosis, prognostication, and monitoring. The circulating metabolome reflects both the systemic metabo-inflammatory milieu and changes in the liver in affected individuals. In this review we summarize the available literature on changes in the different components of the metabolome in MASLD with a focus on changes that are linked to the presence of underlying steatohepatitis, severity of disease activity, and fibrosis stage. We further summarize the existing literature around biomarker panels that are derived from interrogation of the metabolome. Their relevance to disease biology and utility in practice are also discussed. We further highlight potential direction for future studies particularly to ensure they are fit for purpose and suitable for widespread use.
Collapse
Affiliation(s)
- En Ying Tan
- Division of Gastroenterology and Hepatology, Department of Medicine, National University Health System, Singapore, Singapore.
| | - Mark D Muthiah
- Division of Gastroenterology and Hepatology, Department of Medicine, National University Health System, Singapore, Singapore; Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Arun J Sanyal
- Stravitz-Sanyal Institute for Liver Disease and Metabolic Health, Virginia Commonwealth University School of Medicine, Richmond, VA, USA.
| |
Collapse
|
7
|
Choi J, Choi H, Jang Y, Paik HG, Kwon HS, Kwon J. Fermented Gold Kiwifruit Protects Mice Against Non-Alcoholic Fatty Liver Disease in a High-Fat Diet Model. APPLIED SCIENCES 2024; 14:11503. [DOI: 10.3390/app142411503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Gold kiwifruit is known for its high vitamin C content and various benefits. This study investigated the effects and molecular mechanisms of fermented gold kiwifruit (FGK) in a mouse model of high-fat diet (HFD)-induced obesity and hepatic steatosis. FGK powder was prepared using five strains of lactic acid bacteria: L. paracasei, Lc. lactis, L. acidophilus, L. casei, and L. helveticus. ICR mice were fed an HFD for 8 weeks to induce obesity and hepatic steatosis, and FGK supplementation was evaluated for its therapeutic potential. FGK administration significantly reduced serum levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), total cholesterol, triglyceride, and glucose compared to the HFD-only group. Histopathological analysis showed that FGK reduced lipid accumulation and hepatic lesions, as confirmed by hematoxylin and eosin (H&E) staining. Furthermore, administration of FGK activated the sirtuin 1(SIRT1)/adenosine monophosphate-activated protein kinase (AMPK) pathway and inhibited expression of the pro-inflammatory cytokines such as IL-1β, IL-6, and TNF-α in liver tissue. These findings suggest that FGK could reduce the severity of non-alcoholic fatty liver disease (NAFLD) by inhibiting fat synthesis, promoting fat breakdown, and suppressing inflammation in HFD-induced obese mice.
Collapse
Affiliation(s)
- Jihye Choi
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Jeonbuk National University, Iksan-si 54596, Jeollabuk-do, Republic of Korea
| | - Hwal Choi
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Jeonbuk National University, Iksan-si 54596, Jeollabuk-do, Republic of Korea
| | - Yuseong Jang
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Jeonbuk National University, Iksan-si 54596, Jeollabuk-do, Republic of Korea
| | - Hyeon-Gi Paik
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Jeonbuk National University, Iksan-si 54596, Jeollabuk-do, Republic of Korea
| | - Hyuck-Se Kwon
- R&D Team, Food & Supplement Health Claims, Vitech, #602 Giyeon B/D 141 Anjeon-ro, Iseo-myeon, Wanju-gun 55365, Jeollabuk-do, Republic of Korea
| | - Jungkee Kwon
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Jeonbuk National University, Iksan-si 54596, Jeollabuk-do, Republic of Korea
| |
Collapse
|
8
|
Zhang F, Han Y, Wu Y, Bao Z, Zheng G, Liu J, Li W. Association between triglyceride glucose-body mass index and the staging of non-alcoholic steatohepatitis and fibrosis in patients with non-alcoholic fatty liver disease. Ann Med 2024; 56:2409342. [PMID: 39348274 PMCID: PMC11443541 DOI: 10.1080/07853890.2024.2409342] [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: 04/30/2024] [Revised: 07/21/2024] [Accepted: 08/08/2024] [Indexed: 10/02/2024] Open
Abstract
OBJECTIVE The objective of this study was to thoroughly investigate the clinical value of triglyceride glucose-body mass index (TyG-BMI) in patients diagnosed with non-alcoholic fatty liver disease (NAFLD). Specifically, we aimed to determine its association with non-alcoholic steatohepatitis (NASH) and the progression of liver fibrosis. METHODS The study included 393 patients diagnosed with NAFLD after liver biopsy. The patients were divided into two distinct cohorts: a training cohort (N = 320) and a validation cohort (N = 73). The training cohort was further divided into four groups based on TyG-BMI quartiles. The clinical characteristics of the patients in each group were compared in detail, and the association between TyG-BMI and NASH, NAFLD Activity Score (NAS) ≥ 4, at-risk NASH, significant fibrosis, advanced fibrosis, and cirrhosis was analyzed using multiple models. Additionally, we generated receiver operating characteristic (ROC) curves to evaluate the predictive ability of TyG-BMI for NASH and fibrosis staging in patients with NAFLD. RESULTS Patients with higher TyG-BMI values had a significantly higher prevalence of NASH, NAS ≥ 4, at-risk NASH, significant fibrosis, advanced fibrosis, and cirrhosis (all p < .05). TyG-BMI was an independent predictor of these diseases in both unadjusted and adjusted models (all p < .05). ROC curve analysis further revealed the excellent performance of TyG-BMI in predicting NASH, NAS ≥ 4, at-risk NASH, significant fibrosis, advanced fibrosis, and cirrhosis. The validation cohort yielded analogous results. Furthermore, we constructed three multivariate models of TyG-BMI in conjunction with elastography metrics, which demonstrated elevated diagnostic AUC values of 0.782, 0.792, 0.794, 0.785, 0.834, and 0.845, respectively. CONCLUSION This study confirms a significant association between insulin resistance and NAFLD, including at-risk NASH and fibrosis staging, as assessed using the TyG-BMI index. TyG-BMI and its associated multivariate models can be valuable noninvasive indicators for NAFLD diagnosis, risk stratification, and disease course monitoring.
Collapse
Affiliation(s)
- Fan Zhang
- Department of Endocrinology, Changzhou Third People's Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, China
- Department of Clinical Nutrition, Changzhou Third People's Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, China
| | - Yan Han
- Department of Endocrinology, Changzhou Third People's Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, China
- Department of Clinical Nutrition, Changzhou Third People's Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, China
| | - Yunfei Wu
- Department of Pathology, Changzhou Third People's Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, China
| | - Zuowei Bao
- Department of Ultrasonography, Changzhou Third People's Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, China
| | - Guojun Zheng
- Clinical Laboratory, Changzhou Third People's Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, China
| | - Jianhong Liu
- Department of Pathology, Changzhou Third People's Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, China
| | - Wenjian Li
- Department of Urology, Changzhou Third People's Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, China
| |
Collapse
|
9
|
Naoumov NV, Kleiner DE, Chng E, Brees D, Saravanan C, Ren Y, Tai D, Sanyal AJ. Digital quantitation of bridging fibrosis and septa reveals changes in natural history and treatment not seen with conventional histology. Liver Int 2024; 44:3214-3228. [PMID: 39248039 PMCID: PMC11586893 DOI: 10.1111/liv.16092] [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: 03/26/2024] [Revised: 07/31/2024] [Accepted: 08/22/2024] [Indexed: 09/10/2024]
Abstract
BACKGROUND AND AIMS Metabolic dysfunction-associated steatohepatitis (MASH) with bridging fibrosis is a critical stage in the evolution of fatty liver disease. Second harmonic generation/two-photon excitation fluorescence (SHG/TPEF) microscopy with artificial intelligence (AI) provides sensitive and reproducible quantitation of liver fibrosis. This methodology was applied to gain an in-depth understanding of intra-stage fibrosis changes and septa analyses in a homogenous, well-characterised group with MASH F3 fibrosis. METHODS Paired liver biopsies (baseline [BL] and end of treatment [EOT]) of 57 patients (placebo, n = 17 and tropifexor n = 40), with F3 fibrosis stage at BL according to the clinical research network (CRN) scoring, were included. Unstained sections were examined using SHG/TPEF microscopy with AI. Changes in liver fibrosis overall and in five areas of liver lobules were quantitatively assessed by qFibrosis. Progressive, regressive septa, and 12 septa parameters were quantitatively analysed. RESULTS qFibrosis demonstrated fibrosis progression or regression in 14/17 (82%) patients receiving placebo, while the CRN scoring categorised 11/17 (65%) as 'no change'. Radar maps with qFibrosis readouts visualised quantitative fibrosis dynamics in different areas of liver lobules even in cases categorised as 'No Change'. Measurement of septa parameters objectively differentiated regressive and progressive septa (p < .001). Quantitative changes in individual septa parameters (BL to EOT) were observed both in the 'no change' and the 'regression' subgroups, as defined by the CRN scoring. CONCLUSION SHG/TPEF microscopy with AI provides greater granularity and precision in assessing fibrosis dynamics in patients with bridging fibrosis, thus advancing knowledge development of fibrosis evolution in natural history and in clinical trials.
Collapse
Affiliation(s)
| | - David E. Kleiner
- Laboratory of Pathology, Post‐Mortem SectionNational Cancer InstituteBethesdaMarylandUSA
| | | | | | | | - Yayun Ren
- Histoindex Pte. Ltd.SingaporeSingapore
| | - Dean Tai
- Histoindex Pte. Ltd.SingaporeSingapore
| | - Arun J. Sanyal
- Stravitz‐Sanyal Institute of Liver Disease and Metabolic HealthVirginia Commonwealth University School of MedicineRichmondVirginiaUSA
| |
Collapse
|
10
|
Ranneh Y, Bedir AS, Abu-Elsaoud AM, Al Raish S. Polyphenol Intervention Ameliorates Non-Alcoholic Fatty Liver Disease: An Updated Comprehensive Systematic Review. Nutrients 2024; 16:4150. [PMID: 39683546 DOI: 10.3390/nu16234150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 11/22/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) has recently emerged as a challenging metabolic disorder with a strong emphasis on its prevention and management. Polyphenols, a group of naturally occurring plant compounds, have been associated with a decreased risk of various metabolic disorders related to NAFLD. The current systematic review aims to critically assess evidence about the ameliorative effect of polyphenol supplementation on NAFLD patients. A PRISMA systematic search appraisal was conducted in PubMed, Scopus, Web of Science Core Collection, and all relevant studies published prior to April 2024 and met the inclusion criteria were included. Twenty-nine randomized clinical trials (RCTs) comprised 1840 NAFLD patients. The studies primarily examined eleven phenolic compounds, including turmeric, curcumin, resveratrol, genistein, catechin, green tea extract, hesperidin, and silymarin. Turmeric and curcumin decreased liver enzymes, inflammatory cytokines, lipid profile, insulin resistance, and NAFLD score, while resveratrol did not present consistent results across all the studies. Most studies on silymarin showed a reduction in liver enzymes and lipid profile; however, no changes were observed in inflammatory cytokine levels. The dietary supplementation of hesperidin and naringenin or green tea extract caused improvements in liver enzyme, lipid profile, and inflammatory cytokine, while genistein supplementation did not modulate blood lipid profile. In conclusion, dietary supplementation of polyphenols could potentially prevent and ameliorate NAFLD. Still, the inconsistent results across the included RCTs require further clinical research to establish optimal dosage and duration.
Collapse
Affiliation(s)
- Yazan Ranneh
- Department of Nutrition and Dietetics, College of Pharmacy, Al-Ain University, Al-Ain P.O. Box 64141, United Arab Emirates
| | - Alaa S Bedir
- Department of Nutrition, College of Medicine and Health Science, United Arab Emirates University, Al Ain 15551, United Arab Emirates
| | - Abdelghafar M Abu-Elsaoud
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
| | - Seham Al Raish
- Department of Biology, College of Science, United Arab Emirates University, Al-Ain 15551, United Arab Emirates
| |
Collapse
|
11
|
Sang H, Lim J, Kim HI. Association of Comorbidity Duration with the Occurrence and Prognosis of Steatotic Liver Disease. Dig Dis Sci 2024:10.1007/s10620-024-08723-z. [PMID: 39614023 DOI: 10.1007/s10620-024-08723-z] [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: 09/02/2024] [Accepted: 10/26/2024] [Indexed: 12/01/2024]
Abstract
BACKGROUND AND AIMS Steatotic liver disease (SLD) interacts with various comorbidities, impacting outcomes, yet little is known about the duration of comorbidities in SLD occurrence and mortality. We investigated this relationship, focusing on disease predictors and mortality rates. APPROACH Analyzing 2010 and 2015 Korea National Health and Nutrition Examination Survey data for patients aged ≥ 20, we categorized ten comorbidities (hypertension [HTN], diabetes mellitus [DM], dyslipidemia, stroke, myocardial infarction [MI], angina pectoris, asthma, chronic obstructive lung disease [COPD], chronic kidney disease [CKD], and depression) by duration. Association rule mining and logistic regression analyzed the association between SLD occurrence and comorbidity duration, while Cox regression assessed survival. RESULTS The analysis included 2,757 SLD and 9,505 non-SLD cases. Association rule mining showed that the shorter duration of DM and dyslipidemia and the longer duration of HTN comprised the top-ranked component for presence of SLD. DM with a duration ≤ 1 year showed higher risk of SLD than longer periods (odds ratio, 11.53), while the duration of cardiovascular disease, lung disease, or CKD was not significantly associated with the presence of SLD. In terms of prognosis, multivariate Cox regression showed that longer HTN and DM durations were significantly associated with increased hazard ratio (HR) beyond 10 years (HR, 2.22 and 2.11, respectively). Cardiovascular disease duration ≤ 5 years and lung disease duration > 5 years showed statistical significance (HR 2.49and 2.38, respectively). CONCLUSIONS Duration of comorbidities should be considered for comprehensive SLD risk stratification, for both the identification of SLD and the assessment of their prognosis after detection.
Collapse
Affiliation(s)
- Hyunji Sang
- Department of Endocrinology and Metabolism, Kyung Hee University Medical Center, Kyung Hee University College of Medicine, Seoul, South Korea
- Center for Digital Health, Medical Science Research Institute, Kyung Hee University Medical Center, Kyung Hee University College of Medicine, Seoul, South Korea
| | - Jihye Lim
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Catholic University College of Medicine, Seoul, South Korea
| | - Ha Il Kim
- Department of Gastroenterology and Hepatology, Hanyang University Guri Hospital, Guri, South Korea.
| |
Collapse
|
12
|
Yan Y, Gan D, Zhang P, Zou H, Li M. A machine learning-based predictive model discriminates nonalcoholic steatohepatitis from nonalcoholic fatty liver disease. Heliyon 2024; 10:e38848. [PMID: 39512464 PMCID: PMC11539579 DOI: 10.1016/j.heliyon.2024.e38848] [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: 03/14/2024] [Revised: 08/31/2024] [Accepted: 10/01/2024] [Indexed: 11/15/2024] Open
Abstract
Background Non-alcoholic fatty liver disease (NAFLD) is a leading cause of liver-related morbidity and mortality. The diagnosis of non-alcoholic steatohepatitis (NASH) plays a crucial role in the management of NAFLD patients. Objective The aim of our observational study was to build a machine learning model to identify NASH in NAFLD patients. Methods The clinical characteristics of 259 NAFLD patients and their initial laboratory data (Cohort 1) were collected to train the model and carry out internal validation. We compared the models built by five machine learning algorithms and screened out the best models. Receiver operating characteristic (ROC) curves, sensitivity, specificity, and accuracy were used to evaluate the performance of the model. In addition, the NAFLD patients in Cohort 2 (n = 181) were externally verified. Results We finally identified six independent risk factors for predicting NASH, including neutrophil percentage (NEU%), aspartate aminotransferase/alanine aminotransferase (AST/ALT), hematocrit (HCT), creatinine (CREA), uric acid (UA), and prealbumin (PA). The NASH-XGB6 model built using the XGBoost algorithm showed sufficient prediction accuracy, with ROC values of 0.95 (95 % CI, 0.91-0.98) and 0.90 (95 % CI, 0.88-0.93) in Cohort 1 and Cohort 2, respectively. Conclusions NASH-XGB6 can serve as an effective tool for distinguishing NASH patients from NAFLD patients.
Collapse
Affiliation(s)
- Yuqi Yan
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Danhui Gan
- Department of Clinical Pathology, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| | - Ping Zhang
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Haizhu Zou
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - MinMin Li
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| |
Collapse
|
13
|
Yang Y, Li S, An Z, Li S. The correlation between non-high-density lipoprotein cholesterol to high-density lipoprotein cholesterol ratio (NHHR) with non-alcoholic fatty liver disease: an analysis of the population-based NHANES (2017-2018). Front Med (Lausanne) 2024; 11:1477820. [PMID: 39582979 PMCID: PMC11581862 DOI: 10.3389/fmed.2024.1477820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 10/29/2024] [Indexed: 11/26/2024] Open
Abstract
Background/objective Non-alcoholic fatty liver disease (NAFLD) encompasses a spectrum of liver disorders, from benign steatosis to more severe conditions like non-alcoholic steatohepatitis, with risks of progressing to fibrosis, cirrhosis, and hepatocellular carcinoma. The non-high-density lipoprotein cholesterol to high-density lipoprotein cholesterol ratio (NHHR) indicates lipid metabolic dysregulation and is associated with increased risks of various diseases. This study examines the relationship between NHHR and NAFLD to evaluate NHHR as a potential predictive biomarker for NAFLD. Methods Data from the 2017-2018 National Health and Nutrition Examination Survey (NHANES) were used for cross-sectional analysis. After excluding individuals with incomplete data, hepatitis infections, heavy alcohol use, and those under 18, the study included 2,757 adults. The relationship between NHHR and NAFLD was analyzed using multivariable logistic regression, including subgroup analysis and interaction testing. Results Among the 2,757 participants (mean age 49.91 years), 44.9% had NAFLD. NHHR showed a significant positive association with NAFLD, with an unadjusted odds ratio (OR) of 1.71 and a fully adjusted OR of 1.45. Quartile analysis revealed a 228% higher prevalence of NAFLD in the highest NHHR quartile, with an OR of 3.28. This positive association was consistent across various subgroups. Conclusion Our findings suggest that elevated NHHR is positively correlated with the prevalence of NAFLD and possesses predictive value. We recommend that future research validate the clinical utility of NHHR, particularly for early detection of high-risk individuals and guiding personalized interventions.
Collapse
Affiliation(s)
- Yuhao Yang
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| | - Shengxi Li
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Zhenmei An
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China
| | - Shuangqing Li
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
14
|
Smith EE, Biessels GJ, Gao V, Gottesman RF, Liesz A, Parikh NS, Iadecola C. Systemic determinants of brain health in ageing. Nat Rev Neurol 2024; 20:647-659. [PMID: 39375564 DOI: 10.1038/s41582-024-01016-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/20/2024] [Indexed: 10/09/2024]
Abstract
Preservation of brain health is a worldwide priority. The traditional view is that the major threats to the ageing brain lie within the brain itself. Consequently, therapeutic approaches have focused on protecting the brain from these presumably intrinsic pathogenic processes. However, an increasing body of evidence has unveiled a previously under-recognized contribution of peripheral organs to brain dysfunction and damage. Thus, in addition to the well-known impact of diseases of the heart and endocrine glands on the brain, accumulating data suggest that dysfunction of other organs, such as gut, liver, kidney and lung, substantially affects the development and clinical manifestation of age-related brain pathologies. In this Review, a framework is provided to indicate how organ dysfunction can alter brain homeostasis and promote neurodegeneration, with a focus on dementia. We delineate the associations of subclinical dysfunction in specific organs with dementia risk and provide suggestions for public health promotion and clinical management.
Collapse
Affiliation(s)
- Eric E Smith
- Department of Clinical Neurosciences and Hotchkiss Brain Institute, University of Calgary, Calgary, Canada.
| | - Geert Jan Biessels
- Department of Neurology, UMC Utrecht Brain Center, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Virginia Gao
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | | | - Arthur Liesz
- Institute for Stroke and Dementia Research, University Medical Center Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Neal S Parikh
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
15
|
Li J, Yan N, Li X, He S. Association between serum vitamin D concentration and liver fibrosis in diabetes mellitus patients: a cross-sectional study from the NHANES database. Acta Diabetol 2024; 61:1393-1402. [PMID: 38831202 DOI: 10.1007/s00592-024-02292-3] [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: 01/05/2024] [Accepted: 04/14/2024] [Indexed: 06/05/2024]
Abstract
AIM Liver fibrosis (LF) is a common complication of diabetes mellitus (DM). Studies have found that vitamin D (VD), as a modifiable factor has been reported to be associated with LF. The relationship between serum VD concentration and LF in DM patients has rarely been reported. The aim of this study was to assess the association between serum VD concentration and LF in DM patients. METHODS In this cross-sectional study, data of DM patients aged ≥ 45 years were extracted from the National Health and Nutrition Examination Survey (NHANES 2017-2018). Serum VD concentration was measured by high performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS). Vibration controlled transient elastography (VCTE) was used to measure liver stiffness. Covariates included sociodemographic information, lifestyles, laboratory data, diseases history were extracted from the database. The weighted univariable and multivariable logistic regression models were utilized to explore the association between serum VD concentration and LF in DM patients, and were described as odds ratio (ORs) and 95% confidence intervals (CIs). Subgroup analyses based on BMI, liver steatosis, hypertension and dyslipidemia were further assessed the association. RESULTS A total of 799 patients were included, of which 188 (23.53%) had LF. Higher serum VD concentration was associated with the lower odds of LF (OR = 0.33, 95% CI 0.19-0.59) and advanced LF (OR = 0.31, 95% CI 0.17-0.55) in DM patients after adjustment for race, liver steatosis, BMI, smoking, drinking, AST, ALT and physical activity, especially in patients with liver steatosis (OR = 0.28, 95% CI 0.13-0.59) and dyslipidemia (OR = 0.31, 95% CI 0.14-0.66), respectively. CONCLUSIONS High serum VD concentration may have a potential benefit for maintain the liver health in DM patients.
Collapse
Affiliation(s)
- Jing Li
- Department of Endocrinology Diabetes, Shaanxi Provincial People's Hospital, No. 256 Youyi West Road, Beilin District, Xi'an, 710068, China
| | - Ni Yan
- Department of Endocrinology Diabetes, Shaanxi Provincial People's Hospital, No. 256 Youyi West Road, Beilin District, Xi'an, 710068, China
| | - Xiaofeng Li
- Department of Endocrinology Diabetes, Shaanxi Provincial People's Hospital, No. 256 Youyi West Road, Beilin District, Xi'an, 710068, China
| | - Shenglin He
- Department of Endocrinology Diabetes, Shaanxi Provincial People's Hospital, No. 256 Youyi West Road, Beilin District, Xi'an, 710068, China.
| |
Collapse
|
16
|
Link F, Li Y, Zhao J, Munker S, Fan W, Nwosu ZC, Yao Y, Wang S, Huang C, Liebe R, Hammad S, Liu H, Shao C, Gao C, Sun B, Török NJ, Ding H, Ebert MP, Weng H, Ten Dijke P, Drasdo D, Dooley S, Wang S. ECM1 attenuates hepatic fibrosis by interfering with mediators of latent TGF-β1 activation. Gut 2024:gutjnl-2024-333213. [PMID: 39448254 DOI: 10.1136/gutjnl-2024-333213] [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: 06/25/2024] [Accepted: 07/24/2024] [Indexed: 10/26/2024]
Abstract
OBJECTIVE Extracellular matrix protein 1 (ECM1) serves as a gatekeeper of hepatic fibrosis by maintaining transforming growth factor-β1 (TGF-β1) in its latent form. ECM1 knockout (KO) causes latent (L) TGF-β1 activation, resulting in hepatic fibrosis with rapid mortality. In chronic liver disease (CLD), ECM1 decreases with increasing CLD severity. We investigate the regulatory role of ECM1 in TGF-β1 bioavailability and its impact on CLD progression. DESIGN RNAseq was performed to analyse hepatic gene expression. Functional assays were performed using hepatic stellate cells (HSCs), Ecm1-KO and Fxr-KO mice, patient liver tissue and computer simulations. RESULTS Expression of LTGF-β1 activators, including thrombospondins (TSPs), ADAMTS proteases and matrix metalloproteinases (MMPs), increased along with profibrotic gene expression in liver tissue of Ecm1-KO mice. In HSCs, overexpression of ECM1 prevented LTGF-β1 activation mediated by TSP-1, ADAMTS1, and MMP-2/9. In vitro interaction assays demonstrated that ECM1 inhibited LTGF-β1 activation by interacting with TSP-1 and ADAMTS1 via their respective, intrinsic KRFK or KTFR amino acid sequences and by suppressing MMP-2/9 proteolytic activity. In mice, ECM1 overexpression attenuated KRFK-induced LTGF-β1 activation while KTFR treatment reversed Ecm1-KO-mediated and Fxr-KO-mediated liver injury. In patients with CLD, ECM1 expression was inversely correlated with TSP-1, ADAMTS1, MMP-2/9 expression and LTGF-β1 activation. And, these results were complemented by a computational compartment model representing the key network of cellular phenotypes and predicted interactions in liver fibrogenesis. CONCLUSION Our findings underscore the hepatoprotective effect of ECM1, which interferes with mediators of LTGF-β1 activation, suggesting ECM1 or its representative peptide as potential antifibrotic therapies in CLD.
Collapse
Affiliation(s)
- Frederik Link
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Yujia Li
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Jieling Zhao
- INRIA de Saclay, Palaiseau, France
- IfADo, Dortmund, Germany
| | - Stefan Munker
- Department of Medicine II, University Hospital, LMU, Munich, Germany
- Liver Center Munich, University Hospital, LMU, Munich, Germany
| | - Weiguo Fan
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Zeribe C Nwosu
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | - Ye Yao
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Shanshan Wang
- Beijing Institute of Hepatology, Beijing You'an Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Chenjun Huang
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Department of Clinical Laboratory Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Roman Liebe
- Clinic of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke-University, Magdeburg, Germany
| | - Seddik Hammad
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Hui Liu
- Department of Pathology, Beijing You'an Hospital, Affiliated with Capital Medical University, Beijing, People's Republic of China
| | - Chen Shao
- Department of Pathology, Beijing You'an Hospital, Affiliated with Capital Medical University, Beijing, People's Republic of China
| | - Chunfang Gao
- Department of Clinical Laboratory Medicine Center, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Bing Sun
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Natalie J Török
- Gastroenterology and Hepatology, Stanford University, Palo Alto, CA, USA
| | - Huiguo Ding
- Department of Gastroenterology and Hepatology, Beijing You'an Hospital, Affiliated with Capital Medical University, Beijing, People's Republic of China
| | - Matthias Pa Ebert
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- DKFZ-Hector Cancer Institute at the University Medical Center, Mannheim, Germany
| | - Honglei Weng
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Peter Ten Dijke
- Oncode Institute and Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Dirk Drasdo
- INRIA de Saclay, Palaiseau, France
- IfADo, Dortmund, Germany
| | - Steven Dooley
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Sai Wang
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
17
|
Tao SH, Lei YQ, Tan YM, Yang YB, Xie WN. Chinese herbal formula in the treatment of metabolic dysfunction-associated steatotic liver disease: current evidence and practice. Front Med (Lausanne) 2024; 11:1476419. [PMID: 39440040 PMCID: PMC11493624 DOI: 10.3389/fmed.2024.1476419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 09/24/2024] [Indexed: 10/25/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), formerly known as nonalcoholic fatty liver disease, continues to rise with rapid economic development and poses significant challenges to human health. No effective drugs are clinically approved. MASLD is regarded as a multifaceted pathological process encompassing aberrant lipid metabolism, insulin resistance, inflammation, gut microbiota imbalance, apoptosis, fibrosis, and cirrhosis. In recent decades, herbal medicines have gained increasing attention as potential therapeutic agents for the prevention and treatment of MASLD, due to their good tolerance, high efficacy, and low toxicity. In this review, we summarize the pathological mechanisms of MASLD; emphasis is placed on the anti-MASLD mechanisms of Chinese herbal formula (CHF), especially their effects on improving lipid metabolism, inflammation, intestinal flora, and fibrosis. Our goal is to better understand the pharmacological mechanisms of CHF to inform research on the development of new drugs for the treatment of MASLD.
Collapse
Affiliation(s)
- Shao-Hong Tao
- Affiliated Guangdong Hospital of Integrated Traditional Chinese and Western Medicine of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
| | - Yu-Qing Lei
- Affiliated Guangdong Hospital of Integrated Traditional Chinese and Western Medicine of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
| | - Yi-Mei Tan
- Affiliated Guangdong Hospital of Integrated Traditional Chinese and Western Medicine of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Foshan, Guangdong, China
| | - Yu-Bo Yang
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Wei-Ning Xie
- Department of Scientific Research, Guangdong Provincial Hospital of Integrated Traditional Chinese and Western Medicine, Foshan, Guangdong, China
| |
Collapse
|
18
|
Chen Y, Zhang Y, Jin X, Hong S, Tian H. Exerkines: Benign adaptation for exercise and benefits for non-alcoholic fatty liver disease. Biochem Biophys Res Commun 2024; 726:150305. [PMID: 38917635 DOI: 10.1016/j.bbrc.2024.150305] [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: 04/09/2024] [Revised: 06/11/2024] [Accepted: 06/20/2024] [Indexed: 06/27/2024]
Abstract
Exercise has multiple beneficial effects on human metabolic health and is regarded as a "polypill" for various diseases. At present, the lack of physical activity usually causes an epidemic of chronic metabolic syndromes, including obesity, cardiovascular diseases, and non-alcoholic fatty liver disease (NAFLD). Remarkably, NAFLD is emerging as a serious public health issue and is associated with the development of cirrhosis and hepatocellular carcinoma. Unfortunately, specific drug therapies for NAFLD and its more severe form, non-alcoholic steatohepatitis (NASH), are currently unavailable. Lifestyle modification is the foundation of treatment recommendations for NAFLD and NASH, especially for exercise. There are under-appreciated organs that crosstalk to the liver during exercise such as muscle-liver crosstalk. Previous studies have reported that certain exerkines, such as FGF21, GDF15, irisin, and adiponectin, are beneficial for liver metabolism and have the potential to be targeted for NAFLD treatment. In addition, some of exerkines can be modified for the new proteins and get enhanced functions, like IL-6/IC7Fc. Another importance of exercise is the physiological adaptation that combats metabolic diseases. Thus, this review aims to summarize the known exerkines and utilize a multi-omics mining tool to identify more exerkines for the future research. Overall, understanding the mechanisms by which exercise-induced exerkines exert their beneficial effects on metabolic health holds promise for the development of novel therapeutic strategies for NAFLD and related diseases.
Collapse
Affiliation(s)
- Yang Chen
- School of Exercise and Health, Shanghai University of Sport, Shanghai, 200438, China
| | - Yan Zhang
- Clinical Laboratory, Suzhou Yong Ding Hospital, Suzhou, 215200, China
| | - Xingsheng Jin
- School of Exercise and Health, Shanghai University of Sport, Shanghai, 200438, China
| | - Shangyu Hong
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, 200032, China.
| | - Haili Tian
- School of Exercise and Health, Shanghai University of Sport, Shanghai, 200438, China.
| |
Collapse
|
19
|
Zhang J, Chen W, Song K, Song K, Kolls J, Wu T. YAP activation in liver macrophages via depletion of MST1/MST2 enhances liver inflammation and fibrosis in MASLD. FASEB J 2024; 38:e70026. [PMID: 39215627 PMCID: PMC11697422 DOI: 10.1096/fj.202400813rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 08/13/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
Macrophages have been recognized as pivotal players in the progression of MASLD/MASH. However, the molecular mechanisms underlying their multifaceted functions in the disease remain to be further clarified. In the current study, we developed a new mouse model with YAP activation in macrophages to delineate the effect and mechanism of YAP signaling in the pathogenesis of MASLD/MASH. Genetically modified mice, featuring specific depletion of both Mst1 and Mst2 in macrophages/monocytes, were generated and exposed to a high-fat diet for 12 weeks to induce MASLD. Following this period, livers were collected for histopathological examination, and liver non-parenchymal cells were isolated and subjected to various analyses, including single-cell RNA-sequencing, immunofluorescence and immunoblotting and qRT-PCR to investigate the impact of YAP signaling on the progression of MASLD. Our data revealed that Mst1/2 depletion in liver macrophages enhanced liver inflammation and fibrosis in MASLD. Using single-cell RNA-sequencing, we showed that YAP activation via Mst1/2 depletion upregulated the expressions of both pro-inflammatory genes and genes associated with resolution/tissue repair. We observed that YAP activation increases Kupffer cell populations (i.e., Kupffer-2 and Kupffer-3) which are importantly implicated in the pathogenesis of MASLD/MASH. Our data indicate that YAP activation via Mst1/2 deletion enhances both the pro-inflammatory and tissue repairing functions of Kupffer-1 and -2 cells at least in part through C1q. These YAP-regulatory mechanisms control the plasticity of liver macrophages in the context of MASLD/MASH. Our findings provide important evidence supporting the critical regulatory role of YAP signaling in liver macrophage plasticity and the progression of MASLD. Therefore, targeting the Hippo-YAP pathway may present a promising therapeutic strategy for the treatment of MASH.
Collapse
Affiliation(s)
- Jinqiang Zhang
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Weina Chen
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Kyoungsub Song
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Kejing Song
- Tulane Center for Translational Research in Infection & Inflammation, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Jay Kolls
- Tulane Center for Translational Research in Infection & Inflammation, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Tong Wu
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
| |
Collapse
|
20
|
Li S, Hao L, Yu F, Li N, Deng J, Zhang J, Xiong S, Hu X. Capsaicin: a spicy way in liver disease. Front Pharmacol 2024; 15:1451084. [PMID: 39281271 PMCID: PMC11392895 DOI: 10.3389/fphar.2024.1451084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 08/22/2024] [Indexed: 09/18/2024] Open
Abstract
The incidence of liver disease continues to rise, encompassing a spectrum from simple steatosis or non-alcoholic fatty liver disease (NAFLD) to non-alcoholic steatohepatitis (NASH), cirrhosis and liver cancer. Dietary habits in individuals with liver disease may significantly impact the treatment and prevention of these conditions. This article examines the role of chili peppers, a common dietary component, in this context, focusing on capsaicin, the active ingredient in chili peppers. Capsaicin is an agonist of the transient receptor potential vanilloid subfamily 1 (TRPV1) and has been shown to exert protective effects on liver diseases, including liver injury, NAFLD, liver fibrosis and liver cancer. These protective effects are attributed to capsaicin's anti-oxidant, anti-inflammatory, anti-steatosis and anti-fibrosis effects. This article reviewed the different molecular mechanisms of the protective effect of capsaicin on liver diseases.
Collapse
Affiliation(s)
- Shenghao Li
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Liyuan Hao
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fei Yu
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Na Li
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiali Deng
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Junli Zhang
- Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Shuai Xiong
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaoyu Hu
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
21
|
Fernández-Tussy P, Cardelo MP, Zhang H, Sun J, Price NL, Boutagy NE, Goedeke L, Cadena-Sandoval M, Xirouchaki CE, Brown W, Yang X, Pastor-Rojo O, Haeusler RA, Bennett AM, Tiganis T, Suárez Y, Fernández-Hernando C. miR-33 deletion in hepatocytes attenuates MASLD-MASH-HCC progression. JCI Insight 2024; 9:e168476. [PMID: 39190492 PMCID: PMC11466198 DOI: 10.1172/jci.insight.168476] [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: 01/04/2023] [Accepted: 08/21/2024] [Indexed: 08/29/2024] Open
Abstract
The complexity of the mechanisms underlying metabolic dysfunction-associated steatotic liver disease (MASLD) progression remains a significant challenge for the development of effective therapeutics. miRNAs have shown great promise as regulators of biological processes and as therapeutic targets for complex diseases. Here, we study the role of hepatic miR-33, an important regulator of lipid metabolism, during the progression of MASLD and the development of hepatocellular carcinoma (HCC). We report that miR-33 was elevated in the livers of humans and mice with MASLD and that its deletion in hepatocytes (miR-33 HKO) improved multiple aspects of the disease, including steatosis and inflammation, limiting the progression to metabolic dysfunction-associated steatotic hepatitis (MASH), fibrosis, and HCC. Mechanistically, hepatic miR-33 deletion reduced lipid synthesis and promoted mitochondrial fatty acid oxidation, reducing lipid burden. Additionally, absence of miR-33 altered the expression of several known miR-33 target genes involved in metabolism and resulted in improved mitochondrial function and reduced oxidative stress. The reduction in lipid accumulation and liver injury resulted in decreased YAP/TAZ pathway activation, which may be involved in the reduced HCC progression in HKO livers. Together, these results suggest suppressing hepatic miR-33 may be an effective therapeutic approach to temper the development of MASLD, MASH, and HCC in obesity.
Collapse
Affiliation(s)
- Pablo Fernández-Tussy
- Vascular Biology and Therapeutics Program
- Department of Comparative Medicine
- Yale Center for Molecular and System Metabolism, and
| | - Magdalena P. Cardelo
- Vascular Biology and Therapeutics Program
- Department of Comparative Medicine
- Yale Center for Molecular and System Metabolism, and
| | - Hanming Zhang
- Vascular Biology and Therapeutics Program
- Department of Comparative Medicine
- Yale Center for Molecular and System Metabolism, and
| | - Jonathan Sun
- Vascular Biology and Therapeutics Program
- Department of Comparative Medicine
- Yale Center for Molecular and System Metabolism, and
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Nathan L. Price
- Vascular Biology and Therapeutics Program
- Department of Comparative Medicine
- Yale Center for Molecular and System Metabolism, and
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, Maryland, USA
| | - Nabil E. Boutagy
- Vascular Biology and Therapeutics Program
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Leigh Goedeke
- Cardiovascular Research Institute and Division of Cardiology, Department of Medicine; and
- Diabetes, Obesity and Metabolism Institute and Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Martí Cadena-Sandoval
- Department of Pathology & Cell Biology and Naomi Berrie Diabetes Center, Columbia University, New York, New York, USA
| | - Chrysovalantou E. Xirouchaki
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Department of Surgery, Alfred Hospital and Monash University, Melbourne, Victoria, Australia
| | - Wendy Brown
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Xiaoyong Yang
- Department of Comparative Medicine
- Yale Center for Molecular and System Metabolism, and
- Department of Molecular and Cellular Physiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Oscar Pastor-Rojo
- Vascular Biology and Therapeutics Program
- Servicio de Bioquímica Clínica, Hospital Universitario Ramón y Cajal IRYCIS, Madrid, Spain
- Departamento de Biología de Sistemas, Universidad de Alcalá de Henares, Madrid, Spain
| | - Rebecca A. Haeusler
- Department of Pathology & Cell Biology and Naomi Berrie Diabetes Center, Columbia University, New York, New York, USA
| | - Anton M. Bennett
- Yale Center for Molecular and System Metabolism, and
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Tony Tiganis
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Department of Surgery, Alfred Hospital and Monash University, Melbourne, Victoria, Australia
| | - Yajaira Suárez
- Vascular Biology and Therapeutics Program
- Department of Comparative Medicine
- Yale Center for Molecular and System Metabolism, and
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Carlos Fernández-Hernando
- Vascular Biology and Therapeutics Program
- Department of Comparative Medicine
- Yale Center for Molecular and System Metabolism, and
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
22
|
Wutsdorff L, Mougnekabol J, Tang P, Reutzel-Selke A, Sauer IM, Haep N. Unveiling the Multifaceted Role of CIDEB: From Apoptosis to Lipid Metabolism and Liver Health. LIVERS 2024; 4:406-419. [DOI: 10.3390/livers4030030] [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] [Indexed: 01/03/2025] Open
Abstract
Cell-death-inducing DNA fragmentation factor-alpha (DFFA)-like effector b (CIDEB) was first identified as an apoptosis-inducing protein. Further research revealed a pivotal role in lipid metabolism, regulating very-low-density lipoprotein (VLDL), lipid droplets (LD), sterol response element-binding protein (SREBP), and chylomicrons. Recent studies have uncovered that rare germline variants in CIDEB protect against liver diseases, including MAFLD, cirrhosis, and viral hepatitis. Furthermore, CIDEB influences steps of the hepatitis C virus (HCV) replication cycle. This review summarizes the current knowledge about CIDEB’s roles in apoptosis, lipid metabolism, and viral hepatitis, and highlights its critical role in liver diseases.
Collapse
Affiliation(s)
- Louise Wutsdorff
- Department of Surgery, CCM|CVK, Experimental Surgery, Charité—Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, 13353 Berlin, Germany
| | - Julienne Mougnekabol
- Department of Surgery, CCM|CVK, Experimental Surgery, Charité—Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, 13353 Berlin, Germany
| | - Peter Tang
- Department of Surgery, CCM|CVK, Experimental Surgery, Charité—Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, 13353 Berlin, Germany
| | - Anja Reutzel-Selke
- Department of Surgery, CCM|CVK, Experimental Surgery, Charité—Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, 13353 Berlin, Germany
| | - Igor M. Sauer
- Department of Surgery, CCM|CVK, Experimental Surgery, Charité—Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, 13353 Berlin, Germany
| | - Nils Haep
- Department of Surgery, CCM|CVK, Experimental Surgery, Charité—Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, 13353 Berlin, Germany
- Clinician Scientist Program, Berlin Institute of Health at Charité—Universitätsmedizin Berlin, BIH Academy, 10178 Berlin, Germany
| |
Collapse
|
23
|
Dong X, Xiong YT, He T, Zheng C, Li J, Zhuang Y, Xu Y, Xiu Y, Wu Z, Zhao X, Xiao X, Bai Z, Gao L. Protective effects of Nogo-B deficiency in NAFLD mice and its multiomics analysis of gut microbiology and metabolism. GENES & NUTRITION 2024; 19:17. [PMID: 39182019 PMCID: PMC11344411 DOI: 10.1186/s12263-024-00754-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 08/11/2024] [Indexed: 08/27/2024]
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) is a prevalent chronic liver ailment that can lead to serious conditions such as cirrhosis and hepatocellular carcinoma. Hepatic Nogo-B regulates glucose and lipid metabolism, and its inhibition has been shown to be protective against metabolic syndrome. Increasing evidence suggests that imbalances in the gut microbiota (GM) and lipid metabolism disorders are significant contributors to NAFLD progression. Nevertheless, it is not yet known whether Nogo-B can affect NAFLD by influencing the gut microbiota and metabolites. Hence, the aim of the present study was to characterize this process and explore its possible underlying mechanisms. METHODS A NAFLD model was constructed by administering a high-fat diet (HFD) to Nogo-B-/- and WT mice from the same litter, and body weight was measured weekly in each group. The glucose tolerance test (GTT) and insulin tolerance test (ITT) were performed to assess blood glucose levels. At the end of the 12-week period, samples of serum, liver, and intestinal contents were collected and used for serum biochemical marker and inflammatory factor detection; pathology evaluation; and gut microbiome and metabolomics analysis. Spearman's correlation analysis was performed to determine possible correlations between differential gut microbiota and differential serum metabolites between groups. RESULTS Nogo-B deficiency attenuated the effects of the HFD, including weight gain, liver weight gain, impaired glucose tolerance, hepatic steatosis, elevated serum lipid biochemicals levels, and liver function. Nogo-B deficiency suppressed M1 polarization and promoted M2 polarization, thus inhibiting inflammatory responses. Furthermore, Nogo-B-/--HFD-fed mice presented increased gut microbiota richness and diversity, decreased Firmicutes/Bacteroidota (F/B) ratios, and altered serum metabolites compared with those of WT-HFD-fed mice. During analysis, several differential gut microbiota, including Lachnoclostridium, Harryflintia, Odoribacter, UCG-009, and unclassified_f_Butyricoccaceae, were screened between groups. These microbiota were found to be positively correlated with upregulated purine metabolism and bile acid metabolites in Nogo-B deficiency, while they were negatively correlated with downregulated corticosterone and tricarboxylic acid cyclic metabolites in Nogo-B deficiency. CONCLUSION Nogo-B deficiency delayed NAFLD progression, as demonstrated by reduced hepatocellular lipid accumulation, attenuated inflammation and liver injury, and ameliorated gut microbiota dysbiosis and metabolic disorders. Importantly, Odoribacter was strongly positively correlated with ALB and taurodeoxycholic acid, suggesting that it played a considerable role in the influence of Nogo-B on the progression of NAFLD, a specific feature of NAFLD in Nogo-B-/- mice. The regulation of bile acid metabolism by the gut microbiota may be a potential target for Nogo-B deficiency to ameliorate NAFLD.
Collapse
Affiliation(s)
- Xu Dong
- Medical School of Chinese PLA, Beijing, China
- Department of Gastroenterology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
- Department of Hepatology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yu-Ting Xiong
- 307 Clinical Medical College of PLA, Anhui Medical University, Beijing, China
| | - Tingting He
- Department of Hepatology Medicine of Traditional Chinese Medicine, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Congyang Zheng
- Medical School of Chinese PLA, Beijing, China
- Department of Gastroenterology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
- Department of Hepatology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Junjie Li
- Department of Hepatology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
- School of Chengde Medical University, Chengdeshi, China
| | - Yingjie Zhuang
- Department of Gastroenterology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yingjie Xu
- Department of Hepatology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Ye Xiu
- Department of Hepatology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Zhixin Wu
- Department of Hepatology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xiaomei Zhao
- Department of Hepatology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xiaohe Xiao
- Department of Hepatology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China.
- China Military Institute of Chinese Materia, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China.
| | - Zhaofang Bai
- Department of Hepatology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China.
- China Military Institute of Chinese Materia, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China.
| | - Lili Gao
- Medical School of Chinese PLA, Beijing, China.
- Department of Gastroenterology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
24
|
Gu W, Wu G, Chen G, Meng X, Xie Z, Cai S. Polyphenols alleviate metabolic disorders: the role of ubiquitin-proteasome system. Front Nutr 2024; 11:1445080. [PMID: 39188976 PMCID: PMC11345163 DOI: 10.3389/fnut.2024.1445080] [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: 06/06/2024] [Accepted: 07/04/2024] [Indexed: 08/28/2024] Open
Abstract
Metabolic disorders include obesity, nonalcoholic fatty liver disease, insulin resistance and type 2 diabetes. It has become a major health issue around the world. Ubiquitin-proteasome system (UPS) is essential for nearly all cellular processes, functions as a primary pathway for intracellular protein degradation. Recent researches indicated that dysfunctions in the UPS may result in the accumulation of toxic proteins, lipotoxicity, oxidative stress, inflammation, and insulin resistance, all of which contribute to the development and progression of metabolic disorders. An increasing body of evidence indicates that specific dietary polyphenols ameliorate metabolic disorders by preventing lipid synthesis and transport, excessive inflammation, hyperglycemia and insulin resistance, and oxidative stress, through regulation of the UPS. This review summarized the latest research progress of natural polyphenols improving metabolic disorders by regulating lipid accumulation, inflammation, oxidative stress, and insulin resistance through the UPS. In addition, the possible mechanisms of UPS-mediated prevention of metabolic disorders are comprehensively proposed. We aim to provide new angle to the development and utilization of polyphenols in improving metabolic disorders.
Collapse
Affiliation(s)
- Wei Gu
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Sciences and Technology, Anhui Agricultural University, Hefei, Anhui, China
- Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei, Anhui, China
| | - Guohuo Wu
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Sciences and Technology, Anhui Agricultural University, Hefei, Anhui, China
- Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei, Anhui, China
| | - Guijie Chen
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Sciences and Technology, Anhui Agricultural University, Hefei, Anhui, China
- Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei, Anhui, China
| | - Xianghui Meng
- The First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui, China
| | - Zhongwen Xie
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Sciences and Technology, Anhui Agricultural University, Hefei, Anhui, China
- Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei, Anhui, China
| | - Shanbao Cai
- The First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui, China
| |
Collapse
|
25
|
Long C, Ma Q, Huang L, Lin W. A near-infrared fluorescent probe for differentiating cancer cells from normal cells and early diagnosis of liver cirrhosis. Anal Chim Acta 2024; 1316:342802. [PMID: 38969400 DOI: 10.1016/j.aca.2024.342802] [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: 04/14/2024] [Revised: 05/26/2024] [Accepted: 05/29/2024] [Indexed: 07/07/2024]
Abstract
BACKGROUND Cirrhosis represents the terminal stage of liver disease progression and timely intervention in a diseased liver can enhance the likelihood of recovery. Viscosity, a crucial parameter of the cellular microenvironment, is intricately linked to the advancement of cirrhosis. However, viscosity monitoring still faces significant challenges in achieving non-invasive and rapid early diagnosis of cirrhosis. Near-infrared (NIR) fluorescence imaging has the advantages of high sensitivity, non-destructive detection, and ignoring background fluorescence interference, plays an important role in diagnosing and treating various biological diseases. Hence, monitoring cellular viscosity changes with NIR fluorescence probe holds great significance in the early diagnosis of cirrhosis. RESULTS In this study, the NIR fluorescence probe based on the intramolecular charge transfer (TICT) mechanism was developed for imaging applications in mouse model of liver cirrhosis. A molecular rotor-type viscosity-responsive probe was synthesized by linking dioxanthracene groups via carbon-carbon double bonds. The probe demonstrated remarkable sensitivity, high selectivity and photostability, with its responsiveness to viscosity largely unaffected by factors such as polarity, pH, and interfering ions. The probe could effectively detect various drug-induced changes in cellular viscosity, enabling the differentiation between normal cells and cancerous cells. Furthermore, the enhanced tissue penetration capabilities of probe facilitated its successful application in mouse model of liver cirrhosis, allowing for the assessment of liver disease severity based on fluorescence intensity and providing a powerful tool for early diagnosis of cirrhosis. SIGNIFICANCE A NIR viscosity-sensitive fluorescent probe was specifically designed to effectively monitor alterations in cellular and organ viscosity, which could advance the understanding of the biological characteristics of cancer and provide theoretical support for the early diagnosis of cirrhosis. Overall, this probe held immense potential in monitoring viscosity-related conditions, expanding the range of biomedical tools available.
Collapse
Affiliation(s)
- Chenyuan Long
- Institute of Optical Materials and Chemical Biology, Guangxi Key Laboratory of Electrochemical Energy Materials, School of Chemistry and Chemical Engineering, Guangxi University, Nanning, Guangxi, 530004, PR China
| | - Qingqing Ma
- Institute of Optical Materials and Chemical Biology, Guangxi Key Laboratory of Electrochemical Energy Materials, School of Chemistry and Chemical Engineering, Guangxi University, Nanning, Guangxi, 530004, PR China
| | - Ling Huang
- Institute of Optical Materials and Chemical Biology, Guangxi Key Laboratory of Electrochemical Energy Materials, School of Chemistry and Chemical Engineering, Guangxi University, Nanning, Guangxi, 530004, PR China
| | - Weiying Lin
- Institute of Optical Materials and Chemical Biology, Guangxi Key Laboratory of Electrochemical Energy Materials, School of Chemistry and Chemical Engineering, Guangxi University, Nanning, Guangxi, 530004, PR China.
| |
Collapse
|
26
|
Srinivas AN, Suresh D, Vishwanath PM, Satish S, Santhekadur PK, Koka S, Kumar DP. TACE inhibition: a promising therapeutic intervention against AATF-mediated steatohepatitis to hepatocarcinogenesis. Mol Oncol 2024; 18:1940-1957. [PMID: 38558505 PMCID: PMC11306524 DOI: 10.1002/1878-0261.13646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 01/03/2024] [Accepted: 03/18/2024] [Indexed: 04/04/2024] Open
Abstract
Metabolic dysfunction-associated steatohepatitis-driven hepatocellular carcinoma (MASH-HCC) is a global clinical challenge for which there is a limited understanding of disease pathogenesis and a subsequent lack of therapeutic interventions. We previously identified that tumor necrosis factor-alpha (TNF-α) upregulated apoptosis antagonizing transcription factor (AATF) in MASH. Here, we investigated the effect of TNF-α converting enzyme (TACE) inhibition as a promising targeted therapy against AATF-mediated steatohepatitis to hepatocarcinogenesis. A preclinical murine model that recapitulates human MASH-HCC was used in the study. C57Bl/6 mice were fed with chow diet normal water (CD) or western diet sugar water (WD) along with a low dose of carbon tetrachloride (CCl4; 0.2 μL·g-1, weekly) for 24 weeks. TACE activity, TNF-α levels, and AATF expression were measured. The mice were treated with the TACE inhibitor Marimastat for 12 weeks, followed by analyses of liver injury, fibrosis, inflammation, and oncogenic signaling. In vitro experiments using stable clones of AATF control and AATF knockdown were also conducted. We found that AATF expression was upregulated in WD/CCl4 mice, which developed severe MASH at 12 weeks and advanced fibrosis with HCC at 24 weeks. WD/CCl4 mice showed increased TACE activity with reduced hepatic expression of sirtuin 1 (Sirt1) and tissue inhibitor of metalloproteinase 3 (Timp3). The involvement of the SIRT1/TIMP3/TACE axis was confirmed by the release of TNF-α, which upregulated AATF, a key molecular driver of MASH-HCC. Interestingly, TACE inhibition by Marimastat reduced liver injury, dyslipidemia, AATF expression, and oncogenic signaling, effectively preventing hepatocarcinogenesis. Furthermore, Marimastat inhibited the activation of JNK, ERK1/2, and AKT, which are key regulators of tumorigenesis in WD/CCl4 mice and in AATF control cells, but had no effect on AATF knockdown cells. This study shows that TACE inhibition prevents AATF-mediated inflammation, fibrosis, and oncogenesis in MASH-HCC, offering a potential target for therapeutic intervention.
Collapse
Affiliation(s)
- Akshatha N. Srinivas
- Department of Biochemistry, CEMR Lab, JSS Medical CollegeJSS Academy of Higher Education and ResearchMysuruIndia
| | - Diwakar Suresh
- Department of Biochemistry, CEMR Lab, JSS Medical CollegeJSS Academy of Higher Education and ResearchMysuruIndia
| | - Prashant M. Vishwanath
- Department of Biochemistry, CEMR Lab, JSS Medical CollegeJSS Academy of Higher Education and ResearchMysuruIndia
| | - Suchitha Satish
- Department of Pathology, JSS Medical College and HospitalJSS Academy of Higher Education and ResearchMysuruIndia
| | - Prasanna K. Santhekadur
- Department of Biochemistry, CEMR Lab, JSS Medical CollegeJSS Academy of Higher Education and ResearchMysuruIndia
| | - Saisudha Koka
- Department of Pharmaceutical Sciences, Irma Lerma Rangel School of PharmacyTexas A&M UniversityKingsvilleTXUSA
| | - Divya P. Kumar
- Department of Biochemistry, CEMR Lab, JSS Medical CollegeJSS Academy of Higher Education and ResearchMysuruIndia
| |
Collapse
|
27
|
Wang K, Margolis S, Cho JM, Wang S, Arianpour B, Jabalera A, Yin J, Hong W, Zhang Y, Zhao P, Zhu E, Reddy S, Hsiai TK. Non-Invasive Detection of Early-Stage Fatty Liver Disease via an On-Skin Impedance Sensor and Attention-Based Deep Learning. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400596. [PMID: 38887178 PMCID: PMC11336938 DOI: 10.1002/advs.202400596] [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: 01/16/2024] [Revised: 03/17/2024] [Indexed: 06/20/2024]
Abstract
Early-stage nonalcoholic fatty liver disease (NAFLD) is a silent condition, with most cases going undiagnosed, potentially progressing to liver cirrhosis and cancer. A non-invasive and cost-effective detection method for early-stage NAFLD detection is a public health priority but challenging. In this study, an adhesive, soft on-skin sensor with low electrode-skin contact impedance for early-stage NAFLD detection is fabricated. A method is developed to synthesize platinum nanoparticles and reduced graphene quantum dots onto the on-skin sensor to reduce electrode-skin contact impedance by increasing double-layer capacitance, thereby enhancing detection accuracy. Furthermore, an attention-based deep learning algorithm is introduced to differentiate impedance signals associated with early-stage NAFLD in high-fat-diet-fed low-density lipoprotein receptor knockout (Ldlr-/-) mice compared to healthy controls. The integration of an adhesive, soft on-skin sensor with low electrode-skin contact impedance and the attention-based deep learning algorithm significantly enhances the detection accuracy for early-stage NAFLD, achieving a rate above 97.5% with an area under the receiver operating characteristic curve (AUC) of 1.0. The findings present a non-invasive approach for early-stage NAFLD detection and display a strategy for improved early detection through on-skin electronics and deep learning.
Collapse
Affiliation(s)
- Kaidong Wang
- Department of MedicineDavid Geffen School of MedicineUniversity of California Los AngelesLos AngelesCA90095USA
- Department of Bioengineering, Henry Samueli School of Engineering and Applied SciencesUniversity of California Los AngelesLos AngelesCA90095USA
- Department of MedicineGreater Los Angeles Veterans Affairs (VA) Healthcare SystemLos AngelesCA90073USA
| | - Samuel Margolis
- Department of MedicineDavid Geffen School of MedicineUniversity of California Los AngelesLos AngelesCA90095USA
| | - Jae Min Cho
- Department of MedicineDavid Geffen School of MedicineUniversity of California Los AngelesLos AngelesCA90095USA
| | - Shaolei Wang
- Department of Bioengineering, Henry Samueli School of Engineering and Applied SciencesUniversity of California Los AngelesLos AngelesCA90095USA
| | - Brian Arianpour
- Department of Bioengineering, Henry Samueli School of Engineering and Applied SciencesUniversity of California Los AngelesLos AngelesCA90095USA
| | - Alejandro Jabalera
- Department of Bioengineering, Henry Samueli School of Engineering and Applied SciencesUniversity of California Los AngelesLos AngelesCA90095USA
| | - Junyi Yin
- Department of Bioengineering, Henry Samueli School of Engineering and Applied SciencesUniversity of California Los AngelesLos AngelesCA90095USA
| | - Wen Hong
- Department of Materials Science and EngineeringUniversity of California Los AngelesLos AngelesCA90095USA
| | - Yaran Zhang
- Department of Bioengineering, Henry Samueli School of Engineering and Applied SciencesUniversity of California Los AngelesLos AngelesCA90095USA
| | - Peng Zhao
- Department of MedicineDavid Geffen School of MedicineUniversity of California Los AngelesLos AngelesCA90095USA
| | - Enbo Zhu
- Department of MedicineDavid Geffen School of MedicineUniversity of California Los AngelesLos AngelesCA90095USA
- Department of Materials Science and EngineeringUniversity of California Los AngelesLos AngelesCA90095USA
| | - Srinivasa Reddy
- Department of Molecular and Medical PharmacologyUniversity of California Los AngelesLos AngelesCA90095USA
| | - Tzung K. Hsiai
- Department of MedicineDavid Geffen School of MedicineUniversity of California Los AngelesLos AngelesCA90095USA
- Department of Bioengineering, Henry Samueli School of Engineering and Applied SciencesUniversity of California Los AngelesLos AngelesCA90095USA
- Department of MedicineGreater Los Angeles Veterans Affairs (VA) Healthcare SystemLos AngelesCA90073USA
| |
Collapse
|
28
|
Ramírez-Vélez R, Izquierdo M, García-Hermoso A, Correa-Rodríguez M. Reference values and associated factors of controlled attenuation parameter and liver stiffness in adults: A cross-sectional study. Nutr Metab Cardiovasc Dis 2024; 34:1879-1889. [PMID: 38866615 DOI: 10.1016/j.numecd.2024.04.004] [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: 02/17/2024] [Revised: 04/02/2024] [Accepted: 04/08/2024] [Indexed: 06/14/2024]
Abstract
BACKGROUND & AIMS The utilization of non-invasive techniques for liver fibrosis and steatosis assessment has gained acceptance as a viable substitute for liver biopsy in clinical practice. This study aimed to establish normative data for the controlled attenuation parameter (CAP) and liver stiffness measurement (LSM) by age and gender, as well as to explore the relationship between anthropometric measures, clinical status, and biochemical profile according to the 90th percentile cut-off values for CAP/LSM in a U.S. adult population. METHODS AND RESULTS In this cross-sectional analysis, 7.522 US adults aged 20-80 years from the National Health and Nutrition Examination Survey (NHANES 2017-2020) were included. CAP and LSM were quantified using the FibroScan® 502-v2 device. A comprehensive range of data was collected, including sociodemographic, anthropometric, biochemical, lifestyle, and clinical conditions. Participants were segmented by sex and age. The median ± standard deviation (SD) for CAP was significantly lower in women (258.27 ± 61.02 dB/m) than in men (273.43 ± 63.56 dB/m), as was the median ± SD for LSM (women: 5.50 ± 4.12 kPa, men: 6.36 ± 5.63 kPa). Although median CAP and LSM values displayed an upward trend with age, statistical significance was not achieved. Notably, higher liver CAP values (above the 90th percentile) correlated with more pronounced clinical and biochemical profile differences compared to lower CAP values (below the 90th percentile) (p < 0.001). CONCLUSIONS Our study provides age- and sex-stratified standard values for CAP and LSM in a sizeable, nationally representative cohort of adults. The evidence of sex-specific variations in TE test results from our study sets the stage for future research to further corroborate these findings.
Collapse
Affiliation(s)
- Robinson Ramírez-Vélez
- Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain; CIBER of Frailty and Healthy Aging (CIBERFES), Instituto de Salud Carlos III, 28029 Madrid, Spain.
| | - Mikel Izquierdo
- Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain; CIBER of Frailty and Healthy Aging (CIBERFES), Instituto de Salud Carlos III, 28029 Madrid, Spain.
| | - Antonio García-Hermoso
- Navarrabiomed, Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain.
| | - María Correa-Rodríguez
- Department of Nursing, Faculty of Health Sciences, University of Granada (UGR), Av. Ilustración, 60, 18016 Granada, Spain; Instituto de Investigación Biosanitaria Granada (IBIS Granada), Av. de Madrid, 15, Pabellón de consultas externas 2, 2(a) planta, 18012 Granada, Spain.
| |
Collapse
|
29
|
Anasdeen S M, S S, Tm V. Evaluating Bempedoic Acid for Non-alcoholic Fatty Liver Disease: A Review of Preclinical and Clinical Research. Cureus 2024; 16:e67151. [PMID: 39295675 PMCID: PMC11408740 DOI: 10.7759/cureus.67151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 08/18/2024] [Indexed: 09/21/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most prevalent liver disorder globally, characterized by fat accumulation in liver cells, which can progress to inflammation, fibrosis, and cirrhosis. The disease predominantly affects individuals with obesity and high body mass index (BMI). It is a globally prevalent condition, with variations in incidence across different regions. The pathophysiology of NAFLD involves insulin resistance, metabolic disturbances, and genetic and gut microbial factors. Current treatments primarily focus on lifestyle modifications and a limited range of pharmacological options. Bempedoic acid (BA), a novel cholesterol-lowering agent, targets adenosine triphosphate (ATP)-citrate lyase to reduce low-density lipoprotein (LDL) cholesterol and has shown potential in managing NAFLD by decreasing liver fat accumulation and improving lipid profiles. BA's unique mechanism offers a promising add-on therapy, particularly for the patient's intolerant to statins. Despite its potential, comprehensive clinical and preclinical studies are needed to further elucidate its efficacy and safety compared to other NAFLD treatments. Future research should focus on comparing BA with existing and emerging therapies to optimize its role in NAFLD management and enhance patient outcomes.
Collapse
Affiliation(s)
- Mohamed Anasdeen S
- Department of Pharmacy Practice, College of Pharmacy, SRM Institute of Science and Technology, Chennai, IND
| | - Shahinaz S
- Department of Pharmacy Practice, College of Pharmacy, SRM Institute of Science and Technology, Chennai, IND
| | - Vijayakumar Tm
- Department of Pharmacy Practice, College of Pharmacy, SRM Institute of Science and Technology, Chennai, IND
| |
Collapse
|
30
|
Ortenzi VH, Oliveira ACD, Vasconcelos RP, Neves MB, Teixeira AJ, Oliveira KA, Ferreira ACF, Takiya CM, Fortunato RS. High-fat diet elicits sex-based differences in liver inflammatory cytokines and redox homeostasis. Appl Physiol Nutr Metab 2024; 49:1083-1092. [PMID: 38648669 DOI: 10.1139/apnm-2023-0457] [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] [Indexed: 04/25/2024]
Abstract
Sex differences in metabolic dysfunction-associated steatotic liver disease (MASLD) have been reported. Oxidative stress and inflammation are involved in the progression of MASLD. Thus, we aimed to evaluate liver redox homeostasis and inflammation in male and female rats fed a high-fat diet (HFD). Male and female Wistar rats were divided into the following groups: standard chow diet (SCD) or HFD during 12 weeks. HFD groups of both sexes had higher hepatocyte injury, with no differences between the sexes. Portal space liver inflammation was higher in females-HFD compared to females-SCD, whereas no differences were observed in males. Lobular inflammation and overall liver inflammation were higher in HFD groups, regardless of sex. TNF-α, IL-6, and IL-1β levels were higher in males-HFD compared to males-SCD, but no differences were observed in females. Catalase activity was higher in males compared to females, with no differences between the SCD and HFD groups of both sexes. Glutathione peroxidase activity was higher in females compared to males, with no differences between the SCD and HFD groups in both sexes. Lipid peroxidation was higher in female-SCD when compared to male-SCD, and in both male- and female-HFD compared to SCD groups. Furthermore, both cytoplasmic and nuclear NRF2 staining were lower in the HFD group compared to the SCD group in males. However, female-HFD exhibited reduced nuclear NRF2 staining compared to the female-SCD group. In conclusion, our study demonstrated that while both male and female rats developed metabolic dysfunction-associated steatohepatitis after 12 weeks of HFD, the alterations in inflammatory cytokines and redox balance were sexually dimorphic.
Collapse
Affiliation(s)
- Victor Hugo Ortenzi
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | - Marcelo Barbosa Neves
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Keciany Alves Oliveira
- Instituto Superior de Ciências Biomédicas, Universidade Estadual do Ceará, Ceará, Brazil
| | | | - Christina Maeda Takiya
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rodrigo S Fortunato
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
31
|
Huang CH, Hsu HS, Chiang MT. Influence of Varied Dietary Cholesterol Levels on Lipid Metabolism in Hamsters. Nutrients 2024; 16:2472. [PMID: 39125351 PMCID: PMC11314022 DOI: 10.3390/nu16152472] [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: 07/03/2024] [Revised: 07/25/2024] [Accepted: 07/27/2024] [Indexed: 08/12/2024] Open
Abstract
Syrian hamsters are valuable models for studying lipid metabolism due to their sensitivity to dietary cholesterol, yet the precise impact of varying cholesterol levels has not been comprehensively assessed. This study examined the impact of varying dietary cholesterol levels on lipid metabolism in Syrian hamsters. Diets ranging from 0% to 1% cholesterol were administered to assess lipid profiles and oxidative stress markers. Key findings indicate specific cholesterol thresholds for inducing distinct lipid profiles: below 0.13% for normal lipids, 0.97% for elevated LDL-C, 0.43% for increased VLDL-C, and above 0.85% for heightened hepatic lipid accumulation. A cholesterol supplementation of 0.43% induced hypercholesterolemia without adverse liver effects or abnormal lipoprotein expression. Furthermore, cholesterol supplementation significantly increased liver weight, plasma total cholesterol, LDL-C, and VLDL-C levels while reducing the HDL-C/LDL-C ratio. Fecal cholesterol excretion increased, with stable bile acid levels. High cholesterol diets correlated with elevated plasma ALT activities, reduced hepatic lipid peroxidation, and altered leptin and CETP levels. These findings underscore Syrian hamsters as robust models for hyperlipidemia research, offering insights into experimental methodologies. The identified cholesterol thresholds facilitate precise lipid profile manipulation, enhancing the hamster's utility in lipid metabolism studies and potentially informing clinical approaches to managing lipid disorders.
Collapse
Affiliation(s)
| | | | - Meng-Tsan Chiang
- Department of Food Science, College of Life Sciences, National Taiwan Ocean University, Keelung 20224, Taiwan; (C.-H.H.); (H.-S.H.)
| |
Collapse
|
32
|
Pan M, Deng Y, Qiu Y, Pi D, Zheng C, Liang Z, Zhen J, Fan W, Song Q, Pan J, Li Y, Yan H, Yang Q, Zhang Y. Shenling Baizhu powder alleviates non-alcoholic fatty liver disease by modulating autophagy and energy metabolism in high-fat diet-induced rats. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155712. [PMID: 38763008 DOI: 10.1016/j.phymed.2024.155712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 04/18/2024] [Accepted: 05/03/2024] [Indexed: 05/21/2024]
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) has emerged as a burgeoning health problem worldwide, but no specific drug has been approved for its treatment. Shenling Baizhu powder (SL) is extensively used to treat NAFLD in Chinese clinical practice. However, the therapeutic components and pharmacological mechanisms of SL against NAFLD have not been thoroughly investigated. PURPOSE This study aimed to investigate the pharmacological impact and molecular mechanism of SL on NAFLD. METHODS First, we established an animal model of NAFLD by high-fat diet (HFD) feeding, and evaluated the therapeutic efficacy of SL on NAFLD by physiological, biochemical, pathological, and body composition analysis. Next, the effect of SL on autophagic flow in NAFLD rats was evaluated by ultrastructure, immunofluorescence staining, and western blotting. Moreover, an integrated strategy of targeted energy metabolomics and network pharmacology was performed to characterize autophagy-related genes and explore the synergistic effects of SL active compounds. UPLC-MS/MS, molecular docking combined with in vivo and in vitro experiments were conducted to verify the key compounds and genes. Finally, a network was established among SL-herb-compound-genes-energy metabolites-NAFLD, which explains the complicated regulating mechanism of SL on NAFLD. RESULTS We discovered that SL decreased hepatic lipid accumulation, hepatic steatosis, and insulin resistance, and improved systemic metabolic disorders and pathological abnormalities. Subsequently, an integrated strategy of targeted energy metabolomics and network pharmacology identified quercetin, ellagic acid, kaempferol, formononetin, stigmasterol, isorhamnetin and luteolin as key compounds; catalase (CAT), AKT serine/threonine kinase 1 (AKT), nitric oxide synthase 3 (eNOS), NAD(P)H quinone dehydrogenase 1 (NQO1), heme oxygenase 1 (HO-1) and hypoxia-inducible factor 1 subunit alpha (HIF-1α) were identified as key genes; while nicotinamide adenine dinucleotide phosphate (NADP) and succinate emerged as key energy metabolites. Mechanistically, we revealed that SL may exert its anti-NAFLD effect by inducing autophagy activation and forming a comprehensive regulatory network involving key compounds, key genes, and key energy metabolites, ultimately alleviating oxidative stress, endoplasmic reticulum stress, and mitochondrial dysfunction. CONCLUSION Our study demonstrated the therapeutic effect of SL in NAFLD models, and establishes a basis for the development of potential products from SL plant materials for the treatment of NAFLD.
Collapse
Affiliation(s)
- Maoxing Pan
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, Guangdong Province, China
| | - Yuanjun Deng
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, Guangdong Province, China; Shenzhen Traditional Chinese Medicine Hospital, Shenzhen 518033, Guangdong Province, China
| | - Yebei Qiu
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, Guangdong Province, China
| | - Dajin Pi
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, Guangdong Province, China
| | - Chuiyang Zheng
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, Guangdong Province, China
| | - Zheng Liang
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, Guangdong Province, China
| | - Jianwei Zhen
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, Guangdong Province, China
| | - Wen Fan
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, Guangdong Province, China
| | - Qingliang Song
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, Guangdong Province, China
| | - Jinyue Pan
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, Guangdong Province, China
| | - Yuanyou Li
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, Guangdong Province, China
| | - Haizhen Yan
- Guangzhou Red Cross Hospital, Jinan University, Guangzhou 510240, Guangdong Province, China.
| | - Qinhe Yang
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, Guangdong Province, China.
| | - Yupei Zhang
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, Guangdong Province, China.
| |
Collapse
|
33
|
Levy E, Fallet-Bianco C, Auclair N, Patey N, Marcil V, Sané AT, Spahis S. Unraveling Chylomicron Retention Disease Enhances Insight into SAR1B GTPase Functions and Mechanisms of Actions, While Shedding Light of Intracellular Chylomicron Trafficking. Biomedicines 2024; 12:1548. [PMID: 39062121 PMCID: PMC11274388 DOI: 10.3390/biomedicines12071548] [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: 05/22/2024] [Revised: 06/28/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Over the past three decades, significant efforts have been focused on unraveling congenital intestinal disorders that disrupt the absorption of dietary lipids and fat-soluble vitamins. The primary goal has been to gain deeper insights into intra-enterocyte sites, molecular steps, and crucial proteins/regulatory pathways involved, while simultaneously identifying novel therapeutic targets and diagnostic tools. This research not only delves into specific and rare malabsorptive conditions, such as chylomicron retention disease (CRD), but also contributes to our understanding of normal physiology through the utilization of cutting-edge cellular and animal models alongside advanced research methodologies. This review elucidates how modern techniques have facilitated the decoding of CRD gene defects, the identification of dysfunctional cellular processes, disease regulatory mechanisms, and the essential role of coat protein complex II-coated vesicles and cargo receptors in chylomicron trafficking and endoplasmic reticulum (ER) exit sites. Moreover, experimental approaches have shed light on the multifaceted functions of SAR1B GTPase, wherein loss-of-function mutations not only predispose individuals to CRD but also exacerbate oxidative stress, inflammation, and ER stress, potentially contributing to clinical complications associated with CRD. In addition to dissecting the primary disease pathology, genetically modified animal models have emerged as invaluable assets in exploring various ancillary aspects, including responses to environmental challenges such as dietary alterations, gender-specific disparities in disease onset and progression, and embryonic lethality or developmental abnormalities. In summary, this comprehensive review provides an in-depth and contemporary analysis of CRD, offering a meticulous examination of the CRD current landscape by synthesizing the latest research findings and advancements in the field.
Collapse
Affiliation(s)
- Emile Levy
- Azrieli Research Center, CHU Ste-Justine and Department of Nutrition, Université de Montréal, Montreal, QC H3T 1C5, Canada
| | - Catherine Fallet-Bianco
- Azrieli Research Center, CHU Ste-Justine and Pathology & Cell Biology, Université de Montréal, Montreal, QC H3T 1C5, Canada
| | - Nickolas Auclair
- Azrieli Research Center, CHU Ste-Justine and Pharmacology, Université de Montréal, Montreal, QC H3T 1C5, Canada
| | - Natalie Patey
- Azrieli Research Center, CHU Ste-Justine and Pathology & Cell Biology, Université de Montréal, Montreal, QC H3T 1C5, Canada
| | - Valérie Marcil
- Azrieli Research Center, CHU Ste-Justine and Department of Nutrition, Université de Montréal, Montreal, QC H3T 1C5, Canada
| | | | - Schohraya Spahis
- Azrieli Research Center, CHU Ste-Justine and Biochemistry & Molecular Medicine, Université de Montréal, Montreal, QC H3T 1C5, Canada;
| |
Collapse
|
34
|
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
|
35
|
Zheng Y, Zhao L, Xiong Z, Huang C, Yong Q, Fang D, Fu Y, Gu S, Chen C, Li J, Zhu Y, Liu J, Liu F, Li Y. Ursolic acid targets secreted phosphoprotein 1 to regulate Th17 cells against metabolic dysfunction-associated steatotic liver disease. Clin Mol Hepatol 2024; 30:449-467. [PMID: 38623614 PMCID: PMC11261229 DOI: 10.3350/cmh.2024.0047] [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: 01/18/2024] [Revised: 04/12/2024] [Accepted: 04/15/2024] [Indexed: 04/17/2024] Open
Abstract
BACKGROUND/AIMS Metabolic dysfunction-associated steatotic liver disease (MASLD) has become an increasingly important health challenge, with a substantial rise linked to changing lifestyles and global obesity. Ursolic acid, a natural pentacyclic triterpenoid, has been explored for its potential therapeutic effects. Given its multifunctional bioactive properties, this research further revealed the pharmacological mechanisms of ursolic acid on MASLD. METHODS Drug target chips and bioinformatics analysis were combined in this study to explore the potential therapeutic effects of ursolic acid on MASLD. Molecular docking simulations, surface plasmon resonance analyses, pull-down experiments, and co-immunoprecipitation assays were used to verify the direct interactions. Gene knockdown mice were generated, and high-fat diets were used to validate drug efficacy. Furthermore, initial CD4+ T cells were isolated and stimulated to demonstrate our findings. RESULTS In this study, the multifunctional extracellular matrix phosphorylated glycoprotein secreted phosphoprotein 1 (SPP1) was investigated, highlighting its capability to induce Th17 cell differentiation, amplifying inflammatory cascades, and subsequently promoting the evolution of MASLD. In addition, this study revealed that in addition to the canonical TGF-β/IL-6 cytokine pathway, SPP1 can directly interact with ITGB1 and CD44, orchestrating Th17 cell differentiation via their joint downstream ERK signaling pathway. Remarkably, ursolic acid intervention notably suppressed the protein activity of SPP1, suggesting a promising avenue for ameliorating the immunoinflammatory trajectory in MASLD progression. CONCLUSION Ursolic acid could improve immune inflammation in MASLD by modulating SPP1-mediated Th17 cell differentiation via the ERK signaling pathway, which is orchestrated jointly by ITGB1 and CD44, emerging as a linchpin in this molecular cascade.
Collapse
Affiliation(s)
- Yiyuan Zheng
- Department of Gastroenterology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lina Zhao
- Department of Gastroenterology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhekun Xiong
- Department of Spleen, Stomach and Hepatobiliary, Zhongshan Hospital of Traditional Chinese Medicine, Zhongshan, China
| | - Chaoyuan Huang
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Gastroenterology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Qiuhong Yong
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Dan Fang
- Medical Affairs Department, Ton-Bridge Medical Technology Co., Ltd., Zhuhai, China
| | - Yugang Fu
- Department of Gastroenterology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Simin Gu
- Department of Gastroenterology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chong Chen
- Department of Gastroenterology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiacheng Li
- Department of Gastroenterology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yingying Zhu
- Department of Gastroenterology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jing Liu
- Department of Gastroenterology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fengbin Liu
- Department of Gastroenterology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yong Li
- Department of Gastroenterology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
36
|
Xu Q, Xing J, Wang S, Peng H, Liu Y. The role of the cGAS-STING pathway in metabolic diseases. Heliyon 2024; 10:e33093. [PMID: 38988528 PMCID: PMC11234105 DOI: 10.1016/j.heliyon.2024.e33093] [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: 01/18/2024] [Revised: 06/13/2024] [Accepted: 06/13/2024] [Indexed: 07/12/2024] Open
Abstract
The cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway is a critical innate immune pathway primarily due to its vital DNA sensing mechanism in pathogen defence. Recent research advances have shown that excessive activation or damage to the cGAS-STING pathway can exacerbate chronic inflammatory responses, playing a significant role in metabolic dysfunction and aging, leading to the development of related diseases such as obesity, osteoporosis, and neurodegenerative diseases. This article reviews the structure and biological functions of the cGAS-STING signaling pathway and discusses in detail how this pathway regulates the occurrence and development of metabolic and age-related diseases. Additionally, this article introduces potential small molecule drugs targeting cGAS and STING, aiming to provide new research perspectives for studying the pathogenesis and treatment of metabolic-related diseases.
Collapse
Affiliation(s)
- Qian Xu
- Department of Endocrinology, The Affiliated People's Hospital of Jiangsu University, Zhenjiang Medical School of Nanjing Medical University, Zhenjiang, 212002, China
| | - Jie Xing
- Department of Laboratory Medicine, The Affiliated People's Hospital of Jiangsu University, Zhenjiang Medical School of Nanjing Medical University, Zhenjiang, 212002, China
| | - Shengjun Wang
- Department of Laboratory Medicine, The Affiliated People's Hospital of Jiangsu University, Zhenjiang Medical School of Nanjing Medical University, Zhenjiang, 212002, China
| | - Huiyong Peng
- Department of Laboratory Medicine, The Affiliated People's Hospital of Jiangsu University, Zhenjiang Medical School of Nanjing Medical University, Zhenjiang, 212002, China
| | - Yingzhao Liu
- Department of Endocrinology, The Affiliated People's Hospital of Jiangsu University, Zhenjiang Medical School of Nanjing Medical University, Zhenjiang, 212002, China
| |
Collapse
|
37
|
Shakhshir M, Zyoud SH. Mapping global research trends: Nutrition associations with nonalcoholic fatty liver disease - a Scopus bibliometric analysis. World J Gastroenterol 2024; 30:3106-3119. [PMID: 38983957 PMCID: PMC11230064 DOI: 10.3748/wjg.v30.i24.3106] [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: 03/04/2024] [Revised: 05/10/2024] [Accepted: 06/05/2024] [Indexed: 06/25/2024] Open
Abstract
BACKGROUND Several bibliometric analyses have been carried out to identify research hotspots and trends in nonalcoholic fatty liver disease (NAFLD) research. Nonetheless, there are still significant knowledge gaps that must be filled to advance our understanding of and ability to treat NAFLD. AIM To evaluate, through bibliometric and visual analysis, the current status of related research, related research frontiers, and the developmental trends in the field of diet and NAFLD. METHODS We retrieved publications about diet and NAFLD published between 1987 and 2022 from Scopus. Next, we used VOSviewer 1.6.20 to perform bibliometric analysis and visualization. RESULTS We found a total of 1905 studies, including 1637 (85.93%) original articles and 195 (10.24%) reviews, focused on the examination of NAFLD and its correlation with diet that were published between 1987 and 2022. Among the remaining five types of documents, 38 were letters, notes, editorials, meeting minutes, or brief surveys, representing 1.99% of the total documents. The countries with the most publications on this topic were China (n = 539; 28.29%), followed by the United States (n = 379; 19.90%), Japan (n = 133; 6.98%), and South Korea (n = 127; 6.6%). According to the citation analysis, the retrieved papers were cited an average of 32.3 times and had an h-index of 106, with 61014 total citations. The two main clusters on the map included those related to: (1) Inflammation and oxidative stress; and (2) Dietary interventions for NAFLD. CONCLUSION This was the first study to use data taken from Scopus to visualize network mapping in a novel bibliometric analysis of studies focused on diet and NAFLD. After 2017, the two domains that received the most attention were "dietary interventions for NAFL"' and "'inflammation and oxidative stress implicated in NAFLD and its correlation with diet." We believe that this study provides important information for academics, dietitians, and doctors, and that additional research on dietary interventions and NAFLD is warranted.
Collapse
Affiliation(s)
- Muna Shakhshir
- Department of Nutrition, An-Najah National University Hospital, Nablus 44839, Palestine
- Department of Public Health, College of Medicine and Health Sciences, An-Najah National University, Nablus 44839, Palestine
| | - Sa'ed H Zyoud
- Department of Clinical and Community Pharmacy, College of Medicine and Health Sciences, An-Najah National University, Nablus 44839, Palestine
- Poison Control and Drug Information Center, College of Medicine and Health Sciences, An-Najah National University, Nablus 44839, Palestine
- Clinical Research Center, An-Najah National University Hospital, Nablus 44839, Palestine
| |
Collapse
|
38
|
Lu Z, Wu S, Feng E, Chen X, Chen J, Lin F. Association between hepatic steatosis and fibrosis and arthritis among US adults: A population-based study. Clinics (Sao Paulo) 2024; 79:100378. [PMID: 38875754 PMCID: PMC11225167 DOI: 10.1016/j.clinsp.2024.100378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 04/01/2024] [Indexed: 06/16/2024] Open
Abstract
BACKGROUND Lipid metabolism factors may play a role in the development of arthritis and hepatic steatosis and fibrosis. The aim of this study was to explore the potential association between arthritis and hepatic steatosis and liver fibrosis. MATERIALS AND METHODS The nationally representative sample from the National Health and Nutrition Examination Survey was analyzed, with data on arthritis diagnosis, subtype, and liver status obtained. Liver status was assessed using transient elastography. Hepatic steatosis was defined as a Controlled Attenuation Parameter (CAP) score ≥263 dB/m, and liver fibrosis status was defined as F0‒F4. Logistic regression models and subgroup analyses stratified by sex were used to evaluate the associations. Smooth curve fitting was used to describe the associations. RESULTS The present study of 6,840 adults aged 20 years or older found a significant positive correlation between arthritis and CAP in multivariate logistic regression analysis (β = 0.003, 95 % CI 0.001 to 0.0041, p < 0.001). Participants with arthritis had a higher risk of hepatic steatosis (OR = 1.248, 95 % CI 1.036 to 1.504, p = 0.020), particularly those with osteoarthritis or degenerative arthritis, but not rheumatoid arthritis (p = 0.847). The positive correlation was maintained in females (β = 0.004, 95 % CI 0.002 to 0.006, p < 0.001), but not in males. There was no significant relationship between arthritis and liver fibrosis (p = 0.508). CONCLUSION This study indicates that there is a positive correlation between arthritis and hepatic steatosis, particularly in females. Nonetheless, there is no significant relationship between arthritis and the risk of liver fibrosis.
Collapse
Affiliation(s)
- Zhiming Lu
- Fujian Medical University Union Hospital, Fuzhou, China; Fuzhou City Second Hospital, Fuzhou, China; The Third Clinical Medical College, Fujian Medical University, China
| | - Shaojie Wu
- Fuzhou City Second Hospital, Fuzhou, China; Fujian Provincial Clinical Medical Research Center for First 339 Aid and Rehabilitation in Orthopaedic Trauma (2020Y2014), China
| | - Eryou Feng
- Fujian Medical University Union Hospital, Fuzhou, China; The Third Clinical Medical College, Fujian Medical University, China.
| | | | - Jinhua Chen
- Fujian Medical University Union Hospital, Fuzhou, China
| | - Feitai Lin
- Fuzhou City Second Hospital, Fuzhou, China
| |
Collapse
|
39
|
Ma Y, Wang J, Xiao W, Fan X. A review of MASLD-related hepatocellular carcinoma: progress in pathogenesis, early detection, and therapeutic interventions. Front Med (Lausanne) 2024; 11:1410668. [PMID: 38895182 PMCID: PMC11184143 DOI: 10.3389/fmed.2024.1410668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 05/20/2024] [Indexed: 06/21/2024] Open
Abstract
The incidence of metabolic dysfunction-associated steatotic liver disease (MASLD) is continuously rising, evolving into a global health challenge. Concurrently, cases of hepatocellular carcinoma (HCC) associated with MASLD are also on the increase. Although traditional risk factors such as age, gender, and metabolic factors play significant roles in the development of HCC, it cannot be overlooked that MASLD, triggered by changes in modern lifestyle and dietary habits, may also exacerbate the risk of HCC, and this phenomenon is common even among non-obese individuals. Regrettably, MASLD often fails to receive timely diagnosis, resulting in a limited number of patients receiving HCC surveillance. Moreover, there is currently a lack of clear definition for the target population for surveillance beyond patients with cirrhosis. Consequently, MASLD-related HCC is often detected at a late stage, precluding the optimal timing for curative treatment. However, our understanding of the pathogenesis and progression of HCC remains limited. Therefore, this paper reviews relevant literature from recent years, delving into multiple dimensions such as pathogenesis, surveillance and diagnosis, prevention, and treatment, aiming to provide new ideas and directions for the prevention and treatment of MASLD-related HCC.
Collapse
Affiliation(s)
- Yang Ma
- Department of Human Anatomy, School of Basic Medicine, Guilin Medical University, Guilin, China
| | - Jinguo Wang
- School of Public Health, Guilin Medical University, Guilin, China
| | - Wenping Xiao
- Department of Human Anatomy, School of Basic Medicine, Guilin Medical University, Guilin, China
| | - Xiaoming Fan
- Department of Human Anatomy, School of Basic Medicine, Guilin Medical University, Guilin, China
| |
Collapse
|
40
|
Meng X, Long M, Yue N, Li Q, Chen J, Zhao H, Deng W. LncRNA MEG3 Restrains Hepatic Lipogenesis via the FOXO1 Signaling Pathway in HepG2 Cells. Cell Biochem Biophys 2024; 82:1253-1259. [PMID: 38713402 PMCID: PMC11344712 DOI: 10.1007/s12013-024-01278-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2024] [Indexed: 05/08/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD) become a main public health concern, and is characterized by lipid accumulation in the hepatocytes. We found that overexpression of lncRNA MEG3 significantly reduced the expression of FOXO1, ACC1, and FAS, and subsequently decreased the lipid accumulation in HepG2 cells. Moreover, inhibition of lncRNA MEG3 could increase the lipid accumulation and the mRNA and protein levels of FOXO1, ACC1, and FAS. Further study showed that lncRNA MEG3 regulates the lipogenesis process by inhibiting the entry of FOXO1 into the nucleus translocation. Our study demonstrated that lncRNA MEG3 regulates de novo lipogenesis by decreasing the expression and nucleus translocation of FOXO1 in HepG2 cells, suggesting that lncRNA MEG3 could be a promising therapeutic target in lipid metabolic disorders.
Collapse
Affiliation(s)
- Xiangyu Meng
- The Central Laboratory, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Mei Long
- Department of Cardiology, ZiBo Central Hospital, Zibo, Shandong, 255000, China
| | - Nanxi Yue
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education of China, Hebei Medical University, Shijiazhuang, Hebei, 050017, PR China
| | - Quan Li
- Department of Endocrinology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Jia Chen
- Department of Endocrinology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Hongye Zhao
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education of China, Hebei Medical University, Shijiazhuang, Hebei, 050017, PR China.
| | - Wei Deng
- Department of Endocrinology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China.
| |
Collapse
|
41
|
Puengel T, Tacke F. Pharmacotherapeutic options for metabolic dysfunction-associated steatotic liver disease: where are we today? Expert Opin Pharmacother 2024; 25:1249-1263. [PMID: 38954663 DOI: 10.1080/14656566.2024.2374463] [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: 04/29/2024] [Accepted: 06/26/2024] [Indexed: 07/04/2024]
Abstract
INTRODUCTION Metabolic dysfunction-associated steatotic liver disease (MASLD) is defined by hepatic steatosis and cardiometabolic risk factors like obesity, type 2 diabetes, and dyslipidemia. Persistent metabolic injury may promote inflammatory processes resulting in metabolic dysfunction-associated steatohepatitis (MASH) and liver fibrosis. Mechanistic insights helped to identify potential drug targets, thereby supporting the development of novel compounds modulating disease drivers. AREAS COVERED The U.S. Food and Drug Administration has recently approved the thyroid hormone receptor β-selective thyromimetic resmetirom as the first compound to treat MASH and liver fibrosis. This review provides a comprehensive overview of current and potential future pharmacotherapeutic options and their modes of action. Lessons learned from terminated clinical trials are discussed together with the first results of trials investigating novel combinational therapeutic approaches. EXPERT OPINION Approval of resmetirom as the first anti-MASH agent may revolutionize the therapeutic landscape. However, long-term efficacy and safety data for resmetirom are currently lacking. In addition, heterogeneity of MASLD reflects a major challenge to define effective agents. Several lead compounds demonstrated efficacy in reducing obesity and hepatic steatosis, while anti-inflammatory and antifibrotic effects of monotherapy appear less robust. Better mechanistic understanding, exploration of combination therapies, and patient stratification hold great promise for MASLD therapy.
Collapse
Affiliation(s)
- Tobias Puengel
- Department of Hepatology & Gastroenterology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health at Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Frank Tacke
- Department of Hepatology & Gastroenterology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
42
|
Jin Y, Shangguan Z, Pang J, Chen Y, Lin S, Liu H. Pin1 Exacerbates Non-Alcoholic Fatty Liver Disease by Enhancing Its Activity through Binding to ACC1. Int J Mol Sci 2024; 25:5822. [PMID: 38892011 PMCID: PMC11171836 DOI: 10.3390/ijms25115822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 05/17/2024] [Accepted: 05/19/2024] [Indexed: 06/21/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a clinicopathological syndrome characterized by diffuse hepatocellular steatosis due to fatty deposits in hepatocytes, excluding alcohol and other known liver injury factors. However, there are no specific drugs for the clinical treatment of NAFLD. Therefore, research on the pathogenesis of NAFLD at the cellular and molecular levels is a promising approach to finding therapeutic targets and developing targeted drugs for NAFLD. Pin1 is highly expressed during adipogenesis and contributes to adipose differentiation, but its specific mechanism of action in NAFLD is unclear. In this study, we investigated the role of Pin1 in promoting the development of NAFLD and its potential mechanisms in vitro and in vivo. First, Pin1 was verified in the NAFLD model in vitro using MCD diet-fed mice by Western Blot, RT-qPCR and immunohistochemistry (IHC) assays. In the in vitro study, we used the oleic acid (OA) stimulation-induced lipid accumulation model and examined the lipid accumulation in each group of cells by oil red O staining as well as BODIPY staining. The results showed that knockdown of Pin1 inhibited lipid accumulation in hepatocytes in an in vitro lipid accumulation model and improved lipid indices and liver injury levels. Moreover, in vivo, WT and Pin1-KO mice were fed a methionine-choline deficient (MCD) diet for 4 weeks to induce the NAFLD model. The effects of Pin1 on lipid accumulation, hepatic fibrosis, and oxidative stress were evaluated by biochemical analysis, glucose and insulin tolerance tests, histological analysis, IHC, RT-qPCR and Western blot assays. The results indicate that Pin1 knockdown significantly alleviated hepatic steatosis, fibrosis and inflammation in MCD-induced NAFLD mice, improved glucose tolerance and alleviated insulin resistance in mice. Further studies showed that the AMPK/ACC1 signalling pathway might take part in the process by which Pin1 regulates NAFLD, as evidenced by the inhibition of the AMPK/ACC1 pathway. In addition, immunofluorescence (IF), coimmunoprecipitation (Co-IP) and GST pull-down experiments also showed that Pin1 interacts directly with ACC1 and inhibits ACC1 phosphorylation levels. Our study suggests that Pin1 promotes NAFLD progression by inhibiting the activation of the AMPK/ACC1 signalling pathway, and it is possible that this effect is achieved by Pin1 interacting with ACC1 and inhibiting the phosphorylation of ACC1.
Collapse
Affiliation(s)
| | | | | | | | | | - Hekun Liu
- Fujian Key Laboratory of Translational Research in Cancer and Neurodegenerative Diseases, The School of Basic Medical Sciences, Fujian Medical University, No. 1, Xuefu North Road, Fuzhou 350122, China; (Y.J.); (Z.S.); (J.P.); (Y.C.); (S.L.)
| |
Collapse
|
43
|
Mei EH, Yao C, Chen YN, Nan SX, Qi SC. Multifunctional role of oral bacteria in the progression of non-alcoholic fatty liver disease. World J Hepatol 2024; 16:688-702. [PMID: 38818294 PMCID: PMC11135273 DOI: 10.4254/wjh.v16.i5.688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 02/26/2024] [Accepted: 04/07/2024] [Indexed: 05/22/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) encompasses a spectrum of liver disorders of varying severity, ultimately leading to fibrosis. This spectrum primarily consists of NAFL and non-alcoholic steatohepatitis. The pathogenesis of NAFLD is closely associated with disturbances in the gut microbiota and impairment of the intestinal barrier. Non-gut commensal flora, particularly bacteria, play a pivotal role in the progression of NAFLD. Notably, Porphyromonas gingivalis, a principal bacterium involved in periodontitis, is known to facilitate lipid accumulation, augment immune responses, and induce insulin resistance, thereby exacerbating fibrosis in cases of periodontitis-associated NAFLD. The influence of oral microbiota on NAFLD via the "oral-gut-liver" axis is gaining recognition, offering a novel perspective for NAFLD management through microbial imbalance correction. This review endeavors to encapsulate the intricate roles of oral bacteria in NAFLD and explore underlying mechanisms, emphasizing microbial control strategies as a viable therapeutic avenue for NAFLD.
Collapse
Affiliation(s)
- En-Hua Mei
- Shanghai Medical College, Fudan University, Shanghai 200000, China
- Department of Prothodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai 200000, China
- Shanghai Key Laboratory of Craniomaxiofacial Development and Diseases, Fudan University, Shanghai 200000, China
| | - Chao Yao
- Department of Prothodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai 200000, China
- Shanghai Key Laboratory of Craniomaxiofacial Development and Diseases, Fudan University, Shanghai 200000, China
| | - Yi-Nan Chen
- Shanghai Medical College, Fudan University, Shanghai 200000, China
| | - Shun-Xue Nan
- Shanghai Medical College, Fudan University, Shanghai 200000, China
| | - Sheng-Cai Qi
- Department of Prothodontics, Shanghai Stomatological Hospital, Fudan University, Shanghai 200000, China
- Shanghai Key Laboratory of Craniomaxiofacial Development and Diseases, Fudan University, Shanghai 200000, China.
| |
Collapse
|
44
|
Ariyoshi K, Nishiyama K, Kato Y, Mi X, Ito T, Azuma YT, Nishimura A, Nishida M. Inhibition of Drp1-Filamin Protein Complex Prevents Hepatic Lipid Droplet Accumulation by Increasing Mitochondria-Lipid Droplet Contact. Int J Mol Sci 2024; 25:5446. [PMID: 38791484 PMCID: PMC11122359 DOI: 10.3390/ijms25105446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 05/14/2024] [Accepted: 05/14/2024] [Indexed: 05/26/2024] Open
Abstract
Lipid droplet (LD) accumulation in hepatocytes is one of the major symptoms associated with fatty liver disease. Mitochondria play a key role in catabolizing fatty acids for energy production through β-oxidation. The interplay between mitochondria and LD assumes a crucial role in lipid metabolism, while it is obscure how mitochondrial morphology affects systemic lipid metabolism in the liver. We previously reported that cilnidipine, an already existing anti-hypertensive drug, can prevent pathological mitochondrial fission by inhibiting protein-protein interaction between dynamin-related protein 1 (Drp1) and filamin, an actin-binding protein. Here, we found that cilnidipine and its new dihydropyridine (DHP) derivative, 1,4-DHP, which lacks Ca2+ channel-blocking action of cilnidipine, prevent the palmitic acid-induced Drp1-filamin interaction, LD accumulation and cytotoxicity of human hepatic HepG2 cells. Cilnidipine and 1,4-DHP also suppressed the LD accumulation accompanied by reducing mitochondrial contact with LD in obese model and high-fat diet-fed mouse livers. These results propose that targeting the Drp1-filamin interaction become a new strategy for the prevention or treatment of fatty liver disease.
Collapse
Affiliation(s)
- Kohei Ariyoshi
- Department of Physiology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (K.A.); (K.N.); (Y.K.); (X.M.); (T.I.)
| | - Kazuhiro Nishiyama
- Department of Physiology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (K.A.); (K.N.); (Y.K.); (X.M.); (T.I.)
- Laboratory of Prophylactic Pharmacology, Osaka Metropolitan University Graduate School of Veterinary Science, Osaka 598-8531, Japan;
| | - Yuri Kato
- Department of Physiology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (K.A.); (K.N.); (Y.K.); (X.M.); (T.I.)
| | - Xinya Mi
- Department of Physiology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (K.A.); (K.N.); (Y.K.); (X.M.); (T.I.)
| | - Tomoya Ito
- Department of Physiology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (K.A.); (K.N.); (Y.K.); (X.M.); (T.I.)
- National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences (NINS), Okazaki 444-8787, Japan;
| | - Yasu-Taka Azuma
- Laboratory of Prophylactic Pharmacology, Osaka Metropolitan University Graduate School of Veterinary Science, Osaka 598-8531, Japan;
| | - Akiyuki Nishimura
- National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences (NINS), Okazaki 444-8787, Japan;
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences (NINS), Okazaki 444-8787, Japan
- Department of Physiological Sciences, School of Life Science, The Graduate University for Advanced Studies (SOKENDAI), Okazaki 444-8787, Japan
| | - Motohiro Nishida
- Department of Physiology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (K.A.); (K.N.); (Y.K.); (X.M.); (T.I.)
- National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences (NINS), Okazaki 444-8787, Japan;
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences (NINS), Okazaki 444-8787, Japan
- Department of Physiological Sciences, School of Life Science, The Graduate University for Advanced Studies (SOKENDAI), Okazaki 444-8787, Japan
| |
Collapse
|
45
|
Yang S, Li J, Yan L, Wu Y, Zhang L, Li B, Tong H, Lin X. Molecular Mechanisms of Fucoxanthin in Alleviating Lipid Deposition in Metabolic Associated Fatty Liver Disease. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:10391-10405. [PMID: 38669300 DOI: 10.1021/acs.jafc.4c00590] [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: 04/28/2024]
Abstract
Metabolic-associated fatty liver disease (MAFLD) is witnessing a global surge; however, it still lacks effective pharmacological interventions. Fucoxanthin, a natural bioactive metabolite derived from marine brown algae, exhibits promising pharmacological functions, particularly in ameliorating metabolic disorders. However, the mechanisms underlying its therapeutic efficacy in addressing MAFLD remain elusive. Our present findings indicated that fucoxanthin significantly alleviated palmitic acid (PA)-induced hepatic lipid deposition in vitro and obesity-induced hepatic steatosis in ob/ob mice. Moreover, at both the protein and transcriptional levels, fucoxanthin effectively increased the expression of PPARα and CPT1 (involved in fatty acid oxidation) and suppressed FASN and SREBP1c (associated with lipogenesis) in both PA-induced HepG2 cells and hepatic tissues in ob/ob mice. This modulation was accompanied by the activation of AMPK. The capacity of fucoxanthin to improve hepatic lipid deposition was significantly attenuated when utilizing the AMPK inhibitor or siRNA-mediated AMPK silencing. Mechanistically, fucoxanthin activates AMPK, subsequently regulating the KEAP1/Nrf2/ARE signaling pathway to exert antioxidative effects and stimulating the PGC1α/NRF1 axis to enhance mitochondrial biogenesis. These collective actions contribute to fucoxanthin's amelioration of hepatic steatosis induced by metabolic perturbations. These findings offer valuable insights into the prospective utilization of fucoxanthin as a therapeutic strategy for managing MAFLD.
Collapse
Affiliation(s)
- Shouxing Yang
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, P.R. China
| | - Jinhai Li
- Department of Liver and Gall Surgery, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, P.R. China
| | - Liping Yan
- Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, P.R. China
| | - Yu Wu
- Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, P.R. China
| | - Lin Zhang
- Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, P.R. China
| | - Boyang Li
- Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, P.R. China
| | - Haibin Tong
- Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, P.R. China
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Beijing 100700, P.R. China
| | - Xiaochun Lin
- Department of Pediatrics Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, P.R. China
| |
Collapse
|
46
|
Weng C, Shao Z, Xiao M, Song M, Zhao Y, Li A, Pang Y, Huang T, Yu C, Lv J, Li L, Sun D. Association of sex hormones with non-alcoholic fatty liver disease: An observational and Mendelian randomization study. Liver Int 2024; 44:1154-1166. [PMID: 38345150 DOI: 10.1111/liv.15866] [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: 10/18/2023] [Revised: 01/20/2024] [Accepted: 01/28/2024] [Indexed: 04/19/2024]
Abstract
BACKGROUND AND AIMS Sex-specific associations of sex hormone-binding globulin (SHBG) and bioavailable testosterone (BAT) with NAFLD remain indeterminate. We aimed to explore observational and genetically determined relationships between each hormone and NAFLD. METHODS We included 187 395 men and 170 193 women from the UK Biobank. Linear and nonlinear Cox regression models and Mendelian randomization (MR) analysis were used to test the associations. RESULTS During 12.49 years of follow-up, 2209 male and 1886 female NAFLD cases were documented. Elevated SHBG levels were linearly associated with a lower risk of NAFLD in women (HR (95% CI), .71 (.63, .79)), but not in men (a "U" shape, pnon-linear < .001). Higher BAT levels were associated with a lower NAFLD risk in men (HR (95% CI), .81 (.71, .93)) but a higher risk in women (HR (95% CI): 1.25 (1.15, 1.36)). Genetically determined SHBG and BAT levels were linearly associated with NAFLD risk in women (OR (95% CI): .57 (.38, .87) and 2.21 (1.41, 3.26) respectively); in men, an "L-shaped" MR association between SHBG levels and NAFLD risk was found (pnon-linear = .016). The bidirectional MR analysis further revealed the effect of NAFLD on SHBG and BAT levels in both sexes. CONCLUSIONS Consistently, linear associations of lower SHBG and higher BAT levels with increased NAFLD risk were both conventionally and genetically found in women, while in men, SHBG acts in a nonlinear manner. In addition, NAFLD may affect SHBG and BAT levels.
Collapse
Affiliation(s)
- Chenghao Weng
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Zilun Shao
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
| | - Meng Xiao
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
| | - Mingyu Song
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
| | - Yuxuan Zhao
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
| | - Aolin Li
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
| | - Yuanjie Pang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
| | - Tao Huang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
| | - Canqing Yu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
- Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Jun Lv
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
- Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Liming Li
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
- Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Dianjianyi Sun
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
- Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| |
Collapse
|
47
|
Rudolfsen JH, Gluud LL, Grønbæk H, Jensen MK, Vyberg M, Olsen J, Bo Poulsen P, Hovelsø N, Gregersen NT, Thomsen AB, Jepsen P. Societal costs and survival of patients with biopsy-verified non-alcoholic steatohepatitis: Danish nationwide register-based study. Ann Hepatol 2024; 29:101285. [PMID: 38272183 DOI: 10.1016/j.aohep.2024.101285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 12/14/2023] [Accepted: 01/05/2024] [Indexed: 01/27/2024]
Abstract
INTRODUCTION AND OBJECTIVES Studies on the societal burden of patients with biopsy-confirmed non-alcoholic fatty liver disease (NAFLD) are sparse. This study examined this question, comparing NAFLD with matched reference groups. MATERIALS AND METHODS Nationwide Danish healthcare registers were used to include all patients (≥18 years) diagnosed with biopsy-verified NAFLD (1997-2021). Patients were classified as having simple steatosis or non-alcoholic steatohepatitis (NASH) with or without cirrhosis, and all matched with liver-disease free reference groups. Healthcare costs and labour market outcomes were compared from 5 years before to 11 years after diagnosis. Patients were followed for 25 years to analyse risk of disability insurance and death. RESULTS 3,712 patients with biopsy-verified NASH (n = 1,030), simple steatosis (n = 1,540) or cirrhosis (n = 1,142) were identified. The average total costs in the year leading up to diagnosis was 4.1-fold higher for NASH patients than the reference group (EUR 6,318), 6.2-fold higher for cirrhosis patients and 3.1-fold higher for simple steatosis patients. In NASH, outpatient hospital contacts were responsible for 49 % of the excess costs (EUR 3,121). NASH patients had statistically significantly lower income than their reference group as early as five years before diagnosis until nine years after diagnosis, and markedly higher risk of becoming disability insurance recipients (HR: 4.37; 95 % CI: 3.17-6.02) and of death (HR: 2.42; 95 % CI: 1.80-3.25). CONCLUSIONS NASH, simple steatosis and cirrhosis are all associated with substantial costs for the individual and the society with excess healthcare costs and poorer labour market outcomes.
Collapse
Affiliation(s)
| | - Lise Lotte Gluud
- Copenhagen University Hospital Hvidovre, Gastro Unit, DK-2650 Hvidovre, Denmark
| | - Henning Grønbæk
- Aarhus University Hospital, Department of Hepatology & Gastroenterology, DK-8200 Aarhus, Denmark; University of Aarhus, Department of Clinical Medicine, DK-8200 Aarhus, Denmark
| | - Majken K Jensen
- University of Copenhagen, Department of Public Health, Section of Epidemiology, Copenhagen, Denmark
| | - Mogens Vyberg
- Aalborg University Campus Copenhagen, Department of Clinical Medicine, DK-2450 Copenhagen, Denmark; Copenhagen University Hvidovre, Department of Pathology, DK-2650 Hvidovre, Denmark
| | | | | | - Nanna Hovelsø
- Pfizer Denmark Aps, Medical Affairs, DK-2750 Ballerup, Denmark
| | | | | | - Peter Jepsen
- Aarhus University Hospital, Department of Hepatology & Gastroenterology, DK-8200 Aarhus, Denmark; University of Aarhus, Department of Clinical Medicine, DK-8200 Aarhus, Denmark
| |
Collapse
|
48
|
Tyczyńska M, Hunek G, Szczasny M, Brachet A, Januszewski J, Forma A, Portincasa P, Flieger J, Baj J. Supplementation of Micro- and Macronutrients-A Role of Nutritional Status in Non-Alcoholic Fatty Liver Disease. Int J Mol Sci 2024; 25:4916. [PMID: 38732128 PMCID: PMC11085010 DOI: 10.3390/ijms25094916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 04/28/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a condition in which the pathological cumulation of fat with coexisting inflammation and damage of hepatic cells leads to progressive dysfunctions of the liver. Except for the commonly well-known major causes of NAFLD such as obesity, dyslipidemia, insulin resistance, or diabetes, an unbalanced diet and imbalanced nutritional status should also be taken into consideration. In this narrative review, we summarized the current knowledge regarding the micro- and macronutrient status of patients suffering from NAFLD considering various diets and supplementation of chosen supplements. We aimed to summarize the knowledge indicating which nutritional impairments may be associated with the onset and progression of NAFLD at the same time evaluating the potential therapy targets that could facilitate the healing process. Except for the above-mentioned objectives, one of the most important aspects of this review was to highlight the possible strategies for taking care of NAFLD patients taking into account the challenges and opportunities associated with the micronutrient status of the patients. The current research indicates that a supplementation of chosen vitamins (e.g., vitamin A, B complex, C, or D) as well as chosen elements such as zinc may alleviate the symptoms of NAFLD. However, there is still a lack of sufficient data regarding healthy ranges of dosages; thus, further research is of high importance in this matter.
Collapse
Affiliation(s)
- Magdalena Tyczyńska
- Department of Correct, Clinical and Imaging Anatomy, Medical University of Lublin, Jaczewskiego 4, 20-090 Lublin, Poland;
| | - Gabriela Hunek
- Chair and Department of Forensic Medicine, Medical University of Lublin, Jaczewskiego 8b, 20-090 Lublin, Poland; (G.H.); (A.B.)
| | - Martyna Szczasny
- Chair and Department of Anatomy, Medical University of Lublin, Jaczewskiego 4, 20-090 Lublin, Poland; (M.S.); (J.J.)
| | - Adam Brachet
- Chair and Department of Forensic Medicine, Medical University of Lublin, Jaczewskiego 8b, 20-090 Lublin, Poland; (G.H.); (A.B.)
| | - Jacek Januszewski
- Chair and Department of Anatomy, Medical University of Lublin, Jaczewskiego 4, 20-090 Lublin, Poland; (M.S.); (J.J.)
| | - Alicja Forma
- Chair and Department of Forensic Medicine, Medical University of Lublin, Jaczewskiego 8b, 20-090 Lublin, Poland; (G.H.); (A.B.)
| | - Piero Portincasa
- Clinica Medica “A. Murri”, Department of Biomedical Sciences & Human Oncology, University of Bari Medical School, 70124 Bari, Italy;
| | - Jolanta Flieger
- Department of Analytical Chemistry, Medical University of Lublin, Chodźki 4A, 20-093 Lublin, Poland;
| | - Jacek Baj
- Chair and Department of Anatomy, Medical University of Lublin, Jaczewskiego 4, 20-090 Lublin, Poland; (M.S.); (J.J.)
| |
Collapse
|
49
|
Lee EH, Kim JY, Yang HR. Sex-specific differences in ectopic fat and metabolic characteristics of paediatric nonalcoholic fatty liver disease. Int J Obes (Lond) 2024; 48:486-494. [PMID: 38114813 DOI: 10.1038/s41366-023-01439-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 11/22/2023] [Accepted: 12/01/2023] [Indexed: 12/21/2023]
Abstract
BACKGROUND/OBJECTIVES Sex-specific differences in obesity-related metabolic characteristics of non-alcoholic fatty liver disease (NAFLD) have rarely been explored, particularly in children with biopsy-verified NAFLD. The influence of sex hormones on ectopic fat disposition may cause inter-sex differences in various metabolic factors. This study aimed to assess the sex-based differences in ectopic fat and metabolic characteristics in children with NAFLD. SUBJECT/METHODS We enrolled 63 children with biopsy-verified NAFLD (48 boys; mean age, 12.9 ± 3.2 years; mean body mass index z-score [BMI-z], 2.49 ± 1.21). Ectopic fat in the liver and pancreas was quantified based on magnetic resonance imaging within 2 days of the liver biopsy. Laboratory tests, body composition, blood pressure, and anthropometric measurements were also assessed. RESULTS Sex-based differences were neither observed in age, BMI-z, or total body fat percentage nor in the proportions of obesity, abdominal obesity, diabetes, dyslipidaemia, hypertension, or metabolic syndrome. Furthermore, liver enzyme levels, lipid profiles, and pancreatic fat did not differ between the sexes. However, boys had significantly higher fasting insulin (median 133.2 vs. 97.8 pmol/L; p = 0.039), fasting plasma glucose (median 5.30 vs. 4.83 mmol/L; p = 0.013), homeostasis model assessment of insulin resistance (median 5.4 vs. 3.6; p = 0.025), serum uric acid (404.1 ± 101.2 vs. 322.4 ± 87.1 μmol/L; p = 0.009), and liver fat (median 26.3% vs. 16.3%; p = 0.014). CONCLUSIONS Male-predominant hepatic steatosis and insulin resistance caused by sex-specific ectopic fat accumulation may contribute to higher uric acid levels in boys than in girls with NAFLD.
Collapse
Affiliation(s)
- Eun Hye Lee
- Department of Pediatrics, Nowon Eulji Medical Center, Eulji University School of Medicine, Seoul, South Korea
| | - Ji Young Kim
- Department of Radiology, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Hye Ran Yang
- Department of Pediatrics, Seoul National University Bundang Hospital, Seongnam, South Korea.
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, South Korea.
| |
Collapse
|
50
|
Yang R, Chen Z, Pan J, Yang S, Hu F. Non-contrast T1ρ dispersion versus Gd-EOB-DTPA-enhanced T1mapping for the risk stratification of non-alcoholic fatty liver disease in rabbit models. Magn Reson Imaging 2024; 107:130-137. [PMID: 38278311 DOI: 10.1016/j.mri.2024.01.013] [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: 09/14/2023] [Revised: 01/21/2024] [Accepted: 01/23/2024] [Indexed: 01/28/2024]
Abstract
PURPOSE To investigate the diagnostic efficacy of T1ρ dispersion and Gd-EOB-DTPAenhanced T1mapping in the identification of early liver fibrosis (LF) and non-alcoholic steatohepatitis (NASH) in a non-alcoholic fatty liver disease (NAFLD) rabbit model induced by a high-fat diet using histopathological findings as the standard reference. METHODS A total of sixty rabbits were randomly allocated into the standard control group (n = 12) and the NAFLD model groups (8 rabbits per group) corresponding to different high-fat high cholesterol diet feeding weeks. All rabbits underwent noncontrast transverse T1ρ mapping with varying spin-locking frequencies (FSL = 0 Hz and 500 Hz), native T1 mapping, and Gd-EOB-DTPA-enhanced T1 mapping during the hepatobiliary phase. The histopathological findings were assessed based on the NASH CRN Scoring System. Statistical analyses were conducted using the intraclass correlation coefficient, analysis of variance, multiple linear regression, and receiver operating characteristics. RESULTS Except for native T1, T1ρ, T1ρ dispersion, HBP T1, and △T1 values significantly differed among different liver fibrosis groups (F = 14.414, 18.736, 10.15, and 9.799, respectively; all P < 0.05). T1ρ, T1ρ dispersion, HBP T1, and △T1 values also exhibited significant differences among different NASH groups (F = 4.138, 4.594, 21.868, and 22.678, respectively; all P < 0.05). In the multiple regression analysis, liver fibrosis was the only factor that independently influenced T1ρ dispersion (R2 = 0.746, P = 0.000). Among all metrics, T1ρ dispersion demonstrated the best area under curve (AUC) for identifying early LF (≥ F1 stage) and significant LF (≥ F2 stage) (AUC, 0.849 and 0.916, respectively). The performance of △T1 and HBP T1 (AUC, 0.948 and 0.936, respectively) were better than that of T1ρ and T1ρ dispersion (AUC, 0.762 and 0.769, respectively) for diagnosing NASH. CONCLUSION T1⍴ dispersion may be suitable for detecting liver fibrosis in the complex background of NAFLD, while Gd-EOB-DTPA enhanced T1 mapping is superior to nonenhanced T1⍴ mapping (T1⍴ and T1⍴ dispersion) for identifying NASH.
Collapse
Affiliation(s)
- Ru Yang
- Department of Radiology, The First Affiliated Hospital of Chengdu Medical College, No.278, Baoguang Road, Xindu District, Chengdu, Sichuan, China
| | - Zhongshan Chen
- Department of Radiology, The First Affiliated Hospital of Chengdu Medical College, No.278, Baoguang Road, Xindu District, Chengdu, Sichuan, China
| | - Jin Pan
- Department of Radiology, The First Affiliated Hospital of Chengdu Medical College, No.278, Baoguang Road, Xindu District, Chengdu, Sichuan, China
| | - Shimin Yang
- Shanghai United Imaging Healthcare Co., Ltd., No.2258, Chengbei Road, Shanghai, China
| | - Fubi Hu
- Department of Radiology, The First Affiliated Hospital of Chengdu Medical College, No.278, Baoguang Road, Xindu District, Chengdu, Sichuan, China.
| |
Collapse
|