1
|
Hu P, Miao H, Li M, Zhou R, Lou Q, Wang D, Gao J, Guo F. Identification of plasma biomarkers for non-invasive diagnosis of hepatitis B cirrhosis. J Pharm Biomed Anal 2025; 263:116909. [PMID: 40300315 DOI: 10.1016/j.jpba.2025.116909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 05/01/2025]
Abstract
Hepatitis B virus (HBV) infection represents a major public health challenge due to its potential progression to liver cirrhosis and hepatocellular carcinoma, underscoring the importance of early diagnosis for effective management. This study aimed to identify plasma biomarkers for the non-invasive diagnosis of hepatitis B cirrhosis (HBC). We employed quantitative proteomic analysis via liquid chromatography-tandem mass spectrometry on plasma samples from 27 individuals, including 13 patients with HBC and 14 with chronic hepatitis B (CHB). Bioinformatics analysis of 963 identified proteins revealed 234 differential expressed proteins, comprising 115 upregulated and 119 downregulated proteins. Four candidate biomarkers-CHI3L1, IGFBP1, SHBG, and TIMP2-were subsequently selected and validated using ELISA in a cohort of 158 patients, all demonstrating elevated levels in HBC patients. The four-biomarker panel (4MP) demonstrated superior diagnostic performance, achieving an area under the curve (AUC) of 0.902 for distinguishing HBC from CHB. For differentiating decompensated HBC from CHB, the 4MP achieved an AUC of 0.993, with a sensitivity of 93.75 % and specificity of 98.73 %. Mostly, the 4MP also performed well in identifying severe HBC from non-severe HBC, achieving an AUC of 0.911, with a sensitivity of 81.25% and specificity of 93.65%. In conclusion, this study identifies four novel plasma biomarkers for HBC, highlighting their potential to enhance non-invasive diagnostic strategies for monitoring HBC progression.
Collapse
Affiliation(s)
- Piao Hu
- Department of Infectious Diseases, The First People's Hospital of Xiaoshan District, Xiaoshan Affiliated Hospital of Wenzhou Medical University, Hangzhou 311200, China
| | - Haifeng Miao
- Department of Infectious Diseases, The First People's Hospital of Xiaoshan District, Xiaoshan Affiliated Hospital of Wenzhou Medical University, Hangzhou 311200, China
| | - Mei Li
- Department of Infectious Diseases, The First People's Hospital of Xiaoshan District, Xiaoshan Affiliated Hospital of Wenzhou Medical University, Hangzhou 311200, China
| | - Ruxue Zhou
- Cosmos Wisdom Mass Spectrometry Center of Zhejiang University Medical School, Hangzhou 311200, China; Jiaxing Key Laboratory of Clinical Laboratory Diagnostics and Translational Research, Affliated Hospital of Jiaxing University, Jiaxing 314000, China
| | - Qinqin Lou
- Cosmos Wisdom Mass Spectrometry Center of Zhejiang University Medical School, Hangzhou 311200, China; Jiaxing Key Laboratory of Clinical Laboratory Diagnostics and Translational Research, Affliated Hospital of Jiaxing University, Jiaxing 314000, China
| | - Dezhen Wang
- Cosmos Wisdom Mass Spectrometry Center of Zhejiang University Medical School, Hangzhou 311200, China; Jiaxing Key Laboratory of Clinical Laboratory Diagnostics and Translational Research, Affliated Hospital of Jiaxing University, Jiaxing 314000, China
| | - Junli Gao
- Cosmos Wisdom Mass Spectrometry Center of Zhejiang University Medical School, Hangzhou 311200, China; Jiaxing Key Laboratory of Clinical Laboratory Diagnostics and Translational Research, Affliated Hospital of Jiaxing University, Jiaxing 314000, China.
| | - Feng Guo
- Department of Gastroenterology, The First People's Hospital of Xiaoshan District, Xiaoshan Affiliated Hospital of Wenzhou Medical University, Hangzhou 311200, China.
| |
Collapse
|
2
|
Blomdahl J, Åberg M, Fridén M, Ahlström H, Hockings P, Hulthe J, Eriksson N, Gabrysch K, Nasr P, Risérus U, Kechagias S, Rorsman F, Ekstedt M, Vessby J. Proteomic signatures for fibrosis in MASLD: a biopsy-proven dual-cohort study. Scand J Gastroenterol 2025; 60:597-605. [PMID: 40237197 DOI: 10.1080/00365521.2025.2490996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/30/2025] [Accepted: 04/04/2025] [Indexed: 04/18/2025]
Abstract
OBJECTIVES Predicting disease progression in metabolic dysfunction-associated steatotic liver disease (MASLD) is challenging, and current non-invasive tests (NITs) lack the precision to replace liver biopsy. This study aimed to identify plasma biomarkers for different stages of fibrosis using affinity-based proteomics in two biopsy-proven cohorts. The primary objective was to identify biomarkers capable of distinguishing between low-to-no fibrosis (F0-1) and significant fibrosis (F2-4) in MASLD. MATERIALS AND METHODS Participants in the discovery cohort were recruited from Uppsala University Hospital and Swedish CArdioPulmonary bioImage Study (SCAPIS), while the validation cohort was included from Linköping University Hospital. All participants diagnosed with MASLD underwent liver biopsy and were categorized by fibrosis stage (F0-1 or F2-4). A total of 276 plasma proteins were analyzed using Olink® panels, with biomarkers identified through ordinal logistic regression, random forest (RF) analysis and the Boruta algorithm. RESULTS The discovery cohort included 60 participants, with 60% having fibrosis stage F0-1 and 40% having F2-4. The validation cohort had 59 participants, of whom 35 had fibrosis stage F0-1 (59.3%) and 24 had stage F2-4 (40.7%). Five biomarkers were significantly associated with fibrosis stage in the discovery cohort, with four confirmed in the validation cohort. A model combining angiotensin converting enzyme-2 (ACE2), hepatocyte growth factor (HGF) and insulin-like growth factor-binding protein-7 (IGFBP-7) demonstrated strong predictive performance for significant fibrosis (c-statistics 0.82-0.83), outperforming fibrosis-4 (FIB-4) (c-statistics 0.61-0.72). CONCLUSIONS A biomarker model including ACE2, HGF and IGFBP7 shows promise in distinguishing between low-stage and significant fibrosis.
Collapse
Affiliation(s)
- Julia Blomdahl
- Department of Medical Sciences, Gastroenterology Research Group, Uppsala University, Uppsala, Sweden
| | - Mikael Åberg
- Department of Medical Sciences, Clinical Chemistry and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Michael Fridén
- Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, Uppsala University, Uppsala, Sweden
| | - Håkan Ahlström
- Department of Surgical Sciences, Section of Radiology, Uppsala University, Uppsala, Sweden
- Antaros Medical, Mölndal, Sweden
| | | | | | - Niclas Eriksson
- Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
| | - Katja Gabrysch
- Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
| | - Patrik Nasr
- Division of Gastroenterology and Hepatology, Department of Health, Medicine, and Caring Sciences, Linköping University, Linköping, Sweden
- Wallenberg Center for Molecular Medicine, Linköping University, Linköping, Sweden
| | - Ulf Risérus
- Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, Uppsala University, Uppsala, Sweden
| | - Stergios Kechagias
- Division of Gastroenterology and Hepatology, Department of Health, Medicine, and Caring Sciences, Linköping University, Linköping, Sweden
| | - Fredrik Rorsman
- Department of Medical Sciences, Gastroenterology Research Group, Uppsala University, Uppsala, Sweden
| | - Mattias Ekstedt
- Division of Gastroenterology and Hepatology, Department of Health, Medicine, and Caring Sciences, Linköping University, Linköping, Sweden
| | - Johan Vessby
- Department of Medical Sciences, Gastroenterology Research Group, Uppsala University, Uppsala, Sweden
| |
Collapse
|
3
|
Yin W, Gao W, Yang Y, Lin W, Chen W, Zhu X, Zhu R, Zhu L, Jiao N. Disrupted host-microbiota crosstalk promotes nonalcoholic fatty liver disease progression by impaired mitophagy. Microbiol Spectr 2025:e0010025. [PMID: 40401922 DOI: 10.1128/spectrum.00100-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 04/05/2025] [Indexed: 05/23/2025] Open
Abstract
The intricate interplay between host genes and intrahepatic microbes is vital in shaping the hepatic microenvironment. This study aims to elucidate how host-microbiota interactions contribute to the progression of nonalcoholic fatty liver disease (NAFLD). Hepatic gene and microbial profiles were analyzed from 570 samples across five cohorts, including 72 control, 124 nonalcoholic fatty liver (NAFL), 143 Borderline, and 231 nonalcoholic steatohepatitis (NASH) samples. Least absolute shrinkage and selection operator penalized regression and sparse canonical correlation analysis were utilized to identify host-microbiota interactions and their function. Validation was performed using a bulk transcriptomic data set comprising 1,332 samples and a single-cell transcriptomic data set of seven samples. We observed stage-specific gene expression changes of disrupting energy metabolism and immune responses, alongside microbial shifts shaping the NAFLD microenvironment. Additionally, we identified 5,537, 1,937, 1,485, and 2,933 host-microbiota interactions in control, NAFL, Borderline, and NASH samples, respectively. Escherichia coli and Actinomyces naeslundii dominated the interaction network in control but were replaced by Sphingomonadales and Sphingomonadaceae in disease stages from NAFL, preceding the transcriptomic tipping point observed in Borderline. In NASH, interactions significantly weakened, accompanied by the loss of mutualistic interactions between bacteria such as Bacillales, Ralstonia insidiosa, Sphingomonadaceae, and host mitophagy genes including SQSTM1, OPTN, and BNIP3L. Single-cell data sets confirmed these interactions were co-localized in macrophages and monocytes in control, which shifted to hepatocytes and endothelial cells in NAFLD. Shifts in host-microbial interaction signal early microenvironment changes. Disturbed host-microbiota interactions impacting mitophagy can trigger a pro-inflammatory hepatic microenvironment, potentially driving disease progression.IMPORTANCEThis study integrated multiple cohorts to uncover fundamental and generalizable signals in the progression of nonalcoholic fatty liver disease. Key changes in both liver gene expression and microbiota were identified across disease stages, with microbial composition and interactions with host offering earlier insights into microenvironmental changes. Notably, host-microbiota interactions related to mitophagy, crucial in early stages, were destroyed in nonalcoholic steatohepatitis. This disruption may contribute to the worsening inflammation and disease progression.
Collapse
Affiliation(s)
- Wenjing Yin
- Putuo People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Wenxing Gao
- Putuo People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Yuwei Yang
- Putuo People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Weili Lin
- Putuo People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Wanning Chen
- Putuo People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Xinyue Zhu
- Putuo People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Ruixin Zhu
- Putuo People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Lixin Zhu
- Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Na Jiao
- State Key Laboratory of Genetic Engineering, Fudan Microbiome Center, School of Life Sciences, Fudan University, Shanghai, China
| |
Collapse
|
4
|
Liu J, Deng L, Yao B, Zhang Y, Huang J, Huang S, Liang C, Shen Y, Wang X. Carboxylesterase 2A gene knockout or enzyme inhibition alleviates steatohepatitis in rats by regulating PPARγ and endoplasmic reticulum stress. Free Radic Biol Med 2025; 232:279-291. [PMID: 40089078 DOI: 10.1016/j.freeradbiomed.2025.03.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/09/2025] [Accepted: 03/13/2025] [Indexed: 03/17/2025]
Abstract
Metabolic dysfunction associated steatotic liver disease (MASLD) is a widespread liver disease that progresses from simple steatosis to severe steatohepatitis stage. Despite the recognized importance of carboxylesterase 2 (CES2) in hepatic lipid metabolism, the role of CES2 in hepatic inflammation remains unclear. The rat genome encodes six Ces2 genes and Ces2a shows high expression in the liver and intestine. Lipid metabolism, inflammation, fibrosis, and endoplasmic reticulum (ER) stress were investigated in Ces2a knockout (KO) rats. KO rats showed spontaneous liver lipid accumulation due to increased lipogenesis and reduced fatty acid oxidation. Non-targeted lipidomic analysis revealed enhanced lysophosphatidylcholines (LPCs) and phosphatidylcholines (PCs) in KO rats and increased concentrations of ligands, thus activating the expression of PPARγ. Although there was simple lipid accumulation in the liver of KO rats, Ces2a deficiency showed a significant protective effect against LPS and diet-induced hepatic steatohepatitis by inhibiting ER stress regulated by PPARγ activation. In line with this, treatment with tanshinone IIA, a CES2 inhibitor, significantly alleviated the progression of steatohepatitis induced by the MCD diet. In conclusion, the increased PPARγ expression in Ces2a deficiency may counteract liver inflammation and ER stress despite the presence of simple steatosis. Therefore, CES2 inhibition represents a potential therapeutic approach for steatohepatitis.
Collapse
Affiliation(s)
- Jie Liu
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China
| | - Luyao Deng
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China
| | - Bingyi Yao
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China
| | - Yuanjin Zhang
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China
| | - Junze Huang
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China
| | - Shengbo Huang
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China
| | - Chenmeizi Liang
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China
| | - Yifei Shen
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China
| | - Xin Wang
- Changning Maternity and Infant Health Hospital and School of Life Sciences, Shanghai Key Laboratory of Regulatory Biology, East China Normal University, Shanghai, China.
| |
Collapse
|
5
|
Zeng T, Li F, Yang M, Wu Y, Cui W, Mou H, Luo X. Feasibility of Serum Galectin-1 as a Diagnostic Biomarker for Metabolic Dysfunction-Associated Steatotic Liver Disease: A Study on a Segment of the Chinese Population Using Convenience Sampling. Biomedicines 2025; 13:425. [PMID: 40002838 PMCID: PMC11853191 DOI: 10.3390/biomedicines13020425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/30/2025] [Accepted: 01/31/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD) is commonly considered as a hepatic manifestation of metabolic syndrome, posing considerable public health and economic challenges due to its high prevalence. This study investigates the diagnostic potential of serum galectin-1 levels in MASLD patients. Methods: A total of 128 participants were analyzed for this study, comprising 68 healthy controls and 60 MASLD patients. The hepatic steatosis index (HSI) and fatty liver index (FLI) were calculated to evaluate the liver steatosis. Serum galectin-1 levels were measured using an enzyme-linked immunosorbent assay. We additionally conducted a comparative analysis of galectin-1 mRNA and protein expression levels in the liver tissue between the mouse models of MASLD, including ob/ob mice (n = 6), high-fat diet-fed C57 mice (n = 6), and the control group (n = 6). Results: Average serum galectin-1 levels significantly differed between groups, with lower values in the controls (p < 0.01). The frequency of MASLD increased with higher quartiles of galectin-1 levels (p < 0.01). The correlation analysis showed a positive relationship between serum galectin-1 and both HSI and FLI (p < 0.01). The multivariate logistic regression indicated that elevated galectin-1 was associated with an increased risk of MASLD (p < 0.01), yielding an area under the receiver operating characteristic curve for predicting MASLD at 0.745 (95% CI: 0.662-0.829). Hepatic galectin-1 levels were also elevated in the MASLD mouse model at both transcript and protein levels (p < 0.01). Conclusions: Serum galectin-1 can be used as a potential biomarker to help diagnose MASLD.
Collapse
Affiliation(s)
- Ting Zeng
- Department of Laboratory Medicine, Chongqing University Three Gorges Hospital, School of Medicine, Chongqing University, Chongqing 404000, China; (T.Z.); (F.L.); (Y.W.)
| | - Fang Li
- Department of Laboratory Medicine, Chongqing University Three Gorges Hospital, School of Medicine, Chongqing University, Chongqing 404000, China; (T.Z.); (F.L.); (Y.W.)
| | - Min Yang
- Department of Laboratory Medicine, Chongqing University Three Gorges Hospital, School of Medicine, Chongqing University, Chongqing 404000, China; (T.Z.); (F.L.); (Y.W.)
| | - Yao Wu
- Department of Laboratory Medicine, Chongqing University Three Gorges Hospital, School of Medicine, Chongqing University, Chongqing 404000, China; (T.Z.); (F.L.); (Y.W.)
| | - Wei Cui
- The Center of Clinical Research of Endocrinology and Metabolic Diseases in Chongqing, Chongqing University Three Gorges Hospital, School of Medicine, Chongqing University, Chongqing 404000, China
| | - Huaming Mou
- Department of Cardiovascular Medicine, Chongqing University Three Gorges Hospital, School of Medicine, Chongqing University, Chongqing 404000, China
| | - Xiaohe Luo
- Department of Laboratory Medicine, Chongqing University Three Gorges Hospital, School of Medicine, Chongqing University, Chongqing 404000, China; (T.Z.); (F.L.); (Y.W.)
- The Center of Clinical Research of Endocrinology and Metabolic Diseases in Chongqing, Chongqing University Three Gorges Hospital, School of Medicine, Chongqing University, Chongqing 404000, China
| |
Collapse
|
6
|
Hu L, Du H, Zhou Q, Liu C, Zhang T, Yuan M. Web of Science-Based Visualization of Metabolic Dysfunction-Associated Fatty Liver Disease in Pediatric and Adolescent Populations: A Bibliometric Study. Health Sci Rep 2025; 8:e70409. [PMID: 39897463 PMCID: PMC11779742 DOI: 10.1002/hsr2.70409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 12/31/2024] [Accepted: 01/14/2025] [Indexed: 02/04/2025] Open
Abstract
Background and Aims The prevalence of metabolism-associated fatty liver disease (MAFLD) in children is on the rise. This study employs visualization and analysis to evaluate the research implications, current advancements, and emerging trends in pediatric MAFLD, with the aim of elucidating its pathogenesis and informing the development of clinical treatment strategies. Methods Using visualization software, we conducted a visual analysis and mapping of the journal distribution, leading institutions, prominent authors, annual publication trends, and keyword frequencies among the 1179 scholarly articles retrieved from the Web of Science Core Collection for this study. Results The overall publication volume demonstrated an upward trend, with a total of 200 journals, contributions from 63 countries, 882 research institutions, and 5605 authors involved, including 84 who were identified as core authors. The main research team is led by Nobili, Valerio. The main research institutions are concentrated in Italy, the United States, and China. A total of 473 keywords were included, and the keywords with high frequency and medium centricity are insulin resistance, metabolic syndrome, children, steatohepatitis, adolescents, hepatic steatosis, nash, obesity, diagnosis, and fibrosis, which resulted in six keyword clusters. Conclusion MAFLD represents a significant public health concern. Research on children and adolescents with MAFLD continues to attract high interest. Noninvasive diagnostic methods, pathogenesis (intestinal microbiota research), disease prediction (gene research) are current research hotspots.
Collapse
Affiliation(s)
- Liangyu Hu
- Science and Technology Information Center, LibraryShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Huarong Du
- Science and Technology Information Center, LibraryShanghai University of Traditional Chinese MedicineShanghaiChina
| | - QianQian Zhou
- Science and Technology Information Center, LibraryShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Chunlei Liu
- Science and Technology Information Center, LibraryShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Tiansong Zhang
- Department of Traditional Chinese Medicine, Jing'an District Central HospitalFudan UniversityShanghaiChina
| | - Min Yuan
- Science and Technology Information Center, LibraryShanghai University of Traditional Chinese MedicineShanghaiChina
| |
Collapse
|
7
|
Lin Y, Ye H, Chen Y, Zhang R, Chen Y, Ou W. Integrative Analyses of Genes of Pediatric Non-alcoholic Fatty Liver Disease Associated with Energy Metabolism. Dig Dis Sci 2024; 69:4373-4391. [PMID: 39496907 PMCID: PMC11602812 DOI: 10.1007/s10620-024-08702-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 10/18/2024] [Indexed: 11/06/2024]
Abstract
BACKGROUND Pediatric non-alcoholic fatty liver disease (NAFLD) is a chronic steatosis of the liver associated with energy metabolism in children and adolescents, failure to intervene promptly can elevate the risk of developing hepatocellular carcinoma. Therefore, this study aimed to understand the underlying mechanism of pediatric NAFLD and investigate potential biomarkers and therapeutic targets. METHODS We investigated genes using the GSE185051 data set related to energy metabolism from the GeneCards database, constructed protein-protein interaction network, identified hub genes and established networks representing interactions between these hub genes and miRNA, RNA-binding proteins, transcription factors, and drugs. Subsequently, we performed Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) functional enrichment analysis, Gene Set Enrichment Analysis (GSEA), and immune infiltration analysis. RESULTS Our analysis identified 9 hub genes through the PPI network. The target molecules were identified through the interaction network between hub genes and miRNAs, RNA-binding proteins, transcription factors, and drugs. GO analysis revealed that hub genes were associated with oxidative stress responses and other pathways. KEGG analysis highlighted their involvement in pathways such as insulin resistance, among others. GSEA revealed that hub genes were highly enriched in pathways related to Omega-9 fatty acid synthesis, among others. Immune infiltration analysis suggested that mast cells and T follicular helper cells play significant roles in the pathogenesis of NAFLD. CONCLUSION We identified the hub genes in pediatric NAFLD closely related to energy metabolism. These findings offer the potential for identifying potential novel diagnostic biomarkers, and establishing therapeutic targets for pediatric NAFLD.
Collapse
Affiliation(s)
- Yijun Lin
- Department of Pediatrics, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, No.18, Daoshan Road, Gulou District, Fuzhou, 350001, Fujian, China
- Department of Pediatrics, Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), Fuzhou, China
| | - Hong Ye
- Department of Pediatrics, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, No.18, Daoshan Road, Gulou District, Fuzhou, 350001, Fujian, China.
- Department of Pediatrics, Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), Fuzhou, China.
| | - Yan Chen
- Department of Pediatrics, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, No.18, Daoshan Road, Gulou District, Fuzhou, 350001, Fujian, China
- Department of Pediatrics, Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), Fuzhou, China
| | - Rui Zhang
- Department of Pediatrics, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, No.18, Daoshan Road, Gulou District, Fuzhou, 350001, Fujian, China
- Department of Pediatrics, Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), Fuzhou, China
| | - Yuyun Chen
- Department of Pediatrics, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, No.18, Daoshan Road, Gulou District, Fuzhou, 350001, Fujian, China
- Department of Pediatrics, Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), Fuzhou, China
| | - Weijie Ou
- Department of Pediatrics, Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), Fuzhou, China
| |
Collapse
|
8
|
Jiang L, Xu QY, Zhou YC, Xu J, Fan JG. Spatial Transcriptomics Reveals the Transcriptomic Signatures in a Mouse Model of Pediatric Metabolic Dysfunction-Associated Steatohepatitis. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:2341-2355. [PMID: 39222909 DOI: 10.1016/j.ajpath.2024.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 07/24/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024]
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) is considered the progressive form of metabolic dysfunction-associated steatotic liver disease, which is the leading cause of chronic liver disease in children. However, the pathogenesis of pediatric MASH remains poorly understood because of the lack of animal models. In this study, a mouse model of pediatric MASH was developed and its hepatic transcriptomic profile was characterized using spatial transcriptomics technology. C57BL/6J mice were fed a Western diet (WD) along with weekly injections of carbon tetrachloride (CCl4) from the age of 3 weeks and lasting up to 8 weeks. After 5 weeks of feeding, WD + CCl4-treated mice showed significant liver injury without the development of insulin resistance. Histologically, WD + CCl4 induced key features of type 2 MASH, the most common type observed in children, characterized by liver steatosis, portal inflammation, and portal fibrosis. Spatial transcriptomics analysis of liver tissues indicated that cluster 0 in the mouse from the WD + CCl4 group was enriched in pathways associated with lipid metabolism. Further investigation revealed that cytochrome p450 2E1 was the top marker gene of cluster 0, and its expression was increased in the periportal area of mice from the WD + CCl4 group. These findings suggest that this mouse model of pediatric MASH mirrors the histologic features of human MASH, and the up-regulation of cytochrome p450 2E1 may be linked to the disease pathogenesis.
Collapse
Affiliation(s)
- Lu Jiang
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Institute for Pediatric Research, Shanghai, China; Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Qing-Yang Xu
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | - Juan Xu
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian-Gao Fan
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China; Department of Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
9
|
Yang Z, Chen Z, Wang J, Li Y, Zhang H, Xiang Y, Zhang Y, Shao Z, Wu P, Lu D, Lin H, Tong Z, Liu J, Dong Q. Multiple Machine Learning Identifies Key Gene PHLDA1 Suppressing NAFLD Progression. Inflammation 2024:10.1007/s10753-024-02164-6. [PMID: 39496918 DOI: 10.1007/s10753-024-02164-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 10/04/2024] [Accepted: 10/12/2024] [Indexed: 11/06/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD) poses a serious global health threat, with its progression mechanisms not yet fully understood. While several molecular markers for NAFLD have been developed in recent years, a lack of robust evidence hampers their clinical application. Therefore, identifying novel and potent biomarkers would directly aid in the prediction, prevention, and personalized treatment of NAFLD. We downloaded NAFLD-related datasets from the Gene Expression Omnibus (GEO). Differential expression analysis and functional analysis were initially conducted. Subsequently, Weighted Gene Co-expression Network Analysis (WGCNA) and multiple machine learning strategies were employed to screen and identify key genes, and the diagnostic value was assessed using Receiver Operating Characteristic (ROC) analysis. We then explored the relationship between genes and immune cells using transcriptome data and single-cell RNA sequencing (scRNA-seq) data. Finally, we validated our findings in cell and mouse NAFLD models. We obtained 23 overlapping differentially expressed genes (DEGs) across three NAFLD datasets. Enrichment analysis revealed that DEGs were associated with Apoptosis, Parathyroid hormone synthesis, secretion and action, Colorectal cancer, p53 signaling pathway, and Biosynthesis of unsaturated fatty acids. After employing machine learning strategies, we identified one gene, pleckstrin homology like domain family A member 1 (PHLDA1), downregulated in NAFLD and showing high diagnostic accuracy. CIBERSORT analysis revealed significant associations of PHLDA1 with various immune cells. Single-cell data analysis demonstrated downregulation of PHLDA1 in NAFLD, with PHLDA1 exhibiting a significant negative correlation with macrophages. Furthermore, we found PHLDA1 to be downregulated in an in vitro hepatic steatosis cell model, and overexpression of PHLDA1 significantly reduced lipid accumulation, as well as the expression of key molecules involved in hepatic lipogenesis and fatty acid uptake, such as FASN, SCD-1, and CD36. Additionally, gene set enrichment analysis (GSEA) pathway enrichment analysis suggested that PHLDA1 may influence NAFLD progression through pathways such as Cytokine Cytokine Receptor Interaction, Ecm Receptor Interaction, Parkinson's Disease, and Ribosome pathways. Our conclusions were further validated in a mouse model of NAFLD. Our study reveals that PHLDA1 inhibits the progression of NAFLD, as overexpression of PHLDA1 significantly reduces lipid accumulation in cells and markedly decreases the expression of key molecules involved in liver lipogenesis and fatty acid uptake. Therefore, PHLDA1 may emerge as a novel potential target for future prediction, diagnosis, and targeted prevention of NAFLD.
Collapse
Affiliation(s)
- Zhenwei Yang
- Department of Gastroenterology, The Fifth School of Clinical Medicine of Zhejiang, Huzhou Central Hospital, Chinese Medical University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, People's Republic of China.
| | - Zhiqin Chen
- Department of Gastroenterology, The Fifth School of Clinical Medicine of Zhejiang, Huzhou Central Hospital, Chinese Medical University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, People's Republic of China
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, China
| | - Jingchao Wang
- Department of Biochemistry and Molecular Biology, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Shenzhen University School of Medicine, Shenzhen, China
| | - Yizhang Li
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Hailin Zhang
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yu Xiang
- Department of Gastroenterology, The Fifth School of Clinical Medicine of Zhejiang, Huzhou Central Hospital, Chinese Medical University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, People's Republic of China
| | - Yuwei Zhang
- Department of Gastroenterology, The Fifth School of Clinical Medicine of Zhejiang, Huzhou Central Hospital, Chinese Medical University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, People's Republic of China
| | - Zhaozhao Shao
- Department of Gastroenterology, The Fifth School of Clinical Medicine of Zhejiang, Huzhou Central Hospital, Chinese Medical University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, People's Republic of China
| | - Pei Wu
- Department of Gastroenterology, The Fifth School of Clinical Medicine of Zhejiang, Huzhou Central Hospital, Chinese Medical University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, People's Republic of China
| | - Ding Lu
- Department of Gastroenterology, The Fifth School of Clinical Medicine of Zhejiang, Huzhou Central Hospital, Chinese Medical University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, People's Republic of China
| | - Huajiang Lin
- Department of Gastroenterology, The Fifth School of Clinical Medicine of Zhejiang, Huzhou Central Hospital, Chinese Medical University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, People's Republic of China
| | - Zhaowei Tong
- Huzhou Key Laboratory of Precision Medicine Research and Translation for Infectious Diseases, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, 313000, China
| | - Jiang Liu
- Department of Gastroenterology, The Fifth School of Clinical Medicine of Zhejiang, Huzhou Central Hospital, Chinese Medical University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, People's Republic of China
| | - Quan Dong
- Department of Gastroenterology, The Fifth School of Clinical Medicine of Zhejiang, Huzhou Central Hospital, Chinese Medical University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, People's Republic of China
| |
Collapse
|
10
|
Verschuren L, Mak AL, van Koppen A, Özsezen S, Difrancesco S, Caspers MPM, Snabel J, van der Meer D, van Dijk AM, Rashu EB, Nabilou P, Werge MP, van Son K, Kleemann R, Kiliaan AJ, Hazebroek EJ, Boonstra A, Brouwer WP, Doukas M, Gupta S, Kluft C, Nieuwdorp M, Verheij J, Gluud LL, Holleboom AG, Tushuizen ME, Hanemaaijer R. Development of a novel non-invasive biomarker panel for hepatic fibrosis in MASLD. Nat Commun 2024; 15:4564. [PMID: 38811591 PMCID: PMC11137090 DOI: 10.1038/s41467-024-48956-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 05/20/2024] [Indexed: 05/31/2024] Open
Abstract
Accurate non-invasive biomarkers to diagnose metabolic dysfunction-associated steatotic liver disease (MASLD)-related fibrosis are urgently needed. This study applies a translational approach to develop a blood-based biomarker panel for fibrosis detection in MASLD. A molecular gene expression signature identified from a diet-induced MASLD mouse model (LDLr-/-.Leiden) is translated into human blood-based biomarkers based on liver biopsy transcriptomic profiles and protein levels in MASLD patient serum samples. The resulting biomarker panel consists of IGFBP7, SSc5D and Sema4D. LightGBM modeling using this panel demonstrates high accuracy in predicting MASLD fibrosis stage (F0/F1: AUC = 0.82; F2: AUC = 0.89; F3/F4: AUC = 0.87), which is replicated in an independent validation cohort. The overall accuracy of the model outperforms predictions by the existing markers Fib-4, APRI and FibroScan. In conclusion, here we show a disease mechanism-related blood-based biomarker panel with three biomarkers which is able to identify MASLD patients with mild or advanced hepatic fibrosis with high accuracy.
Collapse
Affiliation(s)
| | - Anne Linde Mak
- Department of Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | | | | | | | | | | | | | - Anne-Marieke van Dijk
- Department of Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Elias Badal Rashu
- Gastro Unit, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark and Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Puria Nabilou
- Gastro Unit, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark and Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Mikkel Parsberg Werge
- Gastro Unit, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark and Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Koen van Son
- Department of Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | | | - Amanda J Kiliaan
- Department of Medical Imaging, Anatomy, and Radboud Alzheimer Center, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behavior, Nijmegen, the Netherlands
| | - Eric J Hazebroek
- Department of Bariatric Surgery, Vitalys, Rijnstate Hospital, Arnhem, the Netherlands and Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands
| | - André Boonstra
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Willem P Brouwer
- Department of Gastroenterology and Hepatology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Michail Doukas
- Department of Pathology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Saurabh Gupta
- Translational Medicine, Bristol Meyers Squibb, Princeton Pike, NJ, USA
| | | | - Max Nieuwdorp
- Department of Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Joanne Verheij
- Department of Pathology, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Lise Lotte Gluud
- Gastro Unit, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark and Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Adriaan G Holleboom
- Department of Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Maarten E Tushuizen
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | | |
Collapse
|
11
|
Wang Y, Bo J, Zhao Z, Han Y, Zhang Q, Liu L. Depletion of Igfbp7 alleviates zebrafish NAFLD progression through inhibiting hepatic ferroptosis. Life Sci 2023; 332:122086. [PMID: 37714372 DOI: 10.1016/j.lfs.2023.122086] [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: 04/17/2023] [Revised: 08/31/2023] [Accepted: 09/12/2023] [Indexed: 09/17/2023]
Abstract
AIMS The global increased expression of Insulin-like growth factor binding protein 7 (IGFBP7) has been detected in non-alcoholic fatty liver disease (NAFLD) patients, however, its roles in NAFLD and the mechanism remain largely unclear. The goal of this study is to investigate the effect and mechanism of Igfbp7 using a zebrafish NAFLD model. MAIN METHODS The igfbp7-/- null zebrafish mutant and the Igfbp7 liver overexpressed (LOE) transgenic zebrafish based on Gal4/UAS system were generated by CRISPR/Cas9 and Tol2 transgenic technique, respectively. The zebrafish NAFLD models in wildtypes, igfbp7-/- mutants and Igfbp7 LOE fishes have been established by high-fat diet feeding. The Igfbp7 dynamic expression and its effects on NAFLD progression have been detected and analyzed in both human NAFLD patients and zebrafish models. And the potential mechanism has been investigated through transcriptome analysis and subsequent detection and verification. KEY FINDINGS High Igfbp7 levels in NASH and fibrosis stages have been detected in liver tissues of both human NAFLD patients and zebrafish models. Depletion of Igfbp7 significantly alleviated liver steatosis, inflammation, and fibrosis, whereas liver specific Igfbp7 overexpression dramatically exacerbated liver fibrosis in zebrafish NAFLD model. The hepatic iron deposition, lipid peroxidation products, and ferroptosis-related index were also significantly reduced at the NASH stage in the absence of Igfbp7. Igfbp7 promotes NAFLD progression through regulating ferroptosis, and Ncoa4-mediated ferritinophagy may be the pathway of Igfbp7-regulated ferroptosis. SIGNIFICANCE Igfbp7 is confirmed as an important regulator in NAFLD progression. Depleting Igfbp7 effectively alleviates zebrafish NAFLD progression by inhibiting hepatic ferroptosis, suggesting a novel potential target for NAFLD treatment.
Collapse
Affiliation(s)
- Yanqin Wang
- Department of Gastroenterology and Hepatology, The First Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Jiaqi Bo
- Department of Gastroenterology and Hepatology, The First Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Zhonghua Zhao
- Institutes of Biomedical Sciences, 1331 Local Bio-Resources and Health Industry Collaborative Innovation Center of Shanxi Province, Shanxi University, Taiyuan 030006, China
| | - Yuhang Han
- Department of Gastroenterology and Hepatology, The First Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Qianqian Zhang
- Department of Gastroenterology and Hepatology, The First Hospital of Shanxi Medical University, Taiyuan 030001, China; Experimental Center of Science and Research, The First Hospital of Shanxi Medical University, Taiyuan 030001, China; Key Laboratory of Prevention and Treatment of Liver Injury and Digestive System Neoplasms, Provincial Committee of the Medical and Health, Taiyuan 030001, China
| | - Lixin Liu
- Department of Gastroenterology and Hepatology, The First Hospital of Shanxi Medical University, Taiyuan 030001, China; Experimental Center of Science and Research, The First Hospital of Shanxi Medical University, Taiyuan 030001, China; Key Laboratory of Prevention and Treatment of Liver Injury and Digestive System Neoplasms, Provincial Committee of the Medical and Health, Taiyuan 030001, China.
| |
Collapse
|
12
|
Chokeshaiusaha K, Sananmuang T, Puthier D, Nguyen C. Cross-species analysis of differential transcript usage in humans and chickens with fatty liver disease. Vet World 2023; 16:1964-1973. [PMID: 37859957 PMCID: PMC10583885 DOI: 10.14202/vetworld.2023.1964-1973] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 08/30/2023] [Indexed: 10/21/2023] Open
Abstract
Background and Aim Fatty liver disease is a common condition, characterized by excess fat accumulation in the liver. It can contribute to more severe liver-related health issues, making it a critical concern in avian and human medicine. Apart from modifying the gene expression of liver cells, the disease also alters the expression of specific transcript isoforms, which might serve as new biological markers for both species. This study aimed to identify cross-species genes displaying differential expressions in their transcript isoforms in humans and chickens with fatty liver disease. Materials and Methods We performed differential gene expression and differential transcript usage (DTU) analyses on messenger RNA datasets from the livers of both chickens and humans with fatty liver disease. Using appropriate cross-species gene identification methods, we reviewed the acquired candidate genes and their transcript isoforms to determine their potential role in fatty liver disease's pathogenesis. Results We identified seven genes - ALG5, BRD7, DIABLO, RSU1, SFXN5, STIMATE, TJP3, and VDAC2 - and their corresponding transcript isoforms as potential candidates (false discovery rate ≤0.05). Our findings showed that these genes most likely contribute to fatty disease development and progression. Conclusion This study successfully identified novel human-chicken DTU genes in fatty liver disease. Further research is encouraged to verify the functions and regulations of these transcript isoforms as potential diagnostic markers for fatty liver disease in humans and chickens.
Collapse
Affiliation(s)
- Kaj Chokeshaiusaha
- Department of Veterinary Science, Faculty of Veterinary Medicine, Rajamangala University of Technology Tawan-OK, Chonburi, Thailand
| | - Thanida Sananmuang
- Department of Veterinary Science, Faculty of Veterinary Medicine, Rajamangala University of Technology Tawan-OK, Chonburi, Thailand
| | - Denis Puthier
- Aix-Marseille Université, INSERM, UMR 1090, TAGC, Marseille, France
| | - Catherine Nguyen
- Aix-Marseille Université, INSERM, UMR 1090, TAGC, Marseille, France
| |
Collapse
|
13
|
Nash MJ, Dobrinskikh E, Janssen RC, Lovell MA, Schady DA, Levek C, Jones KL, D’Alessandro A, Kievit P, Aagaard KM, McCurdy CE, Gannon M, Friedman JE, Wesolowski SR. Maternal Western diet is associated with distinct preclinical pediatric NAFLD phenotypes in juvenile nonhuman primate offspring. Hepatol Commun 2023; 7:e0014. [PMID: 36691970 PMCID: PMC9851700 DOI: 10.1097/hc9.0000000000000014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 10/12/2022] [Indexed: 01/25/2023] Open
Abstract
Pediatric NAFLD has distinct and variable pathology, yet causation remains unclear. We have shown that maternal Western-style diet (mWSD) compared with maternal chow diet (CD) consumption in nonhuman primates produces hepatic injury and steatosis in fetal offspring. Here, we define the role of mWSD and postweaning Western-style diet (pwWSD) exposures on molecular mechanisms linked to NAFLD development in a cohort of 3-year-old juvenile nonhuman primates offspring exposed to maternal CD or mWSD followed by CD or Western-style diet after weaning. We used histologic, transcriptomic, and metabolomic analyses to identify hepatic pathways regulating NAFLD. Offspring exposed to mWSD showed increased hepatic periportal collagen deposition but unchanged hepatic triglyceride levels and body weight. mWSD was associated with a downregulation of gene expression pathways underlying HNF4α activity and protein, and downregulation of antioxidant signaling, mitochondrial biogenesis, and PPAR signaling pathways. In offspring exposed to both mWSD and pwWSD, liver RNA profiles showed upregulation of pathways promoting fibrosis and endoplasmic reticulum stress and increased BiP protein expression with pwWSD. pwWSD increased acylcarnitines and decreased anti-inflammatory fatty acids, which was more pronounced when coupled with mWSD exposure. Further, mWSD shifted liver metabolites towards decreased purine catabolism in favor of synthesis, suggesting a mitochondrial DNA repair response. Our findings demonstrate that 3-year-old offspring exposed to mWSD but weaned to a CD have periportal collagen deposition, with transcriptional and metabolic pathways underlying hepatic oxidative stress, compromised mitochondrial lipid sensing, and decreased antioxidant response. Exposure to pwWSD worsens these phenotypes, triggers endoplasmic reticulum stress, and increases fibrosis. Overall, mWSD exposure is associated with altered expression of candidate genes and metabolites related to NAFLD that persist in juvenile offspring preceding clinical presentation of NAFLD.
Collapse
Affiliation(s)
- Michael J. Nash
- Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Evgenia Dobrinskikh
- Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Rachel C. Janssen
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Mark A. Lovell
- Department of Pathology & Laboratory Medicine, Children’s Hospital Colorado, Aurora, Colorado, USA
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Deborah A. Schady
- Department of Pathology & Immunology, Baylor College of Medicine, Texas Children’s Hospital, Houston, Texas, USA
| | - Claire Levek
- Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Kenneth L. Jones
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Angelo D’Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Paul Kievit
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, USA
| | - Kjersti M. Aagaard
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Baylor College of Medicine, Texas Children’s Hospital, Houston, Texas, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Texas Children’s Hospital, Houston, Texas, USA
- Department of Molecular and Cell Biology, Baylor College of Medicine, Texas Children’s Hospital, Houston, Texas, USA
| | - Carrie E. McCurdy
- Department of Human Physiology, University of Oregon, Eugene, Oregon, USA
| | - Maureen Gannon
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jacob E. Friedman
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Stephanie R. Wesolowski
- Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
14
|
Zhang X, Zuo R, Xiao S, Wang L. Association between iron metabolism and non-alcoholic fatty liver disease: results from the National Health and Nutrition Examination Survey (NHANES 2017-2018) and a controlled animal study. Nutr Metab (Lond) 2022; 19:81. [PMID: 36514155 PMCID: PMC9749311 DOI: 10.1186/s12986-022-00715-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 11/30/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Iron metabolism may be involved in the pathogenesis of the non-alcoholic fatty liver disease (NAFLD). The relationship between iron metabolism and NAFLD has not been clearly established. This study aimed to clarify the relationship between biomarkers of iron metabolism and NAFLD. METHODS Based on the National Health and Nutrition Examination Survey (NHANES), restricted cubic spline models and multivariable logistic regression were used to examine the association between iron metabolism [serum iron (SI), serum ferritin (SF), transferrin saturation (TSAT), and soluble transferrin receptor (sTfR)] and the risk for NAFLD. In addition, stratified subgroup analysis was performed for the association between TSAT and NAFLD. Moreover, serum TSAT levels were determined in male mice with NAFLD. The expression of hepcidin and ferroportin, vital regulators of iron metabolism, were analyzed in the livers of mice by quantitative real-time PCR (qRT-PCR) and patients with NAFLD by microarray collected from the GEO data repository. RESULTS Patients with NAFLD showed decreased SI, SF, and TSAT levels and increased STfR levels based on the NHANES. After adjusting for confounding factors, TSAT was significantly negatively correlated with NAFLD. Of note, the relationship between TSAT and NAFLD differed in the four subgroups of age, sex, race, and BMI (P for interaction < 0.05). Consistently, mice with NAFLD exhibited decreased serum TSAT levels. Decreased hepcidin and increased ferroportin gene expression were observed in the livers of patients and mice with NAFLD. CONCLUSION Serum TSAT levels and hepatic hepcidin expression were decreased in both patients and mice with NAFLD. Among multiple biomarkers of iron metabolism, lower TSAT levels were significantly associated with a higher risk of NAFLD in the U.S. general population. These findings might provide new ideas for the prediction, diagnosis, and mechanistic exploration of NAFLD.
Collapse
Affiliation(s)
- Xinxin Zhang
- grid.254147.10000 0000 9776 7793School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198 China
| | - Ronghua Zuo
- grid.412676.00000 0004 1799 0784Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 Jiangsu China
| | - Shengjue Xiao
- grid.263826.b0000 0004 1761 0489Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009 China
| | - Lirui Wang
- grid.41156.370000 0001 2314 964XInstitute of Modern Biology, Nanjing University, 22 Hankou Road, Gulou, Nanjing, 210093 China
| |
Collapse
|
15
|
Han YH, Choi H, Kim HJ, Lee MO. Chemotactic cytokines secreted from Kupffer cells contribute to the sex-dependent susceptibility to non-alcoholic fatty liver diseases in mice. Life Sci 2022; 306:120846. [PMID: 35914587 DOI: 10.1016/j.lfs.2022.120846] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 07/25/2022] [Accepted: 07/26/2022] [Indexed: 12/15/2022]
Abstract
AIMS The global prevalence of non-alcoholic fatty liver disease (NAFLD) has rapidly increased over the last decade due to an elevated occurrence of metabolic syndromes. Importantly, the prevalence and severity of NAFLD is higher in men than in women. Therefore, in the present study we endeavored to identify the mechanistic disparity between male and female mice. MAIN METHODS Global gene transcriptomics analysis was done with the high-fat diet (HFD)-induced NAFLD model of male, female, and ovariectomized (OVX) female mice. The expression of CCL2, CXCL2, and CXCL10 in mRNA level and serum protein level was done by qPCR and ELISA each. Immunohistochemistry staining was used to observe hepatic immune cell infiltration. To analyzing portion of immune cells, flow cytometry was done with isolated liver cells from HFD-fed male and female mice. Primary mouse liver cells were isolated from male and female mice for in vitro studies. KEY FINDINGS We identified sex differences in inflammatory chemokines, CCL2, CXCL2, and CXCL10, with the expression of these chemokines enhanced in male and OVX, but not in female, mice after HFD feeding. Resident Kupffer cells (KCs) were identified as the major source of production of CCL2, CXCL2, and CXCL10 in the mouse NAFLD model. Notably, KCs obtained from male mice expressed higher levels of chemokines than those from female mice, indicating that KCs may mediate the sex discrepancy in NAFLD progression. SIGNIFICANCE Our findings offer new insights into the pathology of sex-specific differences in NAFLD, involving chemokines and KCs.
Collapse
Affiliation(s)
- Yong-Hyun Han
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea; College of Pharmacy, Kangwon National University, Chuncheon, Republic of Korea.
| | - Haena Choi
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea.
| | - Hyeon-Ji Kim
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea; Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea.
| | - Mi-Ock Lee
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea; Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea; Bio-MAX institute, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|