1
|
Alpízar Salazar M, Olguín Reyes SE, Medina Estévez A, Saturno Lobos JA, De Aldecoa Castillo JM, Carrera Aguas JC, Alaniz Monreal S, Navarro Rodríguez JA, Alpízar Sánchez DMF. Natural History of Metabolic Dysfunction-Associated Steatotic Liver Disease: From Metabolic Syndrome to Hepatocellular Carcinoma. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:88. [PMID: 39859069 PMCID: PMC11766802 DOI: 10.3390/medicina61010088] [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: 12/10/2024] [Revised: 12/30/2024] [Accepted: 01/04/2025] [Indexed: 01/27/2025]
Abstract
Introduction: Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD) stems from disrupted lipid metabolism in the liver, often linked to obesity, type 2 diabetes, and dyslipidemia. In Mexico, where obesity affects 36.9% of adults, MASLD prevalence has risen, especially with metabolic syndrome affecting 56.31% by 2018. MASLD can progress to Metabolic Dysfunction-Associated Steatohepatitis (MASH), affecting 5.27% globally, leading to severe complications like cirrhosis and hepatocellular carcinoma. Background: Visceral fat distribution varies by gender, impacting MASLD development due to hormonal influences. Insulin resistance plays a central role in MASLD pathogenesis, exacerbated by high-fat diets and specific fatty acids, leading to hepatic steatosis. Lipotoxicity from saturated fatty acids further damages hepatocytes, triggering inflammation and fibrosis progression in MASH. Diagnosing MASLD traditionally involves invasive liver biopsy, but non-invasive methods like ultrasound and transient elastography are preferred due to their safety and availability. These methods detect liver steatosis and fibrosis with reasonable accuracy, offering alternatives to biopsy despite varying sensitivity and specificity. Conclusions: MASLD as a metabolic disorder underscores its impact on public health, necessitating improved awareness and early management strategies to mitigate its progression to severe liver diseases.
Collapse
Affiliation(s)
- Melchor Alpízar Salazar
- Endocrinology, Specialized Center for Diabetes, Obesity and Prevention of Cardiovascular Diseases (CEDOPEC), Mexico City 11650, Mexico
| | - Samantha Estefanía Olguín Reyes
- Clinical Research, Specialized Center for Diabetes, Obesity and Prevention of Cardiovascular Diseases (CEDOPEC), Mexico City 11650, Mexico; (S.E.O.R.); (A.M.E.); (J.A.S.L.); (S.A.M.); (J.A.N.R.); (D.M.F.A.S.)
| | - Andrea Medina Estévez
- Clinical Research, Specialized Center for Diabetes, Obesity and Prevention of Cardiovascular Diseases (CEDOPEC), Mexico City 11650, Mexico; (S.E.O.R.); (A.M.E.); (J.A.S.L.); (S.A.M.); (J.A.N.R.); (D.M.F.A.S.)
| | - Julieta Alejandra Saturno Lobos
- Clinical Research, Specialized Center for Diabetes, Obesity and Prevention of Cardiovascular Diseases (CEDOPEC), Mexico City 11650, Mexico; (S.E.O.R.); (A.M.E.); (J.A.S.L.); (S.A.M.); (J.A.N.R.); (D.M.F.A.S.)
| | - Jesús Manuel De Aldecoa Castillo
- Clinical Nutrition, Specialized Center for Diabetes, Obesity and Prevention of Cardiovascular Diseases (CEDOPEC), Mexico City 11650, Mexico;
| | - Juan Carlos Carrera Aguas
- Clinical Nutrition, Specialized Center for Diabetes, Obesity and Prevention of Cardiovascular Diseases (CEDOPEC), Mexico City 11650, Mexico;
| | - Samary Alaniz Monreal
- Clinical Research, Specialized Center for Diabetes, Obesity and Prevention of Cardiovascular Diseases (CEDOPEC), Mexico City 11650, Mexico; (S.E.O.R.); (A.M.E.); (J.A.S.L.); (S.A.M.); (J.A.N.R.); (D.M.F.A.S.)
| | - José Antonio Navarro Rodríguez
- Clinical Research, Specialized Center for Diabetes, Obesity and Prevention of Cardiovascular Diseases (CEDOPEC), Mexico City 11650, Mexico; (S.E.O.R.); (A.M.E.); (J.A.S.L.); (S.A.M.); (J.A.N.R.); (D.M.F.A.S.)
| | - Dulce María Fernanda Alpízar Sánchez
- Clinical Research, Specialized Center for Diabetes, Obesity and Prevention of Cardiovascular Diseases (CEDOPEC), Mexico City 11650, Mexico; (S.E.O.R.); (A.M.E.); (J.A.S.L.); (S.A.M.); (J.A.N.R.); (D.M.F.A.S.)
| |
Collapse
|
2
|
Weng J, Cheng Q, Yang J, Jin H, Zhang R, Guan J, Ma Y, Wang L, Chen C, Wang Z. Gal-1-mediated cytochrome p450 activation promotes fibroblast into myofibroblast differentiation in pulmonary fibrosis. Int Immunopharmacol 2024; 141:112920. [PMID: 39137631 DOI: 10.1016/j.intimp.2024.112920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 07/31/2024] [Accepted: 08/07/2024] [Indexed: 08/15/2024]
Abstract
Pulmonary fibrosis (PF) results from excessive extracellular matrix (ECM) deposition and tissue remodeling after activation of fibroblasts into myofibroblasts. Abnormally deposited fibrotic ECM, in turn, promotes fibroblast activation and accelerates loss of lung structure and function. However, the molecular mediators and exact mechanisms by which fibrotic ECM promotes fibroblast activation are unclear. In a bleomycin-induced PF mouse model, we found Galectin-1 (Gal-1) expression was significantly increased in lung tissue, and overexpression of Gal-1 plasmid-transfected fibroblasts were activated into myofibroblasts. Using the decellularization technique to prepare decellularized fibrotic ECM and constructing a 3D in vitro co-culture system with fibroblasts, we found that decellularized fibrotic ECM induced a high expression of Gal-1 and promoted the activation of fibroblasts into myofibroblasts. Therefore, Gal-1 has been identified as a pivotal mediator in PF. Further, we found that decellularized fibrotic ECM delivered mechanical signals to cells through the Gal-1-mediated FAK-Src-P130Cas mechanical signalling pathway, while the CYP450 enzymes (mainly involved in CYP1A1, CYP24A1, CYP3A4, and CYP2D6 isoforms) acted as a chemical signalling pathway to receive mechanical signals transmitted from upstream Gal-1, thereby promoting fibroblast activation. The Gal-1 inhibitor OTX008 or the CYP1A1 inhibitor 7-Hydroxyflavone prevented PF in mice and inhibited the role of fibrotic ECM in promoting fibroblast activation into myofibroblasts, preventing PF. These results reveal novel molecular mechanisms of lung fibrosis formation and identify Gal-1 and its downstream CYP1A1 as potential therapeutic targets for PF disease treatmnts.
Collapse
Affiliation(s)
- Jie Weng
- Department of General Practice, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Wenzhou Key Laboratory of Precision General Practice and Health Management, Wenzhou 325000, China; South Zhejiang Institute of Radiation Medicine and Nuclear Technology, Wenzhou 325014, China
| | - Qianhui Cheng
- Department of Geriatric Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Jingwen Yang
- Department of General Practice, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Wenzhou Key Laboratory of Precision General Practice and Health Management, Wenzhou 325000, China; Department of General Practice, Taizhou Women and Children's Hospital of Wenzhou Medical University, Taizhou 318001, China
| | - Haijuan Jin
- Department of General Practice, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Theorem Clinical College of Wenzhou Medical University, Wenzhou Central Hospital, China
| | - Ran Zhang
- Department of Geriatric Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Jiangan Guan
- Department of Geriatric Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yuan Ma
- Department of Geriatric Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Liang Wang
- Department of Public Health, Robbins College of Health and Human Sciences, Baylor University, Waco, TX, USA
| | - Chan Chen
- Wenzhou Key Laboratory of Precision General Practice and Health Management, Wenzhou 325000, China; Department of Geriatric Medicine, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China.
| | - Zhiyi Wang
- Department of General Practice, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Wenzhou Key Laboratory of Precision General Practice and Health Management, Wenzhou 325000, China; South Zhejiang Institute of Radiation Medicine and Nuclear Technology, Wenzhou 325014, China; Department of General Practice, Taizhou Women and Children's Hospital of Wenzhou Medical University, Taizhou 318001, China.
| |
Collapse
|
3
|
Lee M, Son MJ, Hong S, Ryu J, Min J, Lee D, Lee JH, Kim ND, Park S, Kim D, Joo J, Kwak J, Kim KH, Lee Y, Keum B, Song HS, Jung Y, Kim KS, Kim G. Discovery of a selective cytochrome P450 4A inhibitor for the treatment of metabolic dysfunction-associated fatty liver disease. Clin Transl Med 2024; 14:e1816. [PMID: 39367660 PMCID: PMC11452733 DOI: 10.1002/ctm2.1816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 07/30/2024] [Accepted: 08/05/2024] [Indexed: 10/06/2024] Open
Affiliation(s)
- Minji Lee
- Digital Omics Research CenterKorea Basic Science Institute (KBSI)CheongjuRepublic of Korea
| | - Myung Jin Son
- Stem Cell Convergence Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB)DaejeonRepublic of Korea
- Department of Functional GenomicsKorea University of Science & Technology (UST)DaejeonRepublic of Korea
- School of PharmacySungkyunkwan UniversitySuwonRepublic of Korea
| | - Sin‐Hyoung Hong
- Digital Omics Research CenterKorea Basic Science Institute (KBSI)CheongjuRepublic of Korea
| | - Jae‐Sung Ryu
- New Drug Development CenterOsong Medical Innovation FoundationCheongjuRepublic of Korea
| | - Ji‐Hyeon Min
- Digital Omics Research CenterKorea Basic Science Institute (KBSI)CheongjuRepublic of Korea
- Department of Analytical Science and TechnologyGraduate School of Analytical Science and Technology (GRAST)Chungnam National UniversityDaejeonRepublic of Korea
| | - Dong‐Eon Lee
- Digital Omics Research CenterKorea Basic Science Institute (KBSI)CheongjuRepublic of Korea
- Department of Bio‐Analytical ScienceUniversity of Science and Technology (UST)DaejeonRepublic of Korea
| | - Ji Hoon Lee
- New Drug Development CenterDaegu‐Gyeongbuk Medical Innovation FoundationDaeguRepublic of Korea
| | - Nam Doo Kim
- New Drug Discovery & DevelopmentVORONOIBIO Inc.IncheonRepublic of Korea
| | - Shi‐Young Park
- Korea Mouse Metabolic Phenotyping CenterLee Gil Ya Cancer and Diabetes InstituteGachon UniversityIncheonSouth Korea
| | - Darong Kim
- New Drug Development CenterDaegu‐Gyeongbuk Medical Innovation FoundationDaeguRepublic of Korea
| | - Jeongmin Joo
- New Drug Development CenterDaegu‐Gyeongbuk Medical Innovation FoundationDaeguRepublic of Korea
| | - Jisung Kwak
- Sensor System Research CenterKorea Institute of Science and Technology (KIST)SeoulRepublic of Korea
- Department of Chemical and Biological EngineeringKorea UniversitySeoulRepublic of Korea
| | - Kook Hwan Kim
- Discovery DivisionGI Innovation, Inc.SeoulSouth Korea
| | - Yong‐Ho Lee
- Department of Internal MedicineYonsei University College of MedicineSeoulSouth Korea
| | - Byeong‐Rak Keum
- Department of Life SciencesPohang University of Science and TechnologyPohangRepublic of Korea
| | - Hyun Seok Song
- Sensor System Research CenterKorea Institute of Science and Technology (KIST)SeoulRepublic of Korea
| | - Youngae Jung
- Integrated Metabolomics Research GroupMetropolitan Seoul CenterKorea Basic Science Institute (KBSI)SeoulRepublic of Korea
| | - Koon Soon Kim
- Division of Endocrinology and MetabolismDaejeon Endo Internal MedicineDaejeonRepublic of Korea
| | - Gun‐Hwa Kim
- Digital Omics Research CenterKorea Basic Science Institute (KBSI)CheongjuRepublic of Korea
- Department of Analytical Science and TechnologyGraduate School of Analytical Science and Technology (GRAST)Chungnam National UniversityDaejeonRepublic of Korea
- Department of Bio‐Analytical ScienceUniversity of Science and Technology (UST)DaejeonRepublic of Korea
- Research and Development CenterCYPHARMADaejeonRepublic of Korea
| |
Collapse
|
4
|
Wilson RB, Chen YJ, Zhang R, Maini S, Andrews TS, Wang R, Borradaile NM. Elongation factor 1A1 inhibition elicits changes in lipid droplet size, the bulk transcriptome, and cell type-associated gene expression in MASLD mouse liver. Am J Physiol Gastrointest Liver Physiol 2024; 327:G608-G622. [PMID: 39136056 PMCID: PMC11482270 DOI: 10.1152/ajpgi.00276.2023] [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: 11/18/2023] [Revised: 07/19/2024] [Accepted: 07/31/2024] [Indexed: 09/19/2024]
Abstract
Eukaryotic elongation factor 1A1 (EEF1A1), originally identified for its role in protein synthesis, has additional functions in diverse cellular processes. Of note, we previously discovered a role for EEF1A1 in hepatocyte lipotoxicity. We also demonstrated that a 2-wk intervention with the EEF1A1 inhibitor didemnin B (DB) (50 µg/kg) decreased liver steatosis in a mouse model of obesity and metabolic dysfunction-associated steatotic liver disease (MASLD) [129S6/SvEvTac mice fed Western diet (42% fat) for 26 wk]. Here, we further characterized the hepatic changes occurring in these mice by assessing lipid droplet (LD) size, bulk differential expression, and cell type-associated alterations in gene expression. Consistent with the previously demonstrated decrease in hepatic steatosis, we observed decreased median LD size in response to DB. Bulk RNA sequencing (RNA-Seq) followed by gene set enrichment analysis revealed alterations in pathways related to energy metabolism and proteostasis in DB-treated mouse livers. Deconvolution of bulk data identified decreased cell type association scores for cholangiocytes, mononuclear phagocytes, and mesenchymal cells in response to DB. Overrepresentation analyses of bulk data using cell type marker gene sets further identified hepatocytes and cholangiocytes as the primary contributors to bulk differential expression in response to DB. Thus, we show that chemical inhibition of EEF1A1 decreases hepatic LD size and decreases gene expression signatures associated with several liver cell types implicated in MASLD progression. Furthermore, changes in hepatic gene expression were primarily attributable to hepatocytes and cholangiocytes. This work demonstrates that EEF1A1 inhibition may be a viable strategy to target aspects of liver biology implicated in MASLD progression.NEW & NOTEWORTHY Chemical inhibition of EEF1A1 decreases hepatic lipid droplet size and decreases gene expression signatures associated with liver cell types that contribute to MASLD progression. Furthermore, changes in hepatic gene expression are primarily attributable to hepatocytes and cholangiocytes. This work highlights the therapeutic potential of targeting EEF1A1 in the setting of MASLD, and the utility of RNA-Seq deconvolution to reveal valuable information about tissue cell type composition and cell type-associated gene expression from bulk RNA-Seq data.
Collapse
Affiliation(s)
- Rachel B Wilson
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- Children's Health Research Institute, Lawson Health Research Institute, London, Ontario, Canada
| | - Yun Jin Chen
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Richard Zhang
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Siddhant Maini
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Tallulah S Andrews
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Rennian Wang
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- Children's Health Research Institute, Lawson Health Research Institute, London, Ontario, Canada
| | - Nica M Borradaile
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| |
Collapse
|
5
|
Gao Y, Sun YY, Bai D, Wu XX. Mechanism of the components compatibility of Scutellariae Radix and Coptidis Rhizoma on mice with hyperlipidemia by regulating the Cyp4a family. JOURNAL OF ETHNOPHARMACOLOGY 2024; 331:118263. [PMID: 38677575 DOI: 10.1016/j.jep.2024.118263] [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: 12/26/2023] [Revised: 03/24/2024] [Accepted: 04/24/2024] [Indexed: 04/29/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Scutellaria baicalensis Georgi (Scutellariae Radix, SR) and Coptis chinensis Franch (Coptidis Rhizoma, CR) is a classic herbal pair used in many Traditional Chinese Medicine formulations in the treatment of hyperlipidemia (HLP). As effective ingredients of the drug pair, the effects and mechanisms of berberine and baicalin in the treatment of HLP in the form of components compatibility are still unclear. AIM OF THE STUDY To explore the mechanism of the components compatibility of SR and CR in the treatment of HLP. MATERIALS AND METHODS The HLP model was established by a high-fat diet. Serum biochemical indexes were detected. Transcriptomics and metabolomics were detected. RT-PCR and Western Blot were used to analyze the effect of RA on the expression of the Cyp4a family during the treatment of HLP. RESULTS Berberine-baicalin (RA) has a good effect in the treatment of HLP. RA can significantly reduce the body weight and liver weight of HLP, reduce the levels of total cholesterol (TC), triglyceride (TG), low-density lipoprotein (LDL-C), and increase the level of high-density lipoprotein (HDL-C). Through transcriptomic analysis, RA significantly reversed the gene expression of Cyp4a10, Cyp4a12 b, Cyp4a31, and Cyp4a32 in cytochrome P450 family 4 subfamily a (Cyp4a) which related to fatty acid degradation in the liver of HLP mice. The results of fatty acid detection showed that RA could significantly regulate heptanoic acid, EPA, adrenic acid, DH-γ-linolenic acid, and DPA in the cecum of HLP mice. The Cyp4a family genes regulated by RA are closely related to a variety of fatty acids regulated by RA. RT-PCR confirmed that RA could regulate Cyp4a mRNA expression in HLP mice. WB also showed that RA can regulate the protein expression level of Cyp4a. CONCLUSION The components compatibility of SR and CR can effectively improve the blood lipid level of HLP mice, its mechanism may be related to regulating Cyp4a gene expression and affecting fatty acid degradation, regulating the level of fatty acid metabolism in the body.
Collapse
Affiliation(s)
- Yuan Gao
- The Institute of Basic Theory of Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Yang-Yang Sun
- The Institute of Basic Theory of Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Dong Bai
- The Institute of Basic Theory of Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Xiao-Xia Wu
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| |
Collapse
|
6
|
Yin K, Büttner M, Deligiannis IK, Strzelecki M, Zhang L, Talavera-López C, Theis F, Odom DT, Martinez-Jimenez CP. Polyploidisation pleiotropically buffers ageing in hepatocytes. J Hepatol 2024; 81:289-302. [PMID: 38583492 DOI: 10.1016/j.jhep.2024.03.043] [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: 05/05/2023] [Revised: 03/18/2024] [Accepted: 03/19/2024] [Indexed: 04/09/2024]
Abstract
BACKGROUND & AIMS Polyploidy in hepatocytes has been proposed as a genetic mechanism to buffer against transcriptional dysregulation. Here, we aim to demonstrate the role of polyploidy in modulating gene regulatory networks in hepatocytes during ageing. METHODS We performed single-nucleus RNA sequencing in hepatocyte nuclei of different ploidy levels isolated from young and old wild-type mice. Changes in the gene expression and regulatory network were compared to three independent strains that were haploinsufficient for HNF4A, CEBPA or CTCF, representing non-deleterious perturbations. Phenotypic characteristics of the liver section were additionally evaluated histologically, whereas the genomic allele composition of hepatocytes was analysed by BaseScope. RESULTS We observed that ageing in wild-type mice results in nuclei polyploidy and a marked increase in steatosis. Haploinsufficiency of liver-specific master regulators (HFN4A or CEBPA) results in the enrichment of hepatocytes with tetraploid nuclei at a young age, affecting the genomic regulatory network, and dramatically suppressing ageing-related steatosis tissue wide. Notably, these phenotypes are not the result of subtle disruption to liver-specific transcriptional networks, since haploinsufficiency in the CTCF insulator protein resulted in the same phenotype. Further quantification of genotypes of tetraploid hepatocytes in young and old HFN4A-haploinsufficient mice revealed that during ageing, tetraploid hepatocytes lead to the selection of wild-type alleles, restoring non-deleterious genetic perturbations. CONCLUSIONS Our results suggest a model whereby polyploidisation leads to fundamentally different cell states. Polyploid conversion enables pleiotropic buffering against age-related decline via non-random allelic segregation to restore a wild-type genome. IMPACT AND IMPLICATIONS The functional role of hepatocyte polyploidisation during ageing is poorly understood. Using single-nucleus RNA sequencing and BaseScope approaches, we have studied ploidy dynamics during ageing in murine livers with non-deleterious genetic perturbations. We have identified that hepatocytes present different cellular states and the ability to buffer ageing-associated dysfunctions. Tetraploid nuclei exhibit robust transcriptional networks and are better adapted to genomically overcome perturbations. Novel therapeutic interventions aimed at attenuating age-related changes in tissue function could be exploited by manipulation of ploidy dynamics during chronic liver conditions.
Collapse
Affiliation(s)
- Kelvin Yin
- Helmholtz Pioneer Campus (HPC), Helmholtz Munich, Neuherberg, Germany
| | - Maren Büttner
- Institute of Computational Biology, Computational Health Department, Helmholtz Munich, Neuherberg, Germany
| | | | | | - Liwei Zhang
- Helmholtz Pioneer Campus (HPC), Helmholtz Munich, Neuherberg, Germany
| | - Carlos Talavera-López
- Division of Infectious Diseases and Tropical Medicine, Ludwig-Maximilian-Universität Klinikum, Germany
| | - Fabian Theis
- Institute of Computational Biology, Computational Health Department, Helmholtz Munich, Neuherberg, Germany; Technical University of Munich, Department of Mathematics, 85748 Garching. Munich, Germany; German Cancer Research Centre, Heidelberg, Germany.
| | - Duncan T Odom
- German Cancer Research Center, Division of Regulatory Genomics and Cancer Evolution (B270), Heidelberg, Germany; Cancer Research UK Cambridge Institute, University of Cambridge, CB20RE, United Kingdom.
| | - Celia P Martinez-Jimenez
- Helmholtz Pioneer Campus (HPC), Helmholtz Munich, Neuherberg, Germany; TUM School of Medicine, Technical University of Munich, Munich, Germany; Institute of Biotechnology and Biomedicine (BIOTECMED), Department of Biochemistry and Molecular Biology, University of Valencia, Burjassot, Spain.
| |
Collapse
|
7
|
Zhang Q, Wu S, Chen Q, Zhang Y, Zhang C, Yin R, Ouyang Z, Wei Y. Reducing Oxidative Stress-Mediated Alcoholic Liver Injury by Multiplexed RNAi of Cyp2e1, Cyp4a10, and Cyp4a14. Biomedicines 2024; 12:1505. [PMID: 39062078 PMCID: PMC11274525 DOI: 10.3390/biomedicines12071505] [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: 04/29/2024] [Revised: 06/24/2024] [Accepted: 07/02/2024] [Indexed: 07/28/2024] Open
Abstract
The prevalence of excessive drinking-related alcoholic liver disease (ALD) is rising, yet therapeutic options remain limited. High alcohol consumption and consequent oxidative metabolism by cytochrome P450 (CYP) can lead to extremely high levels of reactive oxygen species, which overwhelm cellular defenses and harm hepatocytes. Our previous investigations showed that inhibiting Cyp2e1 using RNA interference reduced the incidence of ALD. However, compensatory mechanisms other than CYP2E1 contribute to oxidative stress in the liver. Therefore, we coupled triple siRNA lipid nanoparticles (LNPs) targeting Cyp2e1 with two isoenzymes Cyp4a10 and Cyp4a14 to treat ALD mouse models fed with Lieber-Decarli ethanol liquid diet for 12 weeks at the early (1st week), middle (5th week), and late (9th week) stages. The administration of triple siRNA LNPs significantly ameliorated chronic alcoholic liver injury in mice, and early treatment achieved the most profound effects. These effects can be attributed to a reduction in oxidative stress and increased expression of antioxidant genes, including Gsh-Px, Gsh-Rd, and Sod1. Moreover, we observed the alleviation of inflammation, evidenced by the downregulation of Il-1β, Il-6, Tnf-α, and Tgf-β, and the prevention of excessive lipid synthesis, evidenced by the restoration of the expression of Srebp1c, Acc, and Fas. Finally, triple siRNA treatment maintained normal metabolism in lipid oxidation. In brief, our research examined the possible targets for clinical intervention in ALD by examining the therapeutic effects of triple siRNA LNPs targeting Cyp2e1, Cyp4a10, and Cyp4a14. The in vivo knockdown of the three genes in this study is suggested as a promising siRNA therapeutic approach for ALD.
Collapse
Affiliation(s)
- Qi Zhang
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, China; (Q.Z.); (S.W.)
| | - Shuang Wu
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, China; (Q.Z.); (S.W.)
| | - Qiubing Chen
- Department of Urology, Frontier Science Centre for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Yahong Zhang
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, China; (Q.Z.); (S.W.)
| | - Cai Zhang
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, China; (Q.Z.); (S.W.)
| | - Runting Yin
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, China; (Q.Z.); (S.W.)
| | - Zhen Ouyang
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, China; (Q.Z.); (S.W.)
| | - Yuan Wei
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, China; (Q.Z.); (S.W.)
| |
Collapse
|
8
|
Majtan T, Olsen T, Sokolova J, Krijt J, Křížková M, Ida T, Ditrói T, Hansikova H, Vit O, Petrak J, Kuchař L, Kruger WD, Nagy P, Akaike T, Kožich V. Deciphering pathophysiological mechanisms underlying cystathionine beta-synthase-deficient homocystinuria using targeted metabolomics, liver proteomics, sphingolipidomics and analysis of mitochondrial function. Redox Biol 2024; 73:103222. [PMID: 38843767 PMCID: PMC11190558 DOI: 10.1016/j.redox.2024.103222] [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: 02/29/2024] [Revised: 05/17/2024] [Accepted: 06/02/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND Cystathionine β-synthase (CBS)-deficient homocystinuria (HCU) is an inherited disorder of sulfur amino acid metabolism with varying severity and organ complications, and a limited knowledge about underlying pathophysiological processes. Here we aimed at getting an in-depth insight into disease mechanisms using a transgenic mouse model of HCU (I278T). METHODS We assessed metabolic, proteomic and sphingolipidomic changes, and mitochondrial function in tissues and body fluids of I278T mice and WT controls. Furthermore, we evaluated the efficacy of methionine-restricted diet (MRD) in I278T mice. RESULTS In WT mice, we observed a distinct tissue/body fluid compartmentalization of metabolites with up to six-orders of magnitude differences in concentrations among various organs. The I278T mice exhibited the anticipated metabolic imbalance with signs of an increased production of hydrogen sulfide and disturbed persulfidation of free aminothiols. HCU resulted in a significant dysregulation of liver proteome affecting biological oxidations, conjugation of compounds, and metabolism of amino acids, vitamins, cofactors and lipids. Liver sphingolipidomics indicated upregulation of the pro-proliferative sphingosine-1-phosphate signaling pathway. Liver mitochondrial function of HCU mice did not seem to be impaired compared to controls. MRD in I278T mice improved metabolic balance in all tissues and substantially reduced dysregulation of liver proteome. CONCLUSION The study highlights distinct tissue compartmentalization of sulfur-related metabolites in normal mice, extensive metabolome, proteome and sphingolipidome disruptions in I278T mice, and the efficacy of MRD to alleviate some of the HCU-related biochemical abnormalities.
Collapse
Affiliation(s)
- Tomas Majtan
- Department of Pharmacology, University of Fribourg, Faculty of Science and Medicine, Fribourg, 1700, Switzerland.
| | - Thomas Olsen
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Jitka Sokolova
- Department of Pediatrics and Inherited Metabolic Disorders, Charles University-First Faculty of Medicine, Prague, 12808, Czech Republic; Department of Pediatrics and Inherited Metabolic Disorders, General University Hospital in Prague, Prague, 12808, Czech Republic
| | - Jakub Krijt
- Department of Pediatrics and Inherited Metabolic Disorders, Charles University-First Faculty of Medicine, Prague, 12808, Czech Republic; Department of Pediatrics and Inherited Metabolic Disorders, General University Hospital in Prague, Prague, 12808, Czech Republic
| | - Michaela Křížková
- Department of Pediatrics and Inherited Metabolic Disorders, Charles University-First Faculty of Medicine, Prague, 12808, Czech Republic; Department of Pediatrics and Inherited Metabolic Disorders, General University Hospital in Prague, Prague, 12808, Czech Republic
| | - Tomoaki Ida
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan
| | - Tamás Ditrói
- Department of Molecular Immunology and Toxicology and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, 1122, Hungary
| | - Hana Hansikova
- Department of Pediatrics and Inherited Metabolic Disorders, Charles University-First Faculty of Medicine, Prague, 12808, Czech Republic; Department of Pediatrics and Inherited Metabolic Disorders, General University Hospital in Prague, Prague, 12808, Czech Republic
| | - Ondrej Vit
- BIOCEV, First Faculty of Medicine, Charles University, 252 50, Vestec, Czech Republic
| | - Jiri Petrak
- BIOCEV, First Faculty of Medicine, Charles University, 252 50, Vestec, Czech Republic
| | - Ladislav Kuchař
- Department of Pediatrics and Inherited Metabolic Disorders, Charles University-First Faculty of Medicine, Prague, 12808, Czech Republic; Department of Pediatrics and Inherited Metabolic Disorders, General University Hospital in Prague, Prague, 12808, Czech Republic
| | - Warren D Kruger
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Péter Nagy
- Department of Molecular Immunology and Toxicology and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, 1122, Hungary; Department of Anatomy and Histology, HUN-REN-UVMB Laboratory of Redox Biology Research Group, University of Veterinary Medicine, 1078, Budapest, Hungary; Chemistry Institute, University of Debrecen, 4012, Debrecen, Hungary
| | - Takaaki Akaike
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan
| | - Viktor Kožich
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway; Department of Pediatrics and Inherited Metabolic Disorders, Charles University-First Faculty of Medicine, Prague, 12808, Czech Republic.
| |
Collapse
|
9
|
Hu Y, Li W, Cheng X, Yang H, She ZG, Cai J, Li H, Zhang XJ. Emerging Roles and Therapeutic Applications of Arachidonic Acid Pathways in Cardiometabolic Diseases. Circ Res 2024; 135:222-260. [PMID: 38900855 DOI: 10.1161/circresaha.124.324383] [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] [Indexed: 06/22/2024]
Abstract
Cardiometabolic disease has become a major health burden worldwide, with sharply increasing prevalence but highly limited therapeutic interventions. Emerging evidence has revealed that arachidonic acid derivatives and pathway factors link metabolic disorders to cardiovascular risks and intimately participate in the progression and severity of cardiometabolic diseases. In this review, we systemically summarized and updated the biological functions of arachidonic acid pathways in cardiometabolic diseases, mainly focusing on heart failure, hypertension, atherosclerosis, nonalcoholic fatty liver disease, obesity, and diabetes. We further discussed the cellular and molecular mechanisms of arachidonic acid pathway-mediated regulation of cardiometabolic diseases and highlighted the emerging clinical advances to improve these pathological conditions by targeting arachidonic acid metabolites and pathway factors.
Collapse
Affiliation(s)
- Yufeng Hu
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China (Y.H., X.C., H.Y., Z.-G.S., J.C., H.L., X.-J.Z.)
- Key Laboratory of Cardiovascular Disease Prevention and Control, Ministry of Education, First Affiliated Hospital of Gannan Medical University, Ganzhou, China (Y.H., X.C., H.Y.)
| | - Wei Li
- Department of Cardiology, Renmin Hospital of Wuhan University, China (W.L., Z.-G.S., H.L.)
| | - Xu Cheng
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China (Y.H., X.C., H.Y., Z.-G.S., J.C., H.L., X.-J.Z.)
- Key Laboratory of Cardiovascular Disease Prevention and Control, Ministry of Education, First Affiliated Hospital of Gannan Medical University, Ganzhou, China (Y.H., X.C., H.Y.)
| | - Hailong Yang
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China (Y.H., X.C., H.Y., Z.-G.S., J.C., H.L., X.-J.Z.)
- Key Laboratory of Cardiovascular Disease Prevention and Control, Ministry of Education, First Affiliated Hospital of Gannan Medical University, Ganzhou, China (Y.H., X.C., H.Y.)
| | - Zhi-Gang She
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China (Y.H., X.C., H.Y., Z.-G.S., J.C., H.L., X.-J.Z.)
- Department of Cardiology, Renmin Hospital of Wuhan University, China (W.L., Z.-G.S., H.L.)
| | - Jingjing Cai
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China (Y.H., X.C., H.Y., Z.-G.S., J.C., H.L., X.-J.Z.)
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, China (J.C.)
| | - Hongliang Li
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China (Y.H., X.C., H.Y., Z.-G.S., J.C., H.L., X.-J.Z.)
- Department of Cardiology, Renmin Hospital of Wuhan University, China (W.L., Z.-G.S., H.L.)
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan 430071, China (H.L.)
| | - Xiao-Jing Zhang
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China (Y.H., X.C., H.Y., Z.-G.S., J.C., H.L., X.-J.Z.)
- School of Basic Medical Sciences, Wuhan University, China (X.-J.Z.)
| |
Collapse
|
10
|
Bajaj G, Singh V, Sagar P, Gupta R, Singhal NK. Phosphoenolpyruvate carboxykinase-1 targeted siRNA promotes wound healing in type 2 diabetic mice by restoring glucose homeostasis. Int J Biol Macromol 2024; 270:132504. [PMID: 38772464 DOI: 10.1016/j.ijbiomac.2024.132504] [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: 02/24/2024] [Revised: 05/02/2024] [Accepted: 05/17/2024] [Indexed: 05/23/2024]
Abstract
It is well-accepted that the liver plays a vital role in the metabolism of glucose and its homeostasis. Dysregulated hepatic glucose production and utilization, leads to type 2 diabetes (T2DM). In the current study, RNA sequencing and qRT-PCR analysis of nanoformulation-treated T2DM mice (TGthr group) revealed beneficial crosstalk of PCK-1 silencing with other pathways involved in T2DM. The comparison of precise genetic expression profiles of the different experimental groups showed significantly improved hepatic glucose, fatty acid metabolism and several other T2DM-associated crucial markers after the nanoformulation treatment. As a result of these improvements, we observed a significant acceleration in wound healing and improved insulin signaling in vascular endothelial cells in the TGthr group as compared to the T2DM group. Enhanced phosphorylation of PI3K/Akt pathway proteins in the TGthr group resulted in increased angiogenesis as observed by the increased expression of endothelial cell markers (CD31, CD34) thereby improving endothelial dysfunctions in the TGthr group. Additionally, therapeutic nanoformulation has been observed to improve the inflammatory cytokine profile in the TGthr group. Overall, our results demonstrated that the synthesized therapeutic nanoformulation referred to as GPR8:PCK-1siRNA holds the potential in ameliorating hyperglycemia-associated complications such as delayed wound healing in diabetes.
Collapse
Affiliation(s)
- Geetika Bajaj
- National Agri-Food Biotechnology Institute (NABI), Sector-81, S.A.S Nagar, Mohali 140306, Punjab, India; Department of Biotechnology, Panjab University, Sector 25, Chandigarh 160014, India
| | - Vishal Singh
- National Institute for Implementation Research on Non-Communicable Diseases, Jodhpur 342005, India
| | - Poonam Sagar
- National Agri-Food Biotechnology Institute (NABI), Sector-81, S.A.S Nagar, Mohali 140306, Punjab, India
| | - Ritika Gupta
- National Agri-Food Biotechnology Institute (NABI), Sector-81, S.A.S Nagar, Mohali 140306, Punjab, India
| | - Nitin Kumar Singhal
- National Agri-Food Biotechnology Institute (NABI), Sector-81, S.A.S Nagar, Mohali 140306, Punjab, India.
| |
Collapse
|
11
|
Hu J, Li S, Zhong X, Wei Y, Sun Q, Zhong L. Human umbilical cord mesenchymal stem cells attenuate diet-induced obesity and NASH-related fibrosis in mice. Heliyon 2024; 10:e25460. [PMID: 38356602 PMCID: PMC10864966 DOI: 10.1016/j.heliyon.2024.e25460] [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: 09/12/2023] [Revised: 01/26/2024] [Accepted: 01/26/2024] [Indexed: 02/16/2024] Open
Abstract
Non-alcoholic steatohepatitis (NASH) is a progressive form of non-alcoholic fatty liver disease (NAFLD) that may progress to cirrhosis and hepatocellular carcinoma but has no available treatment. Mesenchymal stem cells (MSCs) have become increasingly prominent in cell therapy. Human umbilical cord MSCs (hUC-MSCs) are considered superior to other MSCs due to their strong immunomodulatory ability, ease of collection, low immune rejection, and no tumorigenicity. Though hUC-MSCs have received increasing attention in research, they have been rarely applied in any investigations or treatments of NASH and associated fibrosis. Therefore, this study evaluated the therapeutic efficacy of hUC-MSCs in C57BL/6 mice with diet-induced NASH. At week 32, mice were randomized into two groups: phosphate-buffered saline and MSCs, which were injected into the tail vein. At week 40, glucose metabolism was evaluated using glucose and insulin tolerance tests. NASH-related indicators were examined using various biological methods. hUC-MSC administration alleviated obesity, glucose metabolism, hepatic steatosis, inflammation, and fibrosis. Liver RNA-seq showed that the expression of the acyl-CoA thioesterase (ACOT) family members Acot1, Acot2, and Acot3 involved in fatty acid metabolism were altered. The cytochrome P450 (CYP) members Cyp4a10 and Cyp4a14, which are involved in the peroxisome proliferator-activator receptor (PPAR) signaling pathway, were significantly downregulated after hUC-MSC treatment. In conclusion, hUC-MSCs effectively reduced Western diet-induced obesity, NASH, and fibrosis in mice, partly by regulating lipid metabolism and the PPAR signaling pathway.
Collapse
Affiliation(s)
- Jiali Hu
- Department of Gastroenterology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shan Li
- Department of Gastroenterology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xuan Zhong
- Department of Gastroenterology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yushuang Wei
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200120, China
| | - Qinjuan Sun
- Department of Gastroenterology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lan Zhong
- Department of Gastroenterology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200120, China
| |
Collapse
|
12
|
Zhou X, Liu Z, Yang X, Feng J, Gins MS, Yan T, Han L, Zhang H. The Mechanism Underlying the Hypoglycemic Effect of Epimedin C on Mice with Type 2 Diabetes Mellitus Based on Proteomic Analysis. Nutrients 2023; 16:25. [PMID: 38201855 PMCID: PMC10780735 DOI: 10.3390/nu16010025] [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: 11/19/2023] [Revised: 12/12/2023] [Accepted: 12/13/2023] [Indexed: 01/12/2024] Open
Abstract
Type 2 diabetes mellitus (T2DM) has become a worldwide public health problem. Epimedin C is considered one of the most important flavonoids in Epimedium, a famous edible herb in China and Southeast Asia that is traditionally used in herbal medicine to treat diabetes. In the present study, the therapeutic potential of epimedin C against T2DM was ascertained using a mouse model, and the mechanism underlying the hypoglycemic activity of epimedin C was explored using a label-free proteomic technique for the first time. Levels of fasting blood glucose (FBG), homeostasis model assessment of insulin resistance (HOMA-IR), and oral glucose tolerance, as well as contents of malondialdehyde (MDA) and low-density lipoprotein cholesterol (LDL-C) in the 30 mg·kg-1 epimedin C group (EC30 group), were significantly lower than those in the model control group (MC group) (p < 0.05), while the contents of hepatic glycogen, insulin, and high-density lipoprotein cholesterol (HDL-C), as well as activities of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) in the EC30 group were notably higher than those in the MC group (p < 0.05). The structures of liver cells and tissues were greatly destroyed in the MC group, whereas the structures of cells and tissues were basically complete in the EC30 group, which were similar to those in the normal control group (NC group). A total of 92 differentially expressed proteins (DEPs) were enriched in the gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. In the EC30 vs. MC groups, the expression level of cytosolic phosphoenolpyruvate carboxykinase (Pck1) was down-regulated, while the expression levels of group XIIB secretory phospholipase A2-like protein (Pla2g12b), apolipoprotein B-100 (Apob), and cytochrome P450 4A14 (Cyp4a14) were up-regulated. According to the KEGG pathway assay, Pck1 participated in the gluconeogenesis and insulin signaling pathways, and Pla2g12b, Apob, and Cyp4a14 were the key proteins in the fat digestion and fatty acid degradation pathways. Pck1, Pla2g12b, Apob, and Cyp4a14 seemed to play important roles in the prevention and treatment of T2DM. In summary, epimedin C inhibited Pck1 expression to maintain FBG at a relatively stable level, promoted Pla2g12b, Apob, and Cyp4a14 expressions to alleviate liver lipotoxicity, and protected liver tissues and cells from oxidant stress possibly by its phenolic hydroxyl groups.
Collapse
Affiliation(s)
- Xuexue Zhou
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, Academician and Expert Workstations in Puer City of Yunnan Province, College of Food Engineering and Nutritional Science, Provincial Research Station of Se-Enriched Foods in Hanyin County of Shaanxi Province, International Joint Research Center of Shaanxi Province for Food and Health Sciences, Shaanxi Normal University, Xi’an 710119, China (Z.L.); (T.Y.); (L.H.)
| | - Ziqi Liu
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, Academician and Expert Workstations in Puer City of Yunnan Province, College of Food Engineering and Nutritional Science, Provincial Research Station of Se-Enriched Foods in Hanyin County of Shaanxi Province, International Joint Research Center of Shaanxi Province for Food and Health Sciences, Shaanxi Normal University, Xi’an 710119, China (Z.L.); (T.Y.); (L.H.)
| | - Xiaohua Yang
- Research Station of Selenium-Enriched Tea of Shaanxi Province, Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China
| | - Jing Feng
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, Academician and Expert Workstations in Puer City of Yunnan Province, College of Food Engineering and Nutritional Science, Provincial Research Station of Se-Enriched Foods in Hanyin County of Shaanxi Province, International Joint Research Center of Shaanxi Province for Food and Health Sciences, Shaanxi Normal University, Xi’an 710119, China (Z.L.); (T.Y.); (L.H.)
- Agrarian and Technological Institute, Peoples’ Friendship University of Russia, Moscow 119991, Russia;
| | - Murat Sabirovich Gins
- Agrarian and Technological Institute, Peoples’ Friendship University of Russia, Moscow 119991, Russia;
| | - Tingyu Yan
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, Academician and Expert Workstations in Puer City of Yunnan Province, College of Food Engineering and Nutritional Science, Provincial Research Station of Se-Enriched Foods in Hanyin County of Shaanxi Province, International Joint Research Center of Shaanxi Province for Food and Health Sciences, Shaanxi Normal University, Xi’an 710119, China (Z.L.); (T.Y.); (L.H.)
| | - Lei Han
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, Academician and Expert Workstations in Puer City of Yunnan Province, College of Food Engineering and Nutritional Science, Provincial Research Station of Se-Enriched Foods in Hanyin County of Shaanxi Province, International Joint Research Center of Shaanxi Province for Food and Health Sciences, Shaanxi Normal University, Xi’an 710119, China (Z.L.); (T.Y.); (L.H.)
| | - Huafeng Zhang
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, Academician and Expert Workstations in Puer City of Yunnan Province, College of Food Engineering and Nutritional Science, Provincial Research Station of Se-Enriched Foods in Hanyin County of Shaanxi Province, International Joint Research Center of Shaanxi Province for Food and Health Sciences, Shaanxi Normal University, Xi’an 710119, China (Z.L.); (T.Y.); (L.H.)
| |
Collapse
|
13
|
Duan J, Dong W, Wang G, Xiu W, Pu G, Xu J, Ye C, Zhang X, Zhu Y, Wang C. Senescence-associated 13-HODE production promotes age-related liver steatosis by directly inhibiting catalase activity. Nat Commun 2023; 14:8151. [PMID: 38071367 PMCID: PMC10710422 DOI: 10.1038/s41467-023-44026-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
Aging is a major risk factor for metabolic disorders. Polyunsaturated fatty acid-derived bioactive lipids play critical roles as signaling molecules in metabolic processes. Nonetheless, their effects on age-related liver steatosis remain unknown. Here we show that senescent liver cells induce liver steatosis in a paracrine manner. Linoleic acid-derived 9-hydroxy-octadecadienoic acid (9-HODE) and 13-HODE increase in middle-aged (12-month-old) and aged (20-month-old) male mouse livers and conditioned medium from senescent hepatocytes and macrophages. Arachidonate 15-lipoxygenase, an enzyme for 13-HODE and 9-HODE production, is upregulated in senescent cells. A 9-HODE and 13-HODE mixture induces liver steatosis and activates SREBP1. Furthermore, catalase (CAT) is a direct target of 13-HODE, and its activity is decreased by 13-HODE. CAT overexpression reduces 13-HODE-induced liver steatosis and protects male mice against age-related liver steatosis. Therefore, 13-HODE produced by senescent hepatocytes and macrophages activates SREBP1 by directly inhibiting CAT activity and promotes liver steatosis.
Collapse
Affiliation(s)
- Jinjie Duan
- NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
- School of Public Health, Tianjin Medical University, Tianjin, China
| | - Wenhui Dong
- NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Guangyan Wang
- NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, Tianjin, China
| | - Wenjing Xiu
- NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Guangyin Pu
- NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Jingwen Xu
- NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Chenji Ye
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Xu Zhang
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, Tianjin, China.
| | - Yi Zhu
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China.
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, Tianjin, China.
| | - Chunjiong Wang
- NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China.
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China.
- Tianjin Key Laboratory of Medical Epigenetics, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
14
|
Jiang C, Saiki Y, Hirota S, Iwata K, Wang X, Ito Y, Murakami K, Imura T, Inoue J, Masamune A, Hirayama A, Goto M, Furukawa T. Ablation of Dual-Specificity Phosphatase 6 Protects against Nonalcoholic Fatty Liver Disease via Cytochrome P450 4A and Mitogen-Activated Protein Kinase. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:1988-2000. [PMID: 37741451 DOI: 10.1016/j.ajpath.2023.09.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 07/31/2023] [Accepted: 09/01/2023] [Indexed: 09/25/2023]
Abstract
Dual-specificity phosphatase 6 (DUSP6) is a specific phosphatase for mitogen-activated protein kinase (MAPK). This study used a high-fat diet (HFD)-induced murine nonalcoholic fatty liver disease model to investigate the role of DUSP6 in this disease. Wild-type (WT) and Dusp6-haploinsufficiency mice developed severe obesity and liver pathology consistent with nonalcoholic fatty liver disease when exposed to HFD. In contrast, Dusp6-knockout (KO) mice completely eliminated these phenotypes. Furthermore, primary hepatocytes isolated from WT mice exposed to palmitic and oleic acids exhibited abundant intracellular lipid accumulation, whereas hepatocytes from Dusp6-KO mice showed minimal lipid accumulation. Transcriptome analysis revealed significant down-regulation of genes encoding cytochrome P450 4A (CYP4A), known to promote ω-hydroxylation of fatty acids and hepatic steatosis, in Dusp6-KO hepatocytes compared with that in WT hepatocytes. Diminished CYP4A expression was observed in the liver of Dusp6-KO mice compared with WT and Dusp6-haploinsufficiency mice. Knockdown of DUSP6 in HepG2, a human liver-lineage cell line, also promoted a reduction of lipid accumulation, down-regulation of CYP4A, and up-regulation of phosphorylated/activated MAPK. Furthermore, inhibition of MAPK activity promoted lipid accumulation in DUSP6-knockdown HepG2 cells without affecting CYP4A expression, indicating that CYP4A expression is independent of MAPK activation. These findings highlight the significant role of DUSP6 in HFD-induced steatohepatitis through two distinct pathways involving CYP4A and MAPK.
Collapse
Affiliation(s)
- Can Jiang
- Department of Investigative Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yuriko Saiki
- Department of Investigative Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan.
| | - Shuto Hirota
- Department of Investigative Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kosei Iwata
- Department of Investigative Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Xinyue Wang
- Department of Investigative Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yutaka Ito
- Department of Investigative Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Keigo Murakami
- Department of Investigative Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takehiro Imura
- Division of Transplantation and Regenerative Medicine, Tohoku University, Sendai, Japan
| | - Jun Inoue
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Atsushi Masamune
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Akiyoshi Hirayama
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Japan
| | - Masafumi Goto
- Division of Transplantation and Regenerative Medicine, Tohoku University, Sendai, Japan
| | - Toru Furukawa
- Department of Investigative Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan.
| |
Collapse
|
15
|
Ning L, Zou Y, Li S, Cao Y, Xu B, Zhang S, Cai Y. Anti-PCSK9 Treatment Attenuates Liver Fibrosis via Inhibiting Hypoxia-Induced Autophagy in Hepatocytes. Inflammation 2023; 46:2102-2119. [PMID: 37466835 PMCID: PMC10673768 DOI: 10.1007/s10753-023-01865-8] [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: 04/05/2023] [Revised: 06/25/2023] [Accepted: 06/26/2023] [Indexed: 07/20/2023]
Abstract
Hypoxia and its induced autophagy are involved in the initiation and progression of liver fibrosis. Proprotein convertase subtilisin/kexin type 9 (PCSK9) has been recognized as a potential regulator of autophagy. Our previously reported study found that PCSK9 expression increased in liver fibrosis and that anti-PCSK9 treatment alleviated liver injury. This study aimed to investigate the mechanism of anti-PCSK9 treatment on liver fibrosis by inhibiting hypoxia-induced autophagy. Carbon tetrachloride-induced mouse liver fibrosis and mouse hepatocyte line AML12, cultured under the hypoxic condition, were established to undergo PCSK9 inhibition. The degree of liver fibrosis was shown with histological staining. The reactive oxygen species (ROS) generation was detected by flow cytometry. The expression of PCSK9, hypoxia-inducible factor-1α (HIF-1α), and autophagy-related proteins was examined using Western blot. The autophagic flux was assessed under immunofluorescence and transmission electron microscope. The mouse liver samples were investigated via RNA-sequencing to explore the underlying signaling pathway. The results showed that PCSK9 expression was upregulated with the development of liver fibrosis, which was accompanied by enhanced autophagy. In vitro data verified that PCSK9 increased via hypoxia and inflammation, accompanied by the hypoxia-induced autophagy increased. Then, the validation was acquired of the bidirectional interaction of hypoxia-ROS and PCSK9. The hypoxia reversal attenuated PCSK9 expression and autophagy. Additionally, anti-PCSK9 treatment alleviated liver inflammation and fibrosis, reducing hypoxia and autophagy in vivo. In mechanism, the AMPK/mTOR/ULK1 signaling pathway was identified as a target for anti-PCSK9 therapy. In conclusion, anti-PCSK9 treatment could alleviate liver inflammation and fibrosis by regulating AMPK/mTOR/ULK1 signaling pathway to reduce hypoxia-induced autophagy in hepatocytes.
Collapse
Affiliation(s)
- Liuxin Ning
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Liver Diseases, Shanghai, 200032, China
| | - Yanting Zou
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Liver Diseases, Shanghai, 200032, China
| | - Shuyu Li
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Liver Diseases, Shanghai, 200032, China
| | - Yue Cao
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Liver Diseases, Shanghai, 200032, China
| | - Beili Xu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Liver Diseases, Shanghai, 200032, China
| | - Shuncai Zhang
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Liver Diseases, Shanghai, 200032, China
| | - Yu Cai
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Shanghai Institute of Liver Diseases, Shanghai, 200032, China.
| |
Collapse
|
16
|
Coyne ES, Nie Y, Abdurrachim D, Ong CZL, Zhou Y, Ali AAB, Meyers S, Grein J, Blumenschein W, Gongol B, Liu Y, Hugelshofer C, Carballo-Jane E, Talukdar S. Leukotriene B4 receptor 1 (BLT1) does not mediate disease progression in a mouse model of liver fibrosis. Biochem J 2023; 481:BCJ20230422. [PMID: 38014500 PMCID: PMC10903445 DOI: 10.1042/bcj20230422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/21/2023] [Accepted: 11/27/2023] [Indexed: 11/29/2023]
Abstract
MASH is a prevalent liver disease that can progress to fibrosis, cirrhosis, hepatocellular carcinoma (HCC), and ultimately death, but there are no approved therapies. Leukotriene B4 (LTB4) is a potent pro-inflammatory chemoattractant that drives macrophage and neutrophil chemotaxis, and genetic loss or inhibition of its high affinity receptor, leukotriene B4 receptor 1 (BLT1), results in improved insulin sensitivity and decreased hepatic steatosis. To validate the therapeutic efficacy of BLT1 inhibition in an inflammatory and pro-fibrotic mouse model of MASH and fibrosis, mice were challenged with a choline-deficient, L-amino acid defined high fat diet and treated with a BLT1 antagonist at 30 or 90 mg/kg for 8 weeks. Liver function, histology, and gene expression were evaluated at the end of the study. Treatment with the BLT1 antagonist significantly reduced plasma lipids and liver steatosis but had no impact on liver injury biomarkers or histological endpoints such as inflammation, ballooning, or fibrosis compared to control. Artificial intelligence-powered digital pathology analysis revealed a significant reduction in steatosis co-localized fibrosis in livers treated with the BLT1 antagonist. Liver RNA-seq and pathway analyses revealed significant changes in fatty acid, arachidonic acid, and eicosanoid metabolic pathways with BLT1 antagonist treatment, however, these changes were not sufficient to impact inflammation and fibrosis endpoints. Targeting this LTB4-BLT1 axis with a small molecule inhibitor in animal models of chronic liver disease should be considered with caution, and additional studies are warranted to understand the mechanistic nuances of BLT1 inhibition in the context of MASH and liver fibrosis.
Collapse
Affiliation(s)
| | - Yilin Nie
- Merck & Co., Inc., South San Francisco, CA, U.S.A
| | | | | | | | | | | | - Jeff Grein
- Merck & Co., Inc., South San Francisco, CA, U.S.A
| | | | | | - Yang Liu
- Merck & Co., Inc., South San Francisco, CA, U.S.A
| | | | | | | |
Collapse
|
17
|
Ma R, Liu Q, Liu Z, Sun X, Jiang X, Hou J, Zhang Y, Wu Y, Cheng M, Dong Z. H19/Mir-130b-3p/Cyp4a14 potentiate the effect of praziquantel on liver in the treatment of Schistosoma japonicum infection. Acta Trop 2023; 247:107012. [PMID: 37659685 DOI: 10.1016/j.actatropica.2023.107012] [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: 04/29/2023] [Revised: 08/09/2023] [Accepted: 08/30/2023] [Indexed: 09/04/2023]
Abstract
BACKGROUND Schistosomiasis is a prevalent infectious disease caused by the parasitic trematodes of the genus Schistosoma. Praziquantel (PZQ), a safe and affordable drug, is the recommended oral treatment for schistosomiasis. The main pathologic manifestation of schistosomiasis is liver injury. However, the role and interactions of various RNA molecules in the effect of PZQ on the liver after S. japonicum infection have not been elucidated. RESULTS In this study, C57BL/6 mice were randomly divided into the control group, infection group, and PZQ treatment group. Total RNA was extracted from the livers of the mice. High-throughput whole transcriptome sequencing was performed to detect the RNA expression profiles in the three groups. A co-expression gene-interaction network was established based on the significant differentially expressed genes in the PZQ treatment group; messenger RNA (mRNA) Cyp4a14 was identified as a critical hub gene. Furthermore, competitive endogenous RNA networks were constructed by predicting the specific binding relations between mRNA and long noncoding (lnc) RNA and between lncRNA and microRNA (miRNA) of Cyp4a14, suggesting the involvement of the H19/miR-130b-3p/Cyp4a14 regulatory axis. Dual luciferase reporter assay result proved the specific binding of miR-130b-3p with Cyp4a14 3'UTR. CONCLUSIONS Our findings indicate the involvement of the H19/miR-130b-3p/Cyp4a14 axis in the effect of PZQ on the liver after S. japonicum infection. Moreover, the expression of mRNA Cyp4a14 could be regulated by the bonding of miR-130b-3p with 3'UTR of Cyp4a14. The findings of this study could provide a novel perspective to understand the host response to PZQ against S. japonicum in the future.
Collapse
Affiliation(s)
- Rui Ma
- Department of Health and Disease Management, School of Nursing, Binzhou Medical University, Guanhai Road 346, Yantai, Shandong, 264000, China
| | - Qiang Liu
- Department of Anesthesia, Binzhou Medical University Hospital, Binzhou, Shandong, 256600, China
| | - Zimo Liu
- Electrocardiogram Room, Yantai Yuhuangding Hospital, Yantai, Shandong, 264000, China
| | - Xu Sun
- Department of Health and Disease Management, School of Nursing, Binzhou Medical University, Guanhai Road 346, Yantai, Shandong, 264000, China
| | - Xinze Jiang
- Department of Pathogenic Biology, School of Basic Medical Sciences, Binzhou Medical University, Guanhai Road 346, Yantai, Shandong, 264000, China
| | - Jiangshan Hou
- Department of Pathogenic Biology, School of Basic Medical Sciences, Binzhou Medical University, Guanhai Road 346, Yantai, Shandong, 264000, China
| | - Yumei Zhang
- Department of Pathogenic Biology, School of Basic Medical Sciences, Binzhou Medical University, Guanhai Road 346, Yantai, Shandong, 264000, China
| | - Yulong Wu
- Department of Pathogenic Biology, School of Basic Medical Sciences, Binzhou Medical University, Guanhai Road 346, Yantai, Shandong, 264000, China.
| | - Mei Cheng
- Department of Health and Disease Management, School of Nursing, Binzhou Medical University, Guanhai Road 346, Yantai, Shandong, 264000, China.
| | - Zhouyan Dong
- Department of Pathogenic Biology, School of Basic Medical Sciences, Binzhou Medical University, Guanhai Road 346, Yantai, Shandong, 264000, China.
| |
Collapse
|
18
|
Zhong D, Cai J, Hu C, Chen J, Zhang R, Fan C, Li S, Zhang H, Xu Z, Jia Z, Guo D, Sun Y. Inhibition of mPGES-2 ameliorates NASH by activating NR1D1 via heme. Hepatology 2023; 78:547-561. [PMID: 35839302 DOI: 10.1002/hep.32671] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 06/27/2022] [Accepted: 07/12/2022] [Indexed: 12/08/2022]
Abstract
BACKGROUND AND AIMS Nonalcoholic fatty liver disease (NAFLD), a complex metabolic syndrome, has limited therapeutic options. Microsomal prostaglandin E synthase-2 (mPGES-2) was originally discovered as a prostaglandin E 2 (PGE 2 ) synthase; however, it does not produce PGE 2 in the liver. Moreover, the role of mPGES-2 in NAFLD remains undefined. Herein, we aimed to determine the function and mechanism of mPGES-2 in liver steatosis and steatohepatitis. APPROACH AND RESULTS To evaluate the role of mPGES-2 in NAFLD, whole-body or hepatocyte-specific mPGES-2-deficient mice fed a high-fat or methionine-choline-deficient diet were used. Compared with control mice, mPGES-2-deficient mice showed reduced hepatic lipid accumulation, along with ameliorated liver injury, inflammation, and fibrosis. Furthermore, the protective effect of mPGES-2 deficiency against NAFLD was dependent on decreased cytochrome P450 4A14 and increased acyl-CoA thioesterase 4 levels regulated by the heme receptor nuclear receptor subfamily 1 group D member 1 (NR1D1), but not PGE 2 . Heme regulated the increased NR1D1 activity mediated by mPGES-2 deficiency. Further, we confirmed the protective role of the mPGES-2 inhibitor SZ0232 in NAFLD therapy. CONCLUSION Our study indicates the pathogenic role of mPGES-2 and outlines the mechanism in mediating NAFLD, thereby highlighting the therapeutic potential of mPGES-2 inhibition in liver steatosis and steatohepatitis.
Collapse
Affiliation(s)
- Dandan Zhong
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy , Xuzhou Medical University , Xuzhou , Jiangsu , P. R. China
| | - Jie Cai
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy , Xuzhou Medical University , Xuzhou , Jiangsu , P. R. China
- Nanjing Key Laboratory of Pediatrics , Nanjing Children's Hospital , Nanjing Medical University , Nanjing , Jiangsu , P. R. China
- Public Experimental Research Center of Xuzhou Medical University , Xuzhou Medical University , Xuzhou , Jiangsu , P. R. China
| | - Cheng Hu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy , Xuzhou Medical University , Xuzhou , Jiangsu , P. R. China
| | - Jingshuo Chen
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy , Xuzhou Medical University , Xuzhou , Jiangsu , P. R. China
- Public Experimental Research Center of Xuzhou Medical University , Xuzhou Medical University , Xuzhou , Jiangsu , P. R. China
| | - Rumeng Zhang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy , Xuzhou Medical University , Xuzhou , Jiangsu , P. R. China
- Department of Pharmacology , Xuzhou Central Hospital , Xuzhou , Jiangsu , China
| | - Chenyu Fan
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy , Xuzhou Medical University , Xuzhou , Jiangsu , P. R. China
| | - Shanshan Li
- Jiangsu Medical Engineering Research Center of Gene Detection , Xuzhou , Jiangsu , China
| | - Hongxing Zhang
- Jiangsu Province Key Laboratory of Anesthesiology , Xuzhou Medical University , Xuzhou , Jiangsu , P. R. China
| | - Zhou Xu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy , Xuzhou Medical University , Xuzhou , Jiangsu , P. R. China
- Public Experimental Research Center of Xuzhou Medical University , Xuzhou Medical University , Xuzhou , Jiangsu , P. R. China
| | - Zhanjun Jia
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy , Xuzhou Medical University , Xuzhou , Jiangsu , P. R. China
- Nanjing Key Laboratory of Pediatrics , Nanjing Children's Hospital , Nanjing Medical University , Nanjing , Jiangsu , P. R. China
| | - Dong Guo
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy , Xuzhou Medical University , Xuzhou , Jiangsu , P. R. China
| | - Ying Sun
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy , Xuzhou Medical University , Xuzhou , Jiangsu , P. R. China
| |
Collapse
|
19
|
Li X, Zheng T, Zhang Y, Zhao Y, Liu F, Dai S, Liu X, Zhang M. Dickkopf-1 promotes vascular smooth muscle cell foam cell formation and atherosclerosis development through CYP4A11/SREBP2/ABCA1. FASEB J 2023; 37:e23048. [PMID: 37389895 DOI: 10.1096/fj.202300295r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 05/20/2023] [Accepted: 06/08/2023] [Indexed: 07/01/2023]
Abstract
Vascular smooth muscle cells (VSMCs) are considered to be a crucial source of foam cells in atherosclerosis due to their low expression level of cholesterol exporter ATP-binding cassette transporter A1 (ABCA1) intrinsically. While the definite regulatory mechanisms are complicated and have not yet been fully elucidated, we previously reported that Dickkopf-1 (DKK1) mediates endothelial cell (EC) dysfunction, thereby aggravating atherosclerosis. However, the role of smooth muscle cell (SMC) DKK1 in atherosclerosis and foam cell formation remains unknown. In this study, we established SMC-specific DKK1-knockout (DKK1SMKO ) mice by crossbreeding DKK1flox/flox mice with TAGLN-Cre mice. Then, DKK1SMKO mice were crossed with APOE-/- mice to generate DKK1SMKO /APOE-/- mice, which exhibited milder atherosclerotic burden and fewer SMC foam cells. In vitro loss- and gain-of-function studies of DKK1 in primary human aortic smooth muscle cells (HASMCs) have proven that DKK1 prevented oxidized lipid-induced ABCA1 upregulation and cholesterol efflux and promoted SMC foam cell formation. Mechanistically, RNA-sequencing (RNA-seq) analysis of HASMCs as well as chromatin immunoprecipitation (ChIP) experiments showed that DKK1 mediates the binding of transcription factor CCAAT/enhancer-binding protein delta (C/EBPδ) to the promoter of cytochrome P450 epoxygenase 4A11 (CYP4A11) to regulate its expression. In addition, CYP4A11 as well as its metabolite 20-HETE-promoted activation of transcription factor sterol regulatory element-binding protein 2 (SREBP2) mediated the DKK1 regulation of ABCA1 in SMC. Furthermore, HET0016, the antagonist of CYP4A11, has also shown an alleviating effect on atherosclerosis. In conclusion, our results demonstrate that DKK1 promotes SMC foam cell formation during atherosclerosis via a reduction in CYP4A11-20-HETE/SREBP2-mediated ABCA1 expression.
Collapse
Affiliation(s)
- Xiao Li
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Tengfei Zheng
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Yu Zhang
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Yachao Zhao
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Fengming Liu
- Department of Immunology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Shen Dai
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Xiaolin Liu
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Mei Zhang
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
20
|
Xue L, Liu K, Yan C, Dun J, Xu Y, Wu L, Yang H, Liu H, Xie L, Wang G, Liang Y. Schisandra lignans ameliorate nonalcoholic steatohepatitis by regulating aberrant metabolism of phosphatidylethanolamines. Acta Pharm Sin B 2023; 13:3545-3560. [PMID: 37655337 PMCID: PMC10465965 DOI: 10.1016/j.apsb.2023.04.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 03/16/2023] [Accepted: 04/19/2023] [Indexed: 09/02/2023] Open
Abstract
Nonalcoholic steatohepatitis (NASH) is a spectrum of chronic liver disease characterized by hepatic lipid metabolism disorder. Recent reports emphasized the contribution of triglyceride and diglyceride accumulation to NASH, while the other lipids associated with the NASH pathogenesis remained unexplored. The specific purpose of our study was to explore a novel pathogenesis and treatment strategy of NASH via profiling the metabolic characteristics of lipids. Herein, multi-omics techniques based on LC-Q-TOF/MS, LC-MS/MS and MS imaging were developed and used to screen the action targets related to NASH progress and treatment. A methionine and choline deficient (MCD) diet-induced mouse model of NASH was then constructed, and Schisandra lignans extract (SLE) was applied to alleviate hepatic damage by regulating the lipid metabolism-related enzymes CES2A and CYP4A14. Hepatic lipidomics indicated that MCD-diet led to aberrant accumulation of phosphatidylethanolamines (PEs), and SLE could significantly reduce the accumulation of intrahepatic PEs. Notably, exogenous PE (18:0/18:1) was proved to significantly aggravate the mitochondrial damage and hepatocyte apoptosis. Supplementing PE (18:0/18:1) also deteriorated the NASH progress by up regulating intrahepatic proinflammatory and fibrotic factors, while PE synthase inhibitor exerted a prominent hepatoprotective role. The current work provides new insights into the relationship between PE metabolism and the pathogenesis of NASH.
Collapse
Affiliation(s)
- Lijuan Xue
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Keanqi Liu
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Caixia Yan
- Zhejiang Provincial Key Laboratory for Drug Evaluation and Clinical Research, Department of Clinical Pharmacy, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Junling Dun
- Analytical Applications Center, Shimadzu (China) Co., Ltd., Shanghai 200233, China
| | - Yexin Xu
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Linlin Wu
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Huizhu Yang
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Huafang Liu
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Lin Xie
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Guangji Wang
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Yan Liang
- Key Lab of Drug Metabolism & Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
21
|
Wang L, Derous D, Huang X, Mitchell S, Douglas A, Lusseau D, Wang Y, Speakman J. The Effects of Graded Levels of Calorie Restriction: XIX. Impact of Graded Calorie Restriction on Protein Expression in the Liver. J Gerontol A Biol Sci Med Sci 2023; 78:1125-1134. [PMID: 36757838 PMCID: PMC10329235 DOI: 10.1093/gerona/glad017] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Indexed: 02/10/2023] Open
Abstract
Calorie restriction (CR) extends life span by modulating the mechanisms involved in aging. We quantified the hepatic proteome of male C57BL/6 mice exposed to graded levels of CR (0%-40% CR) for 3 months, and evaluated which signaling pathways were most affected. The metabolic pathways most significantly stimulated by the increase in CR, included the glycolysis/gluconeogenesis pathway, the pentose phosphate pathway, the fatty acid degradation pathway, the valine, leucine, and isoleucine degradation pathway, and the lysine degradation pathway. The metabolism of xenobiotics by cytochrome P450 pathway was activated and feminized by increased CR, while production in major urinary proteins (Mups) was strongly reduced, consistent with a reduced investment in reproduction as predicted by the disposable soma hypothesis. However, we found no evidence of increased somatic protection, and none of the 4 main pathways implied to be linked to the impact of CR on life span (insulin/insulin-like growth factor [IGF-1], nuclear factor-κB [NF-κB], mammalian Target of Rapamycin [mTOR], and sirtuins) as well as pathways in cancer, were significantly changed at the protein level in relation to the increase in CR level. This was despite previous work at the transcriptome level in the same individuals indicating such changes. On the other hand, we found Aldh2, Aldh3a2, and Aldh9a1 in carnitine biosynthesis and Acsl5 in carnitine shuttle system were up-regulated by increased CR, which are consistent with our previous work on metabolome of the same individuals. Overall, the patterns of protein expression were more consistent with a "clean cupboards" than a "disposable soma" interpretation.
Collapse
Affiliation(s)
- Lu Wang
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Yantai University, Yantai, China
| | - Davina Derous
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, UK
| | - Xiahe Huang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Chaoyang, Beijing, China
| | - Sharon E Mitchell
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, UK
| | - Alex Douglas
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, UK
| | - David Lusseau
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, UK
| | - Yingchun Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Chaoyang, Beijing, China
| | - John R Speakman
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, UK
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Chaoyang, Beijing, China
- CAS Centre for Excellence in Animal Evolution and Genetics (CCEAEG), Kunming, China
- Shenzhen Key Laboratory of Metabolic Health, Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced technology, Chinese Academy of Sciences, Shenzhen, China
| |
Collapse
|
22
|
Watanabe K, Kondo M, Ikenaka Y, Nakayama SMM, Ishizuka M. A Comparative Genomic and Phylogenetic Investigation of the Xenobiotic Metabolism Enzymes of Cytochrome P450 in Elephants Shows Loss in CYP2E and CYP4A. Animals (Basel) 2023; 13:1939. [PMID: 37370449 DOI: 10.3390/ani13121939] [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: 04/04/2023] [Revised: 06/04/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
Cytochrome P450 is an important enzyme that metabolizes a variety of chemicals, including exogenous substances, such as drugs and environmental chemicals, and endogenous substances, such as steroids, fatty acids, and cholesterol. Some CYPs show interspecific differences in terms of genetic variation. As little is known about the mechanisms of elephant metabolism, we carried out a comparative genomic and phylogenetic analysis of CYP in elephants. Our results suggest that elephant CYP genes have undergone independent duplication, particularly in the CYP2A, CYP2C, and CYP3A genes, a unique cluster specific to elephant species. However, while CYP2E and CYP4A were conserved in other Afrotheria taxa, their decay in elephants resulted in genetic dysfunction (pseudogene). These findings outline several remarkable characteristics of elephant CYP1-4 genes and provide new insights into elephant xenobiotic metabolism. Further functional investigations are necessary to characterize elephant CYP, including expression patterns and interactions with drugs and sensitivities to other chemicals.
Collapse
Affiliation(s)
- Kanami Watanabe
- Laboratory of Toxicology, Department of Environmental Veterinary Science, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Mitsuki Kondo
- National Institute for Environmental Studies (NIES) Biodiversity Division, Ecological Risk Assessment and Control Section, Tsukuba 305-8506, Japan
| | - Yoshinori Ikenaka
- Laboratory of Toxicology, Department of Environmental Veterinary Science, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
- Water Research Group, Unit for Environmental Sciences and Management, North-West University, Potchefstroom 2520, South Africa
- Translational Research Unit, Veterinary Teaching Hospital, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
- One Health Research Center, Hokkaido University, Sapporo 060-0818, Japan
| | - Shouta M M Nakayama
- Laboratory of Toxicology, Department of Environmental Veterinary Science, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
- Biomedical Sciences Department, School of Veterinary Medicine, The University of Zambia, P.O. Box 32379, Lusaka 10101, Zambia
| | - Mayumi Ishizuka
- Laboratory of Toxicology, Department of Environmental Veterinary Science, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| |
Collapse
|
23
|
Hüttl M, Markova I, Miklánková D, Zapletalova I, Kujal P, Šilhavý J, Pravenec M, Malinska H. Hypolipidemic and insulin sensitizing effects of salsalate beyond suppressing inflammation in a prediabetic rat model. Front Pharmacol 2023; 14:1117683. [PMID: 37077818 PMCID: PMC10106727 DOI: 10.3389/fphar.2023.1117683] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 03/24/2023] [Indexed: 04/05/2023] Open
Abstract
Background and aims: Low-grade chronic inflammation plays an important role in the pathogenesis of metabolic syndrome, type 2 diabetes and their complications. In this study, we investigated the effects of salsalate, a non-steroidal anti-inflammatory drug, on metabolic disturbances in an animal model of prediabetes—a strain of non-obese hereditary hypertriglyceridemic (HHTg) rats.Materials and Methods: Adult male HHTg and Wistar control rats were fed a standard diet without or with salsalate delivering a daily dose of 200 mg/kg of body weight for 6 weeks. Tissue sensitivity to insulin action was measured ex vivo according to basal and insulin-stimulated 14C-U-glucose incorporation into muscle glycogen or adipose tissue lipids. The concentration of methylglyoxal and glutathione was determined using the HPLC-method. Gene expression was measured by quantitative RT-PCR.Results: Salsalate treatment of HHTg rats when compared to their untreated controls was associated with significant amelioration of inflammation, dyslipidemia and insulin resistance. Specificaly, salsalate treatment was associated with reduced inflammation, oxidative and dicarbonyl stress when inflammatory markers, lipoperoxidation products and methylglyoxal levels were significantly decreased in serum and tissues. In addition, salsalate ameliorated glycaemia and reduced serum lipid concentrations. Insulin sensitivity in visceral adipose tissue and skeletal muscle was significantly increased after salsalate administration. Further, salsalate markedly reduced hepatic lipid accumulation (triglycerides −29% and cholesterol −14%). Hypolipidemic effects of salsalate were associated with differential expression of genes coding for enzymes and transcription factors involved in lipid synthesis (Fas, Hmgcr), oxidation (Pparα) and transport (Ldlr, Abc transporters), as well as changes in gene expression of cytochrome P450 proteins, in particular decreased Cyp7a and increased Cyp4a isoforms.Conclusion: These results demonstrate important anti-inflammatory and anti-oxidative effects of salsalate that were associated with reduced dyslipidemia and insulin resistance in HHTg rats. Hypolipidemic effects of salsalate were associated with differential expression of genes regulating lipid metabolism in the liver. These results suggest potential beneficial use of salsalate in prediabetic patients with NAFLD symptoms.
Collapse
Affiliation(s)
- Martina Hüttl
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech
| | - Irena Markova
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech
| | - Denisa Miklánková
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech
| | - Iveta Zapletalova
- Department of Pharmacology, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech
| | - Petr Kujal
- Department of Pathology, Third Faculty of Medicine, Charles University, Prague, Czech
| | - Jan Šilhavý
- Institute of Physiology, Czech Academy of Sciences, Prague, Czech
| | - Michal Pravenec
- Institute of Physiology, Czech Academy of Sciences, Prague, Czech
| | - Hana Malinska
- Center for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech
- *Correspondence: Hana Malinska,
| |
Collapse
|
24
|
Wang GY, Zhang XY, Wang CJ, Guan YF. Emerging novel targets for nonalcoholic fatty liver disease treatment: Evidence from recent basic studies. World J Gastroenterol 2023; 29:75-95. [PMID: 36683713 PMCID: PMC9850950 DOI: 10.3748/wjg.v29.i1.75] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/29/2022] [Accepted: 12/14/2022] [Indexed: 01/04/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD), a leading chronic disease worldwide, affects approximately a quarter of the global population. Nonalcoholic steatohepatitis (NASH) is an advanced form of NAFLD and is more likely to progress to liver fibrosis than simple steatosis. NASH is also identified as the most rapidly growing cause of hepatocellular carcinoma. Although in the past decade, several phase II/III clinical trials have shown promising results in the use of novel drugs targeting lipid synthase, farnesoid X receptor signaling, peroxisome proliferator-activated receptor signaling, hepatocellular injury, and inflammatory signaling, proven pharmaceutical agents to treat NASH are still lacking. Thus, continuous exploration of the mechanism underlying the pathogenesis of NAFLD and the identification of novel therapeutic targets remain urgent tasks in the field. In the current review, we summarize studies reported in recent years that not only provide new insights into the mechanisms of NAFLD development but also explore the possibility of treating NAFLD by targeting newly identified signaling pathways. We also discuss evidence focusing on the intrahepatic targets involved in the pathogenesis of NAFLD as well as extrahepatic targets affecting liver metabolism and function.
Collapse
Affiliation(s)
- Guang-Yan Wang
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin 300070, China
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin 300070, China
- NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin 300070, China
| | - Xiao-Yan Zhang
- Health Science Center, East China Normal University, Shanghai 200241, China
| | - Chun-Jiong Wang
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin 300070, China
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin 300070, China
- NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin 300070, China
| | - You-Fei Guan
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian 116044, Liaoning Province, China
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Dalian Medical University, Dalian 116044, Liaoning Province, China
- Dalian Key Laboratory for Nuclear Receptors in Major Metabolic Diseases, Dalian 116044, Liaoning Province, China
| |
Collapse
|
25
|
Li H, Zheng J, Xu Q, Yang Y, Zhou J, Guo X, Cai Y, Cai JJ, Xie L, Awika J, Han X, Li Q, Kennedy L, Francis H, Glaser S, Huo Y, Alpini G, Wu C. Hepatocyte Adenosine Kinase Promotes Excessive Fat Deposition and Liver Inflammation. Gastroenterology 2023; 164:134-146. [PMID: 36181835 PMCID: PMC9772177 DOI: 10.1053/j.gastro.2022.09.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/23/2022] [Accepted: 09/20/2022] [Indexed: 02/03/2023]
Abstract
BACKGROUND & AIMS Nonalcoholic fatty liver disease is highly associated with obesity and progresses to nonalcoholic steatohepatitis when the liver develops overt inflammatory damage. While removing adenosine in the purine salvage pathway, adenosine kinase (ADK) regulates methylation reactions. We aimed to study whether hepatocyte ADK functions as an obesogenic gene/enzyme to promote excessive fat deposition and liver inflammation. METHODS Liver sections of human subjects were examined for ADK expression using immunohistochemistry. Mice with hepatocyte-specific ADK disruption or overexpression were examined for hepatic fat deposition and inflammation. Liver lipidomics, hepatocyte RNA sequencing (RNA-seq), and single-cell RNA-seq for liver nonparenchymal cells were performed to analyze ADK regulation of hepatocyte metabolic responses and hepatocyte-nonparenchymal cells crosstalk. RESULTS Whereas patients with nonalcoholic fatty liver disease had increased hepatic ADK levels, mice with hepatocyte-specific ADK disruption displayed decreased hepatic fat deposition on a chow diet and were protected from diet-induced excessive hepatic fat deposition and inflammation. In contrast, mice with hepatocyte-specific ADK overexpression displayed increased body weight and adiposity and elevated degrees of hepatic steatosis and inflammation compared with control mice. RNA-seq and epigenetic analyses indicated that ADK increased hepatic DNA methylation and decreased hepatic Ppara expression and fatty acid oxidation. Lipidomic and single-cell RNA-seq analyses indicated that ADK-driven hepatocyte factors, due to mitochondrial dysfunction, enhanced macrophage proinflammatory activation in manners involving increased expression of stimulator of interferon genes. CONCLUSIONS Hepatocyte ADK functions to promote excessive fat deposition and liver inflammation through suppressing hepatocyte fatty acid oxidation and producing hepatocyte-derived proinflammatory mediators. Therefore, hepatocyte ADK is a therapeutic target for managing obesity and nonalcoholic fatty liver disease.
Collapse
Affiliation(s)
- Honggui Li
- Department of Nutrition, Texas A&M University, College Station, Texas
| | - Juan Zheng
- Department of Nutrition, Texas A&M University, College Station, Texas
| | - Qian Xu
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas
| | - Yongjian Yang
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas
| | - Jing Zhou
- Department of Nutrition, Texas A&M University, College Station, Texas
| | - Xinlei Guo
- Department of Nutrition, Texas A&M University, College Station, Texas
| | - Yongfeng Cai
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - James J Cai
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, Texas
| | - Linglin Xie
- Department of Nutrition, Texas A&M University, College Station, Texas
| | - Joseph Awika
- Department of Food Science and Technology, Texas A&M University, College Station, Texas; Department of Soil and Crop Sciences, Texas A&M University, College Station, Texas
| | - Xianlin Han
- Barshop Institute for Longevity and Aging Studies and Department of Medicine, Division of Diabetes, University of Texas Health San Antonio, San Antonio, Texas
| | - Qingsheng Li
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska
| | - Lindsey Kennedy
- Hepatology and Gastroenterology, Medicine, Indiana University, Indianapolis, Indiana; Richard L. Roudebush Veterans Affairs Medical Center, Indianapolis, Indiana
| | - Heather Francis
- Hepatology and Gastroenterology, Medicine, Indiana University, Indianapolis, Indiana; Richard L. Roudebush Veterans Affairs Medical Center, Indianapolis, Indiana
| | - Shannon Glaser
- Department of Medical Physiology, Texas A&M University College of Medicine, Bryan, Texas
| | - Yuqing Huo
- Vascular Biology Center, Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Gianfranco Alpini
- Hepatology and Gastroenterology, Medicine, Indiana University, Indianapolis, Indiana; Richard L. Roudebush Veterans Affairs Medical Center, Indianapolis, Indiana
| | - Chaodong Wu
- Department of Nutrition, Texas A&M University, College Station, Texas.
| |
Collapse
|
26
|
Heintz MM, Eccles JA, Olack EM, Maner-Smith KM, Ortlund EA, Baldwin WS. Human CYP2B6 produces oxylipins from polyunsaturated fatty acids and reduces diet-induced obesity. PLoS One 2022; 17:e0277053. [PMID: 36520866 PMCID: PMC9754190 DOI: 10.1371/journal.pone.0277053] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 10/18/2022] [Indexed: 12/23/2022] Open
Abstract
Multiple factors in addition to over consumption lead to obesity and non-alcoholic fatty liver disease (NAFLD) in the United States and worldwide. CYP2B6 is the only human detoxification CYP whose loss is associated with obesity, and Cyp2b-null mice show greater diet-induced obesity with increased steatosis than wildtype mice. However, a putative mechanism has not been determined. LC-MS/MS revealed that CYP2B6 metabolizes PUFAs, with a preference for metabolism of ALA to 9-HOTrE and to a lesser extent 13-HOTrE with a preference for metabolism of PUFAs at the 9- and 13-positions. To further study the role of CYP2B6 in vivo, humanized-CYP2B6-transgenic (hCYP2B6-Tg) and Cyp2b-null mice were fed a 60% high-fat diet for 16 weeks. Compared to Cyp2b-null mice, hCYP2B6-Tg mice showed reduced weight gain and metabolic disease as measured by glucose tolerance tests, however hCYP2B6-Tg male mice showed increased liver triglycerides. Serum and liver oxylipin metabolite concentrations increased in male hCYP2B6-Tg mice, while only serum oxylipins increased in female hCYP2B6-Tg mice with the greatest increases in LA oxylipins metabolized at the 9 and 13-positions. Several of these oxylipins, specifically 9-HODE, 9-HOTrE, and 13-oxoODE, are PPAR agonists. RNA-seq data also demonstrated sexually dimorphic changes in gene expression related to nuclear receptor signaling, especially CAR > PPAR with qPCR suggesting PPARγ signaling is more likely than PPARα signaling in male mice. Overall, our data indicates that CYP2B6 is an anti-obesity enzyme, but probably to a lesser extent than murine Cyp2b's. Therefore, the inhibition of CYP2B6 by xenobiotics or dietary fats can exacerbate obesity and metabolic disease potentially through disrupted PUFA metabolism and the production of key lipid metabolites.
Collapse
Affiliation(s)
- Melissa M. Heintz
- Biological Sciences, Clemson University, Clemson, South Carolina, United States of America
| | - Jazmine A. Eccles
- Biological Sciences, Clemson University, Clemson, South Carolina, United States of America
| | - Emily M. Olack
- Biological Sciences, Clemson University, Clemson, South Carolina, United States of America
| | - Kristal M. Maner-Smith
- Emory Integrated Metabolomics and Lipodomics Core, Emory University, Atlanta, Georgia, United States of America
| | - Eric A. Ortlund
- Department of Biochemistry, Emory University School of Medicine, Emory University, Atlanta, Georgia, United States of America
| | - William S. Baldwin
- Biological Sciences, Clemson University, Clemson, South Carolina, United States of America
- * E-mail:
| |
Collapse
|
27
|
Shi F, Almerick T Boncan D, Wan HT, Chan TF, Zhang EL, Lai KP, Wong CKC. Hepatic metabolism gene expression and gut microbes in offspring, subjected to in-utero PFOS exposure and postnatal diet challenges. CHEMOSPHERE 2022; 308:136196. [PMID: 36041519 DOI: 10.1016/j.chemosphere.2022.136196] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/19/2022] [Accepted: 08/22/2022] [Indexed: 06/15/2023]
Abstract
We examined the changes in hepatic metabolic gene expression and gut microbiota of offspring exposed to PFOS in-utero. At GD17.5, our data showed that PFOS exposure decreased fetal bodyweights and hepatic metabolic gene expressions but increased relative liver mass and lipid accumulation. At PND21, in-utero high-dose PFOS-exposed offspring exhibited significantly greater bodyweight (catch-up-growth), associated with significant induction of hepatic metabolic gene expression. In addition, 16SrRNA-sequencing of the cecal samples revealed an increase in carbohydrate catabolism but a reduction in microbial polysaccharide synthesis and short-chain fatty acid (SCFA) metabolism. From PND21-80, a postnatal diet-challenge for the offspring was conducted. At PND80 under a normal diet, in-utero high-dose PFOS-exposed offspring maintained the growth "catch-up" effect. In contrast, in a high-fat-diet, the bodyweight of in-utero high-dose PFOS-exposed adult offspring were significantly lesser than the corresponding low-dose and control groups. Even though in the high-fat-diet, the in-utero PFOS-exposed adult offspring showed significant upregulation of hepatic metabolic genes, the lower bodyweight suggests that they had difficulty utilizing high-fat nutrients. Noteworthy, the metagenomic data showed a significant reduction in the biosynthesis of microbial polysaccharides, vitamin B, and SCFAs in the PFOS-exposed adult offspring. Furthermore, the observed effects were significantly reduced in the PFOS-exposed adult offspring with the high-fat diet but supplemented with sucrose. Our study demonstrated that in-utero PFOS exposure caused inefficient fat metabolism and increased the risk of hepatic steatosis in offspring.
Collapse
Affiliation(s)
- Feng Shi
- State Key Laboratory in Environmental and Biological Analysis, Croucher Institute for Environmental Sciences, Department of Biology, Hong Kong Baptist University, Hong Kong SAR, China
| | - Delbert Almerick T Boncan
- School of Life Sciences, State Key Laboratory of Agrobiotechnology, Hong Kong Bioinformatics Centre, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Hin Ting Wan
- State Key Laboratory in Environmental and Biological Analysis, Croucher Institute for Environmental Sciences, Department of Biology, Hong Kong Baptist University, Hong Kong SAR, China
| | - Ting Fung Chan
- School of Life Sciences, State Key Laboratory of Agrobiotechnology, Hong Kong Bioinformatics Centre, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Eric L Zhang
- Department of Computer Science, Hong Kong Baptist University, Hong Kong SAR, China
| | - Keng Po Lai
- Laboratory of Environmental Pollution and Integrative Omics, Guilin Medical University, Guilin, PR China
| | - Chris Kong-Chu Wong
- State Key Laboratory in Environmental and Biological Analysis, Croucher Institute for Environmental Sciences, Department of Biology, Hong Kong Baptist University, Hong Kong SAR, China.
| |
Collapse
|
28
|
20-Hydroxytetraenoic acid induces hepatic fibrosis via the TGF-β1/Smad3 signaling pathway. Toxicol Lett 2022; 373:1-12. [PMID: 36368619 DOI: 10.1016/j.toxlet.2022.11.001] [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: 07/11/2022] [Revised: 10/24/2022] [Accepted: 11/07/2022] [Indexed: 11/10/2022]
Abstract
Hepatic fibrosis is caused by excessive accumulation of extracellular matrix (ECM) due to repeated liver injury. Hepatic stellate cells (HSCs) play a key role in the pathogenesis and progression of hepatic fibrosis. A study showed that CYP4A14 gene defect can inhibit hepatic fibrosis, but the specific mechanism was not clear. In this experiment, patients with hepatic fibrosis, LX-2 cells (a human HSCs line), and mice with liver fibrosis induced by carbon tetrachloride (CCl4) were used to study the effect of 20-Hydroxytetraenoic acid (20-HETE), one of the main metabolites of arachidonic acid (AA) catalyzed by CYP4A enzyme, on hepatic fibrosis and its mechanism. Our experimental results showed that the 20-HETE of patients with hepatic fibrosis is significantly higher than that of normal people and is closely related to the degree of fibrosis. 20-HETE could induce activation of LX-2 cells and 20-HETE antagonist could inhibit the induction of 20-HETE. 20-HETE was significantly increased in CCl4-induced liver fibrosis mice and inhibition of 20-HETE production could attenuate hepatic fibrosis. 20-HETE induced hepatic fibrosis mainly via the TGF- β1/Smad3 signal pathway. In conclusion, the results suggest that 20-HETE plays an important role in hepatic fibrosis and may be a possible target for the clinical treatment of hepatic fibrosis.
Collapse
|
29
|
Krøyer Rasmussen M, Thøgersen R, Horsbøl Lindholm P, Bertram HC, Pilegaard H. Hepatic PGC-1α has minor regulatory effect on the transcriptome and metabolome during high fat high fructose diet and exercise. Gene 2022; 851:147039. [DOI: 10.1016/j.gene.2022.147039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/31/2022] [Accepted: 11/04/2022] [Indexed: 11/10/2022]
|
30
|
Zhang L, Chen H, Cui C, Liang L, Ge H, Meng L, Zhang C. Effects of oocyte vitrification on gene expression in the liver and kidney tissues of adult offspring. J Assist Reprod Genet 2022; 39:2635-2646. [PMID: 36223009 PMCID: PMC9723011 DOI: 10.1007/s10815-022-02611-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 08/31/2022] [Indexed: 12/14/2022] Open
Abstract
Oocyte vitrification is an important assisted reproductive technology (ART) that preserves the fertility of unmarried patients with malignant tumors, and promotes the development of the oocyte donation program. In recent years, the effects of ART, including the vitrification of oocytes and embryos on the health of offspring, have attracted much attention; however, it is difficult to conduct long-term follow-up and biochemical evaluation in humans. In this study, we detected the effect of oocyte vitrification on gene expression in the organs of adult mice offspring by RNA sequencing for the first time. Our results showed that only a small amount of gene expression was significantly affected. Seven genes (Tpm3, Hspe1-rs1, Ntrk2, Cyp4a31, Asic5, Cyp4a14, Retsat) were abnormally expressed in the liver, and ten genes (Lbp, Hspe1-rs1, Prxl2b, Pfn3, Gm9008, Bglap3, Col8a1, Hmgcr, Ero1lb, Ifi44l) were abnormal in the kidney. Several genes were related to metabolism and disease occurrence in the liver or kidney. Besides, we paid special attention to the expression of known imprinted genes and DNA methylation-related genes in adult organs, which are susceptible to oocyte cryopreservation in the preimplantation stage. As a result, some of these transcripts were detected in adult organs, but they were not affected by oocyte vitrification. In conclusion, we first report that oocyte vitrification did not significantly change the global gene expression in offspring organs; nonetheless, it can still influence the transcription of a few functional genes. The potential adverse effects caused by oocyte vitrification need attention and further study.
Collapse
Affiliation(s)
- Lei Zhang
- Reproductive Medicine Center, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital of Henan University, Zhengzhou, Henan, China
- Henan Joint International Research Laboratory of Reproductive Bioengineering, Zhengzhou, Henan Province, China
| | - Huanhuan Chen
- Reproductive Medicine Center, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital of Henan University, Zhengzhou, Henan, China
- Henan Joint International Research Laboratory of Reproductive Bioengineering, Zhengzhou, Henan Province, China
| | - Chenchen Cui
- Reproductive Medicine Center, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital of Henan University, Zhengzhou, Henan, China
- Henan Joint International Research Laboratory of Reproductive Bioengineering, Zhengzhou, Henan Province, China
| | - Linlin Liang
- Reproductive Medicine Center, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital of Henan University, Zhengzhou, Henan, China
- Henan Joint International Research Laboratory of Reproductive Bioengineering, Zhengzhou, Henan Province, China
| | - Hengtao Ge
- Reproductive Medicine Center, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital of Henan University, Zhengzhou, Henan, China
- Henan Joint International Research Laboratory of Reproductive Bioengineering, Zhengzhou, Henan Province, China
| | - Li Meng
- Reproductive Medicine Center, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital of Henan University, Zhengzhou, Henan, China.
- Henan Joint International Research Laboratory of Reproductive Bioengineering, Zhengzhou, Henan Province, China.
| | - Cuilian Zhang
- Reproductive Medicine Center, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital of Henan University, Zhengzhou, Henan, China.
- Henan Joint International Research Laboratory of Reproductive Bioengineering, Zhengzhou, Henan Province, China.
| |
Collapse
|
31
|
Raza GS, Sodum N, Kaya Y, Herzig KH. Role of Circadian Transcription Factor Rev-Erb in Metabolism and Tissue Fibrosis. Int J Mol Sci 2022; 23:12954. [PMID: 36361737 PMCID: PMC9655416 DOI: 10.3390/ijms232112954] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/21/2022] [Accepted: 10/22/2022] [Indexed: 09/12/2023] Open
Abstract
Circadian rhythms significantly affect metabolism, and their disruption leads to cardiometabolic diseases and fibrosis. The clock repressor Rev-Erb is mainly expressed in the liver, heart, lung, adipose tissue, skeletal muscles, and brain, recognized as a master regulator of metabolism, mitochondrial biogenesis, inflammatory response, and fibrosis. Fibrosis is the response of the body to injuries and chronic inflammation with the accumulation of extracellular matrix in tissues. Activation of myofibroblasts is a key factor in the development of organ fibrosis, initiated by hormones, growth factors, inflammatory cytokines, and mechanical stress. This review summarizes the importance of Rev-Erb in ECM remodeling and tissue fibrosis. In the heart, Rev-Erb activation has been shown to alleviate hypertrophy and increase exercise capacity. In the lung, Rev-Erb agonist reduced pulmonary fibrosis by suppressing fibroblast differentiation. In the liver, Rev-Erb inhibited inflammation and fibrosis by diminishing NF-κB activity. In adipose tissue, Rev- Erb agonists reduced fat mass. In summary, the results of multiple studies in preclinical models demonstrate that Rev-Erb is an attractive target for positively influencing dysregulated metabolism, inflammation, and fibrosis, but more specific tools and studies would be needed to increase the information base for the therapeutic potential of these substances interfering with the molecular clock.
Collapse
Affiliation(s)
- Ghulam Shere Raza
- Research Unit of Biomedicine, Medical Research Center, Faculty of Medicine, University of Oulu, 90220 Oulu, Finland
| | - Nalini Sodum
- Research Unit of Biomedicine, Medical Research Center, Faculty of Medicine, University of Oulu, 90220 Oulu, Finland
| | - Yagmur Kaya
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Marmara University, 34854 Istanbul, Turkey
| | - Karl-Heinz Herzig
- Research Unit of Biomedicine, Medical Research Center, Faculty of Medicine, University of Oulu, 90220 Oulu, Finland
- Oulu University Hospital, University of Oulu, 90220 Oulu, Finland
- Pediatric Gastroenterology and Metabolic Diseases, Pediatric Institute, Poznan University of Medical Sciences, 60-572 Poznań, Poland
| |
Collapse
|
32
|
Weber AA, Yang X, Mennillo E, Ding J, Watrous JD, Jain M, Chen S, Karin M, Tukey RH. Lactational delivery of Triclosan promotes non-alcoholic fatty liver disease in newborn mice. Nat Commun 2022; 13:4346. [PMID: 35896521 PMCID: PMC9329322 DOI: 10.1038/s41467-022-31947-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 07/11/2022] [Indexed: 11/21/2022] Open
Abstract
Here we show that Triclosan (TCS), a high-volume antimicrobial additive that has been detected in human breastmilk, can be efficiently transferred by lactation to newborn mice, causing significant fatty liver (FL) during the suckling period. These findings are relevant since pediatric non-alcoholic fatty liver disease (NAFLD) is escalating in the United States, with a limited mechanistic understanding. Lactational delivery stimulated hepatosteatosis, triglyceride accumulation, endoplasmic reticulum (ER) stress, signs of inflammation, and liver fibrosis. De novo lipogenesis (DNL) induced by lactational TCS exposure is shown to be mediated in a PERK-eIF2α-ATF4-PPARα cascade. The administration of obeticholic acid (OCA), a potent FXR agonist, as well as activation of intestinal mucosal-regenerative gp130 signaling, led to reduced liver ATF4 expression, PPARα signaling, and DNL when neonates were exposed to TCS. It is yet to be investigated but mother to child transmission of TCS or similar toxicants may underlie the recent increases in pediatric NAFLD.
Collapse
Affiliation(s)
- André A Weber
- Laboratory of Environmental Toxicology, Department of Pharmacology, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Xiaojing Yang
- Laboratory of Environmental Toxicology, Department of Pharmacology, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Elvira Mennillo
- Laboratory of Environmental Toxicology, Department of Pharmacology, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Jeffrey Ding
- Departments of Medicine and Pharmacology, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Jeramie D Watrous
- Departments of Medicine and Pharmacology, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Mohit Jain
- Departments of Medicine and Pharmacology, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Shujuan Chen
- Laboratory of Environmental Toxicology, Department of Pharmacology, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Robert H Tukey
- Laboratory of Environmental Toxicology, Department of Pharmacology, University of California, San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
33
|
Soundharrajan I, Karnan M, Jung JS, Lee KD, Lee JC, Ramesh T, Kim D, Choi KC. A Transcriptomic Response to Lactiplantibacillus plantarum-KCC48 against High-Fat Diet-Induced Fatty Liver Diseases in Mice. Int J Mol Sci 2022; 23:6750. [PMID: 35743193 PMCID: PMC9224190 DOI: 10.3390/ijms23126750] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 06/15/2022] [Accepted: 06/15/2022] [Indexed: 02/05/2023] Open
Abstract
The most prevalent chronic liver disorder in the world is fatty liver disease caused by a high-fat diet. We examined the effects of Lactiplantibacillus plantarum-KCC48 on high-fat diet-induced (HFD) fatty liver disease in mice. We used the transcriptome tool to perform a systematic evaluation of hepatic mRNA transcripts changes in high-fat diet (HFD)-fed animals and high-fat diet with L. plantarum (HFLPD)-fed animals. HFD causes fatty liver diseases in animals, as evidenced by an increase in TG content in liver tissues compared to control animals. Based on transcriptome data, 145 differentially expressed genes (DEGs) were identified in the liver of HFD-fed mice compared to control mice. Moreover, 61 genes were differentially expressed in the liver of mice fed the HFLPD compared to mice fed the HFD. Additionally, 43 common DEGs were identified between HFD and HFLPD. These genes were enriched in metabolic processes, retinol metabolism, the PPAR signaling pathway, fatty acid degradation, arachidonic metabolism, and steroid hormone synthesis. Taking these data into consideration, it can be concluded that L. plantarum-KCC48 treatment significantly regulates the expression of genes involved in hepatosteatosis caused by HFD, which may prevent fatty liver disease.
Collapse
Affiliation(s)
- Ilavenil Soundharrajan
- Grassland and Forage Division, Rural Development Administration, National Institute of Animal Science, Cheonan 31000, Korea; (I.S.); (M.K.); (J.-S.J.)
| | - Muthusamy Karnan
- Grassland and Forage Division, Rural Development Administration, National Institute of Animal Science, Cheonan 31000, Korea; (I.S.); (M.K.); (J.-S.J.)
| | - Jeong-Sung Jung
- Grassland and Forage Division, Rural Development Administration, National Institute of Animal Science, Cheonan 31000, Korea; (I.S.); (M.K.); (J.-S.J.)
| | - Kyung-Dong Lee
- Department of Companion Animals, Dongsin University, Naju 58245, Korea;
| | - Jeong-Chae Lee
- Department of Bioactive Material Sciences and Research Center of Bioactive Materials, Jeonbuk National University, Jeonju 54896, Korea;
| | - Thiyagarajan Ramesh
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia;
| | - Dahye Kim
- Animal Genomics and Bioinformatics Division, National Institute of Animal Science, Wanju 55365, Korea
| | - Ki-Choon Choi
- Grassland and Forage Division, Rural Development Administration, National Institute of Animal Science, Cheonan 31000, Korea; (I.S.); (M.K.); (J.-S.J.)
| |
Collapse
|
34
|
Zhang B, Chen T, Cao M, Yuan C, Reiter RJ, Zhao Z, Zhao Y, Chen L, Fan W, Wang X, Zhou X, Li C. Gut Microbiota Dysbiosis Induced by Decreasing Endogenous Melatonin Mediates the Pathogenesis of Alzheimer's Disease and Obesity. Front Immunol 2022; 13:900132. [PMID: 35619714 PMCID: PMC9127079 DOI: 10.3389/fimmu.2022.900132] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 04/13/2022] [Indexed: 01/15/2023] Open
Abstract
Lifestyle choices, external environment, aging, and other factors influence the synthesis of melatonin. Although the physiological functions of melatonin have been widely studied in relation to specific organs, the systemic effects of endogenous melatonin reduction has not been reported. This study evaluates the systemic changes and possible pathogenic risks in an endogenous melatonin reduction (EMR) mouse model deficient in the rate limiting enzyme in melatonin production, arylalkylamine N-acetyltransferase (Aanat) gene. Using this model, we identified a new relationship between melatonin, Alzheimer’s disease (AD), and gut microbiota. Systematic changes were evaluated using multi-omics analysis. Fecal microbiota transplantation (FMT) was performed to examine the role of gut microbiota in the pathogenic risks of EMR. EMR mice exhibited a pan-metabolic disorder, with significant transcriptome changes in 11 organs, serum metabolome alterations as well as microbiota dysbiosis. Microbiota dysbiosis was accompanied by increased gut permeability along with gut and systemic inflammation. Correlation analysis revealed that systemic inflammation may be related to the increase of Ruminiclostridium_5 relative abundance. 8-month-old EMR mice had AD-like phenotypes, including Iba-1 activation, A β protein deposition and decreased spatial memory ability. Moreover, EMR mice showed decreased anti stress ability, under high-fat diet, EMR mice had greater body weight and more obvious hepatic steatosis compared with WT group. FMT improved gut permeability, systemic inflammation, and AD-related phenotypes, while reducing obesity in EMR mice. Our findings suggest EMR causes systemic changes mediated by gut microbiota dysbiosis, which may be a pathogenic factor for AD and obesity, we further proved the gut microbiota is a potential target for the prevention and treatment of AD and obesity.
Collapse
Affiliation(s)
- Boqi Zhang
- College of Animal Sciences, Jilin University, Changchun, China
| | - Tong Chen
- College of Animal Sciences, Jilin University, Changchun, China
| | - Maosheng Cao
- College of Animal Sciences, Jilin University, Changchun, China
| | - Chenfeng Yuan
- College of Animal Sciences, Jilin University, Changchun, China
| | - Russel J Reiter
- Department of Cellular and Structural Biology, University of Texas Health Science Center, San Antonio, TX, United States
| | - Zijiao Zhao
- College of Animal Sciences, Jilin University, Changchun, China
| | - Yun Zhao
- College of Animal Sciences, Jilin University, Changchun, China
| | - Lu Chen
- College of Animal Sciences, Jilin University, Changchun, China
| | - Wenjing Fan
- College of Animal Sciences, Jilin University, Changchun, China
| | - Xin Wang
- College of Animal Sciences, Jilin University, Changchun, China
| | - Xu Zhou
- College of Animal Sciences, Jilin University, Changchun, China
| | - Chunjin Li
- College of Animal Sciences, Jilin University, Changchun, China
| |
Collapse
|
35
|
Zhang H, Li Y, Zhang C, Huang K, Zhao J, Le S, Jiang L, Liu H, Yang P, Xiao X, Yu J, Wu J, Ye P, Xia J. B-cell lymphoma 6 alleviates nonalcoholic fatty liver disease in mice through suppression of fatty acid transporter CD36. Cell Death Dis 2022; 13:359. [PMID: 35436984 PMCID: PMC9016081 DOI: 10.1038/s41419-022-04812-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 03/22/2022] [Accepted: 03/30/2022] [Indexed: 02/06/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is an ubiquitous disease that exists across a wide spectrum ranging from steatosis, steatohepatitis, advanced fibrosis, and liver cirrhosis. Hallmarks of NAFLD are lipid accumulation, insulin resistance, and chronic low-grade inflammation. However, there currently are no medications approved for NAFLD. B-cell lymphoma 6 (BCL6) is a transcriptional inhibitor that is vital for germinal center B-cell formation. Our study identified BCL6 as a critical modulator of hepatic lipid metabolism and appears to contribute to the initiation and progression of NAFLD. In our research, we induced hepatic BCL6 overexpression using adeno-associated virus (AAV), as well as conditional liver-specific BCL6 knockout mice (BCL6-CKO). With these models, we noted that BCL6 overexpression improved insulin resistance and hepatic steatosis in mice models maintained on a HFD diet. Conversely, these parameters worsened in the livers of mice with downregulated BCL6 levels. Mechanistically, the translocase fatty acid CD36 was determined to be a transcriptional target of BCL6 that influences its role in hepatic steatosis. BCL6 bound directly to the CD36 promoter region, restraining CD36 transcription under physiological conditions. We conclude that the hepatocyte BCL6 inhibits the NAFLD progression in mice, including deranged lipid accumulation and glucose metabolism, through a CD36-dependent manner. These results indicate that BCL6 may potentially be targeted in NAFLD treatment.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yue Li
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chao Zhang
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kun Huang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Zhao
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sheng Le
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lang Jiang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hao Liu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Peiwen Yang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyue Xiao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jizhang Yu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jie Wu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Ping Ye
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Jiahong Xia
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
36
|
Scalcon V, Folda A, Lupo MG, Tonolo F, Pei N, Battisti I, Ferri N, Arrigoni G, Bindoli A, Holmgren A, Coppo L, Rigobello MP. Mitochondrial depletion of glutaredoxin 2 induces metabolic dysfunction-associated fatty liver disease in mice. Redox Biol 2022; 51:102277. [PMID: 35290904 PMCID: PMC8921303 DOI: 10.1016/j.redox.2022.102277] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/18/2022] [Accepted: 02/26/2022] [Indexed: 12/13/2022] Open
Abstract
Glutaredoxin 2 (Grx2) is a glutathione-dependent oxidoreductase that facilitates glutathionylation/de-glutathionylation of target proteins. The main variants of Grx2 are the mitochondrial Grx2a and the cytosolic Grx2c. The aim of this study was to investigate the specific role of mitochondrial Grx2 in vivo using a mitochondrial Grx2 depleted (mGD) mouse model. mGD mice displayed an altered mitochondrial morphology and functioning. Furthermore, the lack of Grx2 in the mitochondrial compartment is responsible for increased blood lipid levels under a normal diet, a metabolic dysfunction-associated fatty liver disease (MAFLD) phenotype and a decreased glycogen storage capacity. In addition, depleting Grx2a leads to an alteration in abundance and in glutathionylation pattern of different mitochondrial enzymes, highlighting the selective role of Grx2 in the regulation of metabolic pathways. Overall, our findings identify the involvement of mitochondrial Grx2a in the regulation of cell metabolism and highlight a previously unknown association between Grx2 and MAFLD. Mitochondrial Grx2 depleted (mGD) mice display mitochondrial impairment. mGD mice show alterations in lipid metabolism and glycogen storage in the liver. Upon Grx2 deficiency the glutathionylation pattern of metabolic enzymes changes Mitochondrial Grx2 depletion is associated with MAFLD development
Collapse
Affiliation(s)
- Valeria Scalcon
- Department of Biomedical Sciences, University of Padova, 35131, Padova, Italy
| | - Alessandra Folda
- Department of Biomedical Sciences, University of Padova, 35131, Padova, Italy
| | | | - Federica Tonolo
- Department of Biomedical Sciences, University of Padova, 35131, Padova, Italy
| | - Naixuan Pei
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-17165, Stockholm, Sweden
| | - Ilaria Battisti
- Department of Biomedical Sciences, University of Padova, 35131, Padova, Italy; Proteomics Center, University of Padova and Azienda Ospedaliera di Padova, 35129, Padova, Italy
| | - Nicola Ferri
- Department of Medicine, University of Padova, 35121, Padova, Italy
| | - Giorgio Arrigoni
- Department of Biomedical Sciences, University of Padova, 35131, Padova, Italy; Proteomics Center, University of Padova and Azienda Ospedaliera di Padova, 35129, Padova, Italy
| | - Alberto Bindoli
- Institute of Neuroscience, CNR c/o Department of Biomedical Sciences, University of Padova, 35131, Padova, Italy
| | - Arne Holmgren
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-17165, Stockholm, Sweden
| | - Lucia Coppo
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-17165, Stockholm, Sweden.
| | - Maria Pia Rigobello
- Department of Biomedical Sciences, University of Padova, 35131, Padova, Italy.
| |
Collapse
|
37
|
Hu R. Grifola frondosa may play an anti-obesity role by affecting intestinal microbiota to increase the production of short-chain fatty acids. Front Endocrinol (Lausanne) 2022; 13:1105073. [PMID: 36733799 PMCID: PMC9886863 DOI: 10.3389/fendo.2022.1105073] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 12/29/2022] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Grifola frondosa (G. frondosa) is a fungus with good economic exploitation prospects of food and medicine homologation. This study aims to investigate the effects of G. frondosa powder suspension (GFPS) on the intestinal contents microbiota and the indexes related to oxidative stress and energy metabolism in mice, to provide new ideas for developing G. frondosa weight loss products. METHODS Twenty Kunming mice were randomly divided into control (CC), low-dose GFPS (CL), medium-dose GFPS (CM), and high-dose GFPS (CH) groups. The mice in CL, CM, and CH groups were intragastrically administered with 1.425 g/(kg·d), 2.85 g/(kg·d), and 5.735 g/(kg·d) GFPS, respectively. The mice in CC group were given the same dose of sterile water. After 8 weeks, liver and muscle related oxidative stress and energy metabolism indicators were detected, and the intestinal content microbiota of the mice was detected by 16S rRNA high-throughput sequencing. RESULTS After eight weeks of GFPS intervention, all mice lost weight. Compared with the CC group, lactate dehydrogenase (LDH) and malondialdehyde (MDA) contents in CL, CM, and CH groups were increased, while Succinate dehydrogenase (SDH) and Superoxide Dismutase (SOD) contents in the liver were decreased. The change trends of LDH and SDH in muscle were consistent with those in the liver. Among the above indexes, the change in CH is the most significant. The Chao1, ACE, Shannon, and Simpson index in CL, CM, and CH groups were increased. In the taxonomic composition, after the intervention with GFPS, the short-chain fatty acid (SCFA)-producing bacteria such as unclassified Muribaculaceae, Alloprevotella, and unclassified Lachnospiraceae increased. In linear discriminant analysis effect size (LEfSe) analysis, the characteristic bacteria in CC, CL, CM, and CH groups showed significant differences. In addition, some characteristic bacteria significantly correlated with related energy metabolism indicators. CONCLUSION The preventive effect of G. frondosa on obesity is related to changing the structure of intestinal content microbiota and promoting the growth of SCFAs. While excessive intake of G. frondosa may not be conducive to the antioxidant capacity and energy metabolism.
Collapse
|
38
|
Jarukamjorn K, Sukkasem N, Chatuphonprasert W. Alteration of murine cytochrome P450 profiles in fatty liver disease by hesperidin and myricetin. Pharmacogn Mag 2022. [DOI: 10.4103/pm.pm_321_21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
39
|
Hafner H, Mulcahy MC, Carlson Z, Hartley P, Sun H, Westerhoff M, Qi N, Bridges D, Gregg B. Lactational High Fat Diet in Mice Causes Insulin Resistance and NAFLD in Male Offspring Which Is Partially Rescued by Maternal Metformin Treatment. Front Nutr 2021; 8:759690. [PMID: 34977118 PMCID: PMC8714922 DOI: 10.3389/fnut.2021.759690] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 10/26/2021] [Indexed: 01/16/2023] Open
Abstract
Maternal metabolic disease and diet during pregnancy and lactation have important implications for the programming of offspring metabolic disease. In addition, high-fat diets during pregnancy and lactation can predispose the offspring to non-alcoholic fatty liver disease (NAFLD), a rising health threat in the U.S. We developed a model of maternal high-fat feeding exclusively during the lactation period. We previously showed that offspring from dams, given lactational high-fat diet (HFD), are predisposed to obesity, glucose intolerance, and inflammation. In separate experiments, we also showed that lactational metformin treatment can decrease offspring metabolic risk. The purpose of these studies was to understand the programming implications of lactational HFD on offspring metabolic liver disease risk. Dams were fed a 60% lard-based HFD from the day of delivery through the 21-day lactation period. A subset of dams was also given metformin as a co-treatment. Starting at weaning, the offspring were fed normal fat diet until 3 months of age; at which point, a subset was challenged with an additional HFD stressor. Lactational HFD led male offspring to develop hepatic insulin resistance. The post-weaning HFD challenge led male offspring to progress to NAFLD with more severe outcomes in the lactational HFD-challenged offspring. Co-administration of metformin to lactating dams on HFD partially rescued the offspring liver metabolic defects in males. Lactational HFD or post-weaning HFD had no impact on female offspring who maintained a normal insulin sensitivity and liver phenotype. These findings indicate that HFD, during the lactation period, programs the adult offspring to NAFLD risk in a sexually dimorphic manner. In addition, early life intervention with metformin via maternal exposure may prevent some of the liver programming caused by maternal HFD.
Collapse
Affiliation(s)
- Hannah Hafner
- Division of Endocrinology, Department of Pediatrics, Michigan Medicine, Ann Arbor, MI, United States
| | - Molly C. Mulcahy
- Department of Nutritional Sciences, School of Public Health, University of Michigan, Ann Arbor, MI, United States
| | - Zach Carlson
- Division of Endocrinology, Department of Pediatrics, Michigan Medicine, Ann Arbor, MI, United States
| | - Phillip Hartley
- Department of Nutritional Sciences, School of Public Health, University of Michigan, Ann Arbor, MI, United States
| | - Haijing Sun
- Division of Endocrinology, Department of Pediatrics, Michigan Medicine, Ann Arbor, MI, United States
| | - Maria Westerhoff
- Department of Pathology, Michigan Medicine, Ann Arbor, MI, United States
| | - Nathan Qi
- Department of Molecular and Integrative Physiology, Michigan Medicine, Ann Arbor, MI, United States
| | - Dave Bridges
- Department of Nutritional Sciences, School of Public Health, University of Michigan, Ann Arbor, MI, United States
| | - Brigid Gregg
- Division of Endocrinology, Department of Pediatrics, Michigan Medicine, Ann Arbor, MI, United States
- Department of Nutritional Sciences, School of Public Health, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
40
|
Manek R, Zhang YV, Berthelette P, Hossain M, Cornell CS, Gans J, Anarat-Cappillino G, Geller S, Jackson R, Yu D, Singh K, Ryan S, Bangari DS, Xu EY, Kyostio-Moore SRM. Blood phenylalanine reduction reverses gene expression changes observed in a mouse model of phenylketonuria. Sci Rep 2021; 11:22886. [PMID: 34819582 PMCID: PMC8613214 DOI: 10.1038/s41598-021-02267-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 11/09/2021] [Indexed: 12/02/2022] Open
Abstract
Phenylketonuria (PKU) is a genetic deficiency of phenylalanine hydroxylase (PAH) in liver resulting in blood phenylalanine (Phe) elevation and neurotoxicity. A pegylated phenylalanine ammonia lyase (PEG-PAL) metabolizing Phe into cinnamic acid was recently approved as treatment for PKU patients. A potentially one-time rAAV-based delivery of PAH gene into liver to convert Phe into tyrosine (Tyr), a normal way of Phe metabolism, has now also entered the clinic. To understand differences between these two Phe lowering strategies, we evaluated PAH and PAL expression in livers of PAHenu2 mice on brain and liver functions. Both lowered brain Phe and increased neurotransmitter levels and corrected animal behavior. However, PAL delivery required dose optimization, did not elevate brain Tyr levels and resulted in an immune response. The effect of hyperphenylalanemia on liver functions in PKU mice was assessed by transcriptome and proteomic analyses. We observed an elevation in Cyp4a10/14 proteins involved in lipid metabolism and upregulation of genes involved in cholesterol biosynthesis. Majority of the gene expression changes were corrected by PAH and PAL delivery though the role of these changes in PKU pathology is currently unclear. Taken together, here we show that blood Phe lowering strategy using PAH or PAL corrects both brain pathology as well as previously unknown lipid metabolism associated pathway changes in liver.
Collapse
Affiliation(s)
- Rachna Manek
- Genomic Medicine Unit, Sanofi, Framingham, MA, USA.
| | - Yao V Zhang
- Genomic Medicine Unit, Sanofi, Framingham, MA, USA
| | | | | | | | - Joseph Gans
- Translational Sciences, Sanofi, Framingham, MA, USA
| | | | - Sarah Geller
- Pre-Development Sciences NA, Analytical R&D, Sanofi, Framingham, MA, USA
| | | | - Dan Yu
- Genomic Medicine Unit, Sanofi, Framingham, MA, USA
| | - Kuldeep Singh
- Global Discovery Pathology, Sanofi, Framingham, MA, USA
| | - Sue Ryan
- Global Discovery Pathology, Sanofi, Framingham, MA, USA
| | | | - Ethan Y Xu
- Translational Sciences, Sanofi, Framingham, MA, USA
- Excision BioTherapeutics, Cambridge, MA, USA
| | | |
Collapse
|
41
|
Sellers J, Brooks A, Fernando S, Westenberger G, Junkins S, Smith S, Min K, Lawan A. Fasting-Induced Upregulation of MKP-1 Modulates the Hepatic Response to Feeding. Nutrients 2021; 13:nu13113941. [PMID: 34836195 PMCID: PMC8619756 DOI: 10.3390/nu13113941] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 10/27/2021] [Accepted: 11/02/2021] [Indexed: 12/14/2022] Open
Abstract
The liver plays a key role in whole-body, glucose and lipid homeostasis. Nutritional signals in response to fasting and refeeding regulate hepatic lipid synthesis. It is established that activation of mitogen-activated protein kinase (MAPK) phosphatase-1 (MKP-1) in response to overnutrition regulates MAPK-dependent pathways that control lipid metabolism in the liver. However, the regulatory mechanisms and the impact of the actions of MKP-1 in hepatic response to fasting remains unclear. We investigated the effect of fasting on the expression of MKP-1 and the impact on hepatic response to feeding. In this study, we demonstrate that fasting stress induced upregulation of hepatic MKP-1 protein levels with a corresponding downregulation of p38 MAPK and JNK phosphorylation in mouse livers. We found that MKP-1-deficient livers are resistant to fasting-induced hepatic steatosis. Hepatic MKP-1 deficiency impaired fasting-induced changes in the levels of key transcription factors involved in the regulation of fatty acid and cholesterol metabolism including Srebf2 and Srebf1c. Mechanistically, MKP-1 negatively regulates Srebf2 expression by attenuating p38 MAPK pathway, suggesting its contribution to the metabolic effects of MKP-1 deficiency in the fasting liver. These findings support the hypothesis that upregulation of MKP-1 is a physiological relevant response and might be beneficial in hepatic lipid utilization during fasting in the liver. Collectively, these data unravel some of the complexity and tissue specific interaction of MKP-1 action in response to changes in nutritional cues, including fasting and excess nutrients
Collapse
Affiliation(s)
- Jacob Sellers
- Department of Biological Sciences, University of Alabama in Huntsville, Huntsville, AL 35899, USA; (J.S.); (A.B.); (S.F.); (G.W.); (S.J.); (S.S.)
| | - Abigail Brooks
- Department of Biological Sciences, University of Alabama in Huntsville, Huntsville, AL 35899, USA; (J.S.); (A.B.); (S.F.); (G.W.); (S.J.); (S.S.)
| | - Savanie Fernando
- Department of Biological Sciences, University of Alabama in Huntsville, Huntsville, AL 35899, USA; (J.S.); (A.B.); (S.F.); (G.W.); (S.J.); (S.S.)
| | - Gabrielle Westenberger
- Department of Biological Sciences, University of Alabama in Huntsville, Huntsville, AL 35899, USA; (J.S.); (A.B.); (S.F.); (G.W.); (S.J.); (S.S.)
| | - Sadie Junkins
- Department of Biological Sciences, University of Alabama in Huntsville, Huntsville, AL 35899, USA; (J.S.); (A.B.); (S.F.); (G.W.); (S.J.); (S.S.)
| | - Shauri Smith
- Department of Biological Sciences, University of Alabama in Huntsville, Huntsville, AL 35899, USA; (J.S.); (A.B.); (S.F.); (G.W.); (S.J.); (S.S.)
| | - Kisuk Min
- Department of Kinesiology, University of Texas at El Paso, El Paso, TX 79968, USA;
| | - Ahmed Lawan
- Department of Biological Sciences, University of Alabama in Huntsville, Huntsville, AL 35899, USA; (J.S.); (A.B.); (S.F.); (G.W.); (S.J.); (S.S.)
- Correspondence: ; Tel.: +1-256-824-6264
| |
Collapse
|
42
|
Hüttl M, Markova I, Miklankova D, Zapletalova I, Poruba M, Haluzik M, Vaněčkova I, Malinska H. In a Prediabetic Model, Empagliflozin Improves Hepatic Lipid Metabolism Independently of Obesity and before Onset of Hyperglycemia. Int J Mol Sci 2021; 22:ijms222111513. [PMID: 34768942 PMCID: PMC8584090 DOI: 10.3390/ijms222111513] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 10/19/2021] [Accepted: 10/21/2021] [Indexed: 12/11/2022] Open
Abstract
Recent studies suggest that treatment with SGLT-2 inhibitors can reduce hepatic lipid storage and ameliorate non-alcoholic fatty liver disease (NAFLD) development beyond their glycemic benefits. However, the exact mechanism involved is still unclear. We investigated the hepatic metabolic effect of empagliflozin (10 mg/kg/day for eight weeks) on the development of NAFLD and its complications using HHTg rats as a non-obese prediabetic rat model. Empagliflozin treatment reduced neutral triacylglycerols and lipotoxic diacylglycerols in the liver and was accompanied by significant changes in relative mRNA expression of lipogenic enzymes (Scd-1, Fas) and transcription factors (Srebp1, Pparγ). In addition, alterations in the gene expression of cytochrome P450 proteins, particularly Cyp2e1 and Cyp4a, together with increased Nrf2, contributed to the improvement of hepatic lipid metabolism after empagliflozin administration. Decreased circulating levels of fetuin-A improved lipid metabolism and attenuated insulin resistance in the liver and in peripheral tissues. Our results highlight the beneficial effect of empagliflozin on hepatic lipid metabolism and lipid accumulation independent of obesity, with the mechanisms understood to involve decreased lipogenesis, alterations in cytochrome P450 proteins, and decreased fetuin-A. These changes help to alleviate NAFLD symptoms in the early phase of the disease and before the onset of diabetes.
Collapse
Affiliation(s)
- Martina Hüttl
- Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, 14321 Prague, Czech Republic; (M.H.); (I.M.); (D.M.)
| | - Irena Markova
- Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, 14321 Prague, Czech Republic; (M.H.); (I.M.); (D.M.)
| | - Denisa Miklankova
- Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, 14321 Prague, Czech Republic; (M.H.); (I.M.); (D.M.)
| | - Iveta Zapletalova
- Department of Pharmacology, Faculty of Medicine and Dentistry, Palacky University Olomouc, 77900 Olomouc, Czech Republic; (I.Z.); (M.P.)
| | - Martin Poruba
- Department of Pharmacology, Faculty of Medicine and Dentistry, Palacky University Olomouc, 77900 Olomouc, Czech Republic; (I.Z.); (M.P.)
| | - Martin Haluzik
- Diabetes Centre, Institute for Clinical and Experimental Medicine, 14321 Prague, Czech Republic;
| | - Ivana Vaněčkova
- Department of Experimental Hypertension, Institute of Physiology, Czech Academy of Sciences, 14220 Prague, Czech Republic;
| | - Hana Malinska
- Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, 14321 Prague, Czech Republic; (M.H.); (I.M.); (D.M.)
- Correspondence: ; Tel.: +420-261-365-369; Fax: +420-261-363-027
| |
Collapse
|
43
|
Jiang L, Hong Y, Xie G, Zhang J, Zhang H, Cai Z. Comprehensive multi-omics approaches reveal the hepatotoxic mechanism of perfluorohexanoic acid (PFHxA) in mice. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 790:148160. [PMID: 34380288 DOI: 10.1016/j.scitotenv.2021.148160] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 05/24/2021] [Accepted: 05/27/2021] [Indexed: 06/13/2023]
Abstract
Perfluorohexanoic acid (PFHxA), one of the short-chain perfluoroalkyl acids (PFAAs), is considered as a substitute of perfluorooctane sulfonate (PFOS). This emerging organic pollutant is persistent and highly bioavailable to humans, raising concerns about its potential health risks. There are currently few researches on the toxicity of PFHxA. Liver has been suggested to be the main target of PFHxA toxicity, and the mechanism remains unclear. Herein, we investigated the transcriptomic, proteomic, and metabolomic landscape in PFHxA-exposed mice. Using these approaches, we identified several valuable biological processes involved in the process of liver injury, comprising fatty acid biosynthesis and degradation pathways, which might be induced by peroxisome proliferator-activated receptor (PPAR) signaling pathway. These processes further promoted oxidative stress and induced liver injury. Meanwhile, abnormalities in purine metabolism and glutathione metabolism were observed during the liver injury induced by PFHxA, indicating the production of oxidative stress. Finally, our present multi-omics studies provided new insights into the mechanisms involved in PFHxA-induced liver injury.
Collapse
Affiliation(s)
- Lilong Jiang
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong, China; Shenzhen Research Institute and Continuing Education, Hong Kong Baptist University, Shenzhen, China
| | - Yanjun Hong
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong, China; Shenzhen Research Institute and Continuing Education, Hong Kong Baptist University, Shenzhen, China; School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, China.
| | - Guangshan Xie
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong, China
| | - Jinghui Zhang
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong, China
| | - Hongna Zhang
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong, China; Shenzhen Research Institute and Continuing Education, Hong Kong Baptist University, Shenzhen, China
| | - Zongwei Cai
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong, China.
| |
Collapse
|
44
|
Smolka C, Schlösser D, Hohnloser C, Bemtgen X, Jänich C, Schneider L, Martin J, Pfeifer D, Moser M, Hasselblatt P, Bode C, Grundmann S, Pankratz F. MiR-100 overexpression attenuates high fat diet induced weight gain, liver steatosis, hypertriglyceridemia and development of metabolic syndrome in mice. Mol Med 2021; 27:101. [PMID: 34488621 PMCID: PMC8422764 DOI: 10.1186/s10020-021-00364-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 08/25/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Diet-induced obesity can result in the development of a diverse spectrum of cardiovascular and metabolic diseases, including type 2 diabetes, dyslipidemia, non-alcoholic liver steatosis and atherosclerotic disease. MicroRNAs have been described to be important regulators of metabolism and disease development. METHODS In the current study, we investigated the effects of ubiquitous miR-100 overexpression on weight gain and the metabolic phenotype in a newly generated transgenic mouse strain under normal chow and high fat diet and used microarray expression analysis to identify new potential target genes of miR-100. RESULTS While transgenic overexpression of miR-100 did not significantly affect weight and metabolism under a normal diet, miR-100 overexpressing mice showed a reduced weight gain under a high fat diet compared to wildtype mice, despite an equal calorie intake. This was accompanied by less visceral and subcutaneous fat development and lover serum LDL cholesterol. In addition, transgenic miR-100 mice were more glucose tolerant and insulin sensitive and demonstrated increased energy expenditure under high fat diet feeding. A comprehensive gene expression profiling revealed the differential expression of several genes involved in lipid storage- and metabolism, among them CD36 and Cyp4A14. Our data showed a direct regulation of CD36 by miR-100, leading to a reduced fatty acid uptake in primary hepatocytes overexpressing miR-100 and the downregulation of several downstream mediators of lipid metabolism such as ACC1, FABP4, FAS and PPARγ in the liver. CONCLUSIONS Our findings demonstrate a protective role of miR-100 in high fat diet induced metabolic syndrome and liver steatosis, partially mediated by the direct repression of CD36 and attenuation of hepatic lipid storage, implicating miR-100 as a possible therapeutic target in liver steatosis.
Collapse
Affiliation(s)
- Christian Smolka
- Department of Cardiology and Angiology I, University Heart Center Freiburg - Bad Krozingen, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Delia Schlösser
- Department of Cardiology and Angiology I, University Heart Center Freiburg - Bad Krozingen, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Catherine Hohnloser
- Department of Cardiology and Angiology I, University Heart Center Freiburg - Bad Krozingen, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Xavier Bemtgen
- Department of Cardiology and Angiology I, University Heart Center Freiburg - Bad Krozingen, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Caterina Jänich
- Department of Cardiology and Angiology I, University Heart Center Freiburg - Bad Krozingen, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Laura Schneider
- Department of Cardiology and Angiology I, University Heart Center Freiburg - Bad Krozingen, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Julien Martin
- Department of Cardiology and Angiology I, University Heart Center Freiburg - Bad Krozingen, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Dietmar Pfeifer
- Department of Hematology, Oncology and Stem Cell Transplantation, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Martin Moser
- Department of Cardiology and Angiology I, University Heart Center Freiburg - Bad Krozingen, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Peter Hasselblatt
- Department of Medicine II, Gastroenterology, Hepatology, Endocrinology, and Infectious Diseases, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christoph Bode
- Department of Cardiology and Angiology I, University Heart Center Freiburg - Bad Krozingen, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sebastian Grundmann
- Department of Cardiology and Angiology I, University Heart Center Freiburg - Bad Krozingen, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Franziska Pankratz
- Department of Cardiology and Angiology I, University Heart Center Freiburg - Bad Krozingen, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
45
|
Yang Z, Smalling RV, Huang Y, Jiang Y, Kusumanchi P, Bogaert W, Wang L, Delker DA, Skill NJ, Han S, Zhang T, Ma J, Huda N, Liangpunsakul S. The role of SHP/REV-ERBα/CYP4A axis in the pathogenesis of alcohol-associated liver disease. JCI Insight 2021; 6:e140687. [PMID: 34423788 PMCID: PMC8410014 DOI: 10.1172/jci.insight.140687] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 07/14/2021] [Indexed: 12/20/2022] Open
Abstract
Alcohol-associated liver disease (ALD) represents a spectrum of histopathological changes, including alcoholic steatosis, steatohepatitis, and cirrhosis. One of the early responses to excessive alcohol consumption is lipid accumulation in the hepatocytes. Lipid ω-hydroxylation of medium- and long-chain fatty acid metabolized by the cytochrome P450 4A (CYP4A) family is an alternative pathway for fatty acid metabolism. The molecular mechanisms of CYP4A in ALD pathogenesis have not been elucidated. In this study, WT and Shp−/− mice were fed with a modified ethanol-binge, National Institute on Alcohol Abuse and Alcoholism model (10 days of ethanol feeding plus single binge). Liver tissues were collected every 6 hours for 24 hours and analyzed using RNA-Seq. The effects of REV-ERBα agonist (SR9009, 100 mg/kg/d) or CYP4A antagonist (HET0016, 5 mg/kg/d) in ethanol-fed mice were also evaluated. We found that hepatic Cyp4a10 and Cyp4a14 expression were significantly upregulated in WT mice, but not in Shp−/− mice, fed with ethanol. ChIP quantitative PCR and promoter assay revealed that REV-ERBα is the transcriptional repressor of Cyp4a10 and Cyp4a14. Rev-Erbα−/− hepatocytes had a marked induction of both Cyp4a genes and lipid accumulation. REV-ERBα agonist SR9009 or CYP4A antagonist HET0016 attenuated Cyp4a induction by ethanol and prevented alcohol-induced steatosis. Here, we have identified a role for the SHP/REV-ERBα/CYP4A axis in the pathogenesis of ALD. Our data also suggest REV-ERBα or CYP4A as the potential therapeutic targets for ALD.
Collapse
Affiliation(s)
- Zhihong Yang
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Rana V Smalling
- Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Yi Huang
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut, USA
| | - Yanchao Jiang
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Praveen Kusumanchi
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Will Bogaert
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut, USA
| | - Li Wang
- Department of Internal Medicine, Section of Digestive Diseases, Yale University, New Haven, Connecticut, USA
| | - Don A Delker
- Divisions of Gastroenterology, University of Utah, Salt Lake City, Utah, USA
| | - Nicholas J Skill
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Sen Han
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Ting Zhang
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Jing Ma
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Nazmul Huda
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Suthat Liangpunsakul
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Roudebush Veterans Administration Medical Center, Indianapolis, Indiana, USA.,Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
46
|
Kozaczek M, Bottje W, Albataineh D, Hakkak R. Effects of Short- and Long-Term Soy Protein Feeding on Hepatic Cytochrome P450 Expression in Obese Nonalcoholic Fatty Liver Disease Rat Model. Front Nutr 2021; 8:699620. [PMID: 34262928 PMCID: PMC8273275 DOI: 10.3389/fnut.2021.699620] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 06/03/2021] [Indexed: 12/16/2022] Open
Abstract
Obesity can lead to chronic health complications such as nonalcoholic fatty liver disease (NAFLD). NAFLD is characterized by lipid aggregation in the hepatocytes and inflammation of the liver tissue as a consequence that can contribute to the development of cirrhosis and hepatocellular carcinoma (HCC). Previously, we reported that feeding obese Zucker rats with soy protein isolate (SPI) can reduce liver steatosis when compared with a casein (CAS) diet as a control. However, the effects of SPI on cytochrome P450 (CYP) in an obese rat model are less known. In addition, there is a lack of information concerning the consumption of soy protein in adolescents and its effect in reducing the early onset of NAFLD in this group. Our main goal was to understand if the SPI diet had any impact on the hepatic CYP gene expression when compared with the CAS diet. For this purpose, we used the transcriptomic data obtained in a previous study in which liver samples were collected from obese rats after short-term (eight-week) and long-term (16-week) feeding of SPI (n = 8 per group). To analyze this RNAseq data, we used Ingenuity Pathway Analysis (IPA) software. Comparing short- vs long-term feeding revealed an increase in the number of downregulated CYP genes from three at 8 weeks of SPI diet to five at 16 weeks of the same diet (P ≤ 0.05). On the other hand, upregulated CYP gene numbers showed a small increase in the long-term SPI diet compared to the short-term SPI diet, from 14 genes at 8 weeks to 17 genes at 16 weeks (P ≤ 0.05). The observed changes may have an important role in the attenuation of liver steatosis.
Collapse
Affiliation(s)
- Melisa Kozaczek
- Department of Dietetics and Nutrition, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Department of Poultry Science and The Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR, United States.,Arkansas Children's Research Institute, Little Rock, AR, United States
| | - Walter Bottje
- Department of Poultry Science and The Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR, United States
| | - Diyana Albataineh
- Department of Poultry Science and The Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR, United States
| | - Reza Hakkak
- Department of Dietetics and Nutrition, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Arkansas Children's Research Institute, Little Rock, AR, United States.,Department of Pediatrics, University of Arkansas for Medical Sciences, Arkansas Children's Hospital, Little Rock, AR, United States
| |
Collapse
|
47
|
Razdan A, Main NM, Chiu V, Shackel NA, de Souza P, Bryant K, Scott KF. Targeting the eicosanoid pathway in hepatocellular carcinoma. Am J Cancer Res 2021; 11:2456-2476. [PMID: 34249410 PMCID: PMC8263695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 04/06/2021] [Indexed: 06/13/2023] Open
Abstract
Liver cancer has variable incidence worldwide and high mortality. Histologically, the most common subtype of liver cancer is hepatocellular carcinoma (HCC). Approximately 30-40% of HCC patients are diagnosed at an advanced stage, and at present, there are limited treatment options for such patients. The current first-line therapy with tyrosine kinase inhibitors, sorafenib or lenvatinib, prolongs survival by a median of about 2.5-3 months after which the disease normally progresses. Additionally, many patients discontinue the use of tyrosine kinase inhibitors due to toxicity or may not be suitable candidates due to co-morbidity or frailty. It is, therefore, imperative to identify novel therapeutic targets for advanced HCC patients. Persistent injury to the liver as a result of insults such as hepatitis B or C viral (HBV or HCV) infections, alcohol abuse, and non-alcoholic fatty liver disease (NAFLD), results in chronic inflammation, which progresses to hepatic fibrosis and later, cirrhosis, provides the conditions for initiation of HCC. One of the key pathways studied for its role in inflammation and carcinogenesis is the eicosanoid pathway. In this review, we briefly outline the eicosanoid pathway, describe the mechanisms by which some pathway members either facilitate or counter the development of liver diseases, with the focus on NAFLD/hepatic fibrosis/cirrhosis, and HCC. We describe the link between the eicosanoid pathway, inflammation and these liver diseases, and identify components of the eicosanoid pathway that may be used as potential therapeutic targets in HCC.
Collapse
Affiliation(s)
- Anshuli Razdan
- School of Medicine, Western Sydney UniversitySydney, NSW, Australia
- Department of Medical Oncology, Ingham Institute for Applied Medical ResearchSydney, NSW, Australia
| | - Nathan M Main
- Gastroenterology and Liver Laboratory, Ingham Institute for Applied Medical ResearchSydney, NSW, Australia
| | - Vincent Chiu
- Gastroenterology and Liver Laboratory, Ingham Institute for Applied Medical ResearchSydney, NSW, Australia
| | - Nicholas A Shackel
- Gastroenterology and Liver Laboratory, Ingham Institute for Applied Medical ResearchSydney, NSW, Australia
| | - Paul de Souza
- School of Medicine, Western Sydney UniversitySydney, NSW, Australia
- Department of Medical Oncology, Ingham Institute for Applied Medical ResearchSydney, NSW, Australia
- School of Medicine, University of WollongongWollongong, NSW, Australia
| | - Katherine Bryant
- Gastroenterology and Liver Laboratory, Ingham Institute for Applied Medical ResearchSydney, NSW, Australia
| | - Kieran F Scott
- School of Medicine, Western Sydney UniversitySydney, NSW, Australia
- Department of Medical Oncology, Ingham Institute for Applied Medical ResearchSydney, NSW, Australia
| |
Collapse
|
48
|
Hamilton MC, Heintz MM, Pfohl M, Marques E, Ford L, Slitt AL, Baldwin WS. Increased toxicity and retention of perflourooctane sulfonate (PFOS) in humanized CYP2B6-Transgenic mice compared to Cyp2b-null mice is relieved by a high-fat diet (HFD). Food Chem Toxicol 2021; 152:112175. [PMID: 33838175 PMCID: PMC8154739 DOI: 10.1016/j.fct.2021.112175] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 03/24/2021] [Accepted: 03/30/2021] [Indexed: 01/11/2023]
Abstract
PFOS is a persistent, fluorosurfactant used in multiple products. Murine Cyp2b's are induced by PFOS and high-fat diets (HFD) and therefore we hypothesized that human CYP2B6 may alleviate PFOS-induced steatosis. Cyp2b-null and hCYP2B6-Tg mice were treated with 0, 1, or 10 mg/kg/day PFOS by oral gavage for 21-days while provided a chow diet (ND) or HFD. Similar to murine Cyp2b10, CYP2B6 is inducible by PFOS. Furthermore, three ND-fed hCYP2B6-Tg females treated with 10 mg/kg/day PFOS died during the exposure period; neither Cyp2b-null nor HFD-fed mice died. hCYP2B6-Tg mice retained more PFOS in serum and liver than Cyp2b-null mice presumably causing the observed toxicity. In contrast, serum PFOS retention was reduced in the HFD-fed hCYP2B6-Tg mice; the opposite trend observed in HFD-fed Cyp2b-null mice. Hepatotoxicity biomarkers, ALT and ALP, were higher in PFOS-treated mice and repressed by a HFD. However, PFOS combined with a HFD exacerbated steatosis in all mice, especially in the hCYP2B6-Tg mice with significant disruption of key lipid metabolism genes such as Srebp1, Pparg, and Hmgcr. In conclusion, CYP2B6 is induced by PFOS but does not alleviate PFOS toxicity presumably due to increased retention. CYP2B6 protects from PFOS-mediated steatosis in ND-fed mice, but increases steatosis when co-treated with a HFD.
Collapse
Affiliation(s)
- Matthew C Hamilton
- Environmental Toxicology Program, Clemson University, Clemson, SC, 29634, USA
| | - Melissa M Heintz
- Environmental Toxicology Program, Clemson University, Clemson, SC, 29634, USA
| | - Marisa Pfohl
- College of Pharmacy, University of Rhode Island, Kingston, RI, 02881, USA
| | - Emily Marques
- College of Pharmacy, University of Rhode Island, Kingston, RI, 02881, USA
| | - Lucie Ford
- College of Pharmacy, University of Rhode Island, Kingston, RI, 02881, USA
| | - Angela L Slitt
- College of Pharmacy, University of Rhode Island, Kingston, RI, 02881, USA
| | - William S Baldwin
- Environmental Toxicology Program, Clemson University, Clemson, SC, 29634, USA.
| |
Collapse
|
49
|
Li S, Wang C, Zhang X, Su W. Cytochrome P450 Omega-Hydroxylase 4a14 Attenuates Cholestatic Liver Fibrosis. Front Physiol 2021; 12:688259. [PMID: 34135776 PMCID: PMC8201794 DOI: 10.3389/fphys.2021.688259] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 04/22/2021] [Indexed: 12/12/2022] Open
Abstract
Background Cholestasis is a pathological condition involving obstruction of bile secretion and excretion that results in hepatotoxicity, inflammation, fibrosis, cirrhosis, and eventually liver failure. Common bile duct ligation (BDL) model is a well-established murine model to mimic cholestatic liver fibrosis. We previously reported that cytochrome P450 omega-hydroxylase 4a14 (Cyp4a14) plays an important role in the pathogenesis of non-alcoholic fatty liver disease (NAFLD)-related fibrosis. The goal of this study was to determine the role of Cyp4a14 in cholestatic-induced liver fibrosis. Methods C57BL/6 mice were subjected to BDL for 14 days, and Cyp4a14 mRNA and protein levels were examined and compared with those of the sham group. Cyp4a14 knockout mice and adeno-associated virus (AAV)-mediated overexpression of Cyp4a14 in C57BL/6 mice underwent BDL and liver histology, and key fibrosis markers were examined. Results Both hepatic Cyp4a14 mRNA and protein levels were markedly reduced in BDL liver compared with the time-matched sham group. Cyp4a14 gene-deficient mice aggravates whereas its overexpression alleviates BDL-induced hepatic fibrosis, which were determined by liver function, liver histology, and levels of key fibrotic markers including α-smooth muscle actin (α-SMA), transforming growth factor-β1 (TGF-β1), and collagen 1a2 (Col1a2). Conclusion Cyp4a14 exerts a contrasting role in different hepatic fibrosis models. Strategies that enhance Cyp4a14 activity may be potential strategies to cholestatic related liver fibrosis.
Collapse
Affiliation(s)
- Sha Li
- Medical College, Hebei University of Engineering, Handan, China.,Hebei Key Laboratory of Applied Basic Research of Blood Purification, Affiliated Hospital of Hebei Engineering University, Handan, China.,Health Science Center, Shenzhen University, Shenzhen, China
| | - Chenghai Wang
- Hebei Key Laboratory of Applied Basic Research of Blood Purification, Affiliated Hospital of Hebei Engineering University, Handan, China
| | - Xiaxia Zhang
- Department of Gastroenterology and Hepatology, Handan Central Hospital, Handan, China
| | - Wen Su
- Health Science Center, Shenzhen University, Shenzhen, China.,Shenzhen Key Laboratory of Metabolism and Cardiovascular Homeostasis, Shenzhen, China
| |
Collapse
|
50
|
Malinská H, Hüttl M, Miklánková D, Trnovská J, Zapletalová I, Poruba M, Marková I. Ovariectomy-Induced Hepatic Lipid and Cytochrome P450 Dysmetabolism Precedes Serum Dyslipidemia. Int J Mol Sci 2021; 22:ijms22094527. [PMID: 33926097 PMCID: PMC8123580 DOI: 10.3390/ijms22094527] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/23/2021] [Accepted: 04/23/2021] [Indexed: 12/13/2022] Open
Abstract
Ovarian hormone deficiency leads to increased body weight, visceral adiposity, fatty liver and disorders associated with menopausal metabolic syndrome. To better understand the underlying mechanisms of these disorders in their early phases of development, we investigated the effect of ovariectomy on lipid and glucose metabolism. Compared to sham-operated controls, ovariectomized Wistar female rats markedly increased whole body and visceral adipose tissue weight (p ˂ 0.05) and exhibited insulin resistance in peripheral tissues. Severe hepatic triglyceride accumulation (p ˂ 0.001) after ovariectomy preceded changes in both serum lipids and glucose intolerance, reflecting alterations in some CYP proteins. Increased CYP2E1 (p ˂ 0.05) and decreased CYP4A (p ˂ 0.001) after ovariectomy reduced fatty acid oxidation and induced hepatic steatosis. Decreased triglyceride metabolism and secretion from the liver contributed to hepatic triglyceride accumulation in response to ovariectomy. In addition, interscapular brown adipose tissue of ovariectomized rats exhibited decreased fatty acid oxidation (p ˂ 0.01), lipogenesis (p ˂ 0.05) and lipolysis (p ˂ 0.05) despite an increase in tissue weight. The results provide evidence that impaired hepatic triglycerides and dysregulation of some CYP450 proteins may have been involved in the development of hepatic steatosis. The low metabolic activity of brown adipose tissue may have contributed to visceral adiposity as well as triglyceride accumulation during the postmenopausal period.
Collapse
Affiliation(s)
- Hana Malinská
- Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, 14021 Prague, Czech Republic; (M.H.); (D.M.); (J.T.); (I.M.)
- Correspondence: ; Tel.: +420-261-365-369; Fax: +420-261-363-027
| | - Martina Hüttl
- Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, 14021 Prague, Czech Republic; (M.H.); (D.M.); (J.T.); (I.M.)
| | - Denisa Miklánková
- Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, 14021 Prague, Czech Republic; (M.H.); (D.M.); (J.T.); (I.M.)
| | - Jaroslava Trnovská
- Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, 14021 Prague, Czech Republic; (M.H.); (D.M.); (J.T.); (I.M.)
| | - Iveta Zapletalová
- Department of Pharmacology, Faculty of Medicine and Dentistry, Palacky University, 77900 Olomouc, Czech Republic; (I.Z.); (M.P.)
| | - Martin Poruba
- Department of Pharmacology, Faculty of Medicine and Dentistry, Palacky University, 77900 Olomouc, Czech Republic; (I.Z.); (M.P.)
| | - Irena Marková
- Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, 14021 Prague, Czech Republic; (M.H.); (D.M.); (J.T.); (I.M.)
| |
Collapse
|