1
|
Kim DH, Choi G, Song EB, Lee H, Kim J, Jang YS, Park J, Chi S, Han J, Kim SM, Kim D, Bae SH, Lee HW, Park JY, Kang SG, Cha SH, Han YH. Treatment of IL-18-binding protein biologics suppresses fibrotic progression in metabolic dysfunction-associated steatohepatitis. Cell Rep Med 2025; 6:102047. [PMID: 40239621 PMCID: PMC12047490 DOI: 10.1016/j.xcrm.2025.102047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 10/18/2024] [Accepted: 03/07/2025] [Indexed: 04/18/2025]
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) is a chronic liver disease characterized by inflammation and fibrosis, with enhanced interleukin-18 (IL-18) signaling. IL-18-binding protein (IL-18BP) neutralizes IL-18, but its therapeutic potential in MASH is unclear. We find elevated IL-18BP and IL-18 levels in patients with MASH and mice, with free IL-18 correlating with disease severity. IL-18 stimulates interferon-gamma (IFNγ) production in CD4 T cells, increasing hepatic IL-18BP. IL-18BP-deficient mice show worsened liver inflammation and fibrosis. We develop a human IL-18BP biologics (APB-R3) and inject it to mice to evaluate its pharmacologic efficacy. APB-R3 significantly improves MASH in reducing fibrosis and inflammation and inhibits hepatic stellate cell activation via the cGMP pathway. This study proposes that abrogation of IL-18 signaling by boosting IL-18BP can strongly inhibit the development of MASH-induced fibrosis, and our engineered IL-18BP biologics can become a promising therapeutic candidate for curing MASH.
Collapse
Affiliation(s)
- Dong-Hyun Kim
- Laboratory of Pathology and Physiology, College of Pharmacy, Kangwon National University, Chuncheon 24341, South Korea
| | - Gona Choi
- Laboratory of Pathology and Physiology, College of Pharmacy, Kangwon National University, Chuncheon 24341, South Korea
| | - Eun-Bi Song
- Laboratory of Pathology and Physiology, College of Pharmacy, Kangwon National University, Chuncheon 24341, South Korea
| | - Hanna Lee
- Laboratory of Pathology and Physiology, College of Pharmacy, Kangwon National University, Chuncheon 24341, South Korea; Multidimensional Genomics Research Center, Kangwon National University, Chuncheon 24341, South Korea
| | - Jaehui Kim
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, Chuncheon 24341, South Korea; Institute of Bioscience and Biotechnology, College of Biomedical Science, Kangwon National University, Chuncheon 24341, South Korea
| | - Young-Saeng Jang
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, Chuncheon 24341, South Korea; Institute of Bioscience and Biotechnology, College of Biomedical Science, Kangwon National University, Chuncheon 24341, South Korea
| | - JinJoo Park
- AprilBio Co., Ltd, Biomedical Science Building, Kangwon National University, Chuncheon 24341, South Korea
| | - Susan Chi
- AprilBio Co., Ltd, Biomedical Science Building, Kangwon National University, Chuncheon 24341, South Korea
| | - Jaekyu Han
- AprilBio Co., Ltd, Biomedical Science Building, Kangwon National University, Chuncheon 24341, South Korea
| | - Sun-Mi Kim
- AprilBio Co., Ltd, Biomedical Science Building, Kangwon National University, Chuncheon 24341, South Korea
| | - Dongyoon Kim
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Soo Han Bae
- Severance Biomedical Science Institute, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, South Korea
| | - Hye Won Lee
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, South Korea
| | - Jun Yong Park
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, South Korea
| | - Seung Goo Kang
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, Chuncheon 24341, South Korea; Institute of Bioscience and Biotechnology, College of Biomedical Science, Kangwon National University, Chuncheon 24341, South Korea.
| | - Sang-Hoon Cha
- Institute of Bioscience and Biotechnology, College of Biomedical Science, Kangwon National University, Chuncheon 24341, South Korea; AprilBio Co., Ltd, Biomedical Science Building, Kangwon National University, Chuncheon 24341, South Korea.
| | - Yong-Hyun Han
- Laboratory of Pathology and Physiology, College of Pharmacy, Kangwon National University, Chuncheon 24341, South Korea; Multidimensional Genomics Research Center, Kangwon National University, Chuncheon 24341, South Korea.
| |
Collapse
|
2
|
Brothwell MJ, Cao G, Maschek JA, Poss AM, Peterlin AD, Wang L, Baker TB, Shahtout JL, Siripoksup P, Pearce QJ, Johnson JM, Finger FM, Prola A, Pellizzari SA, Hale GL, Manuel AM, Watanabe S, Miranda ER, Affolter KE, Tippetts TS, Nikolova LS, Choi RH, Decker ST, Patil M, Catrow JL, Holland WL, Nowinski SM, Lark DS, Fisher-Wellman KH, Mimche PN, Evason KJ, Cox JE, Summers SA, Gerhart-Hines Z, Funai K. Cardiolipin deficiency disrupts electron transport chain to drive steatohepatitis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.10.10.617517. [PMID: 39416056 PMCID: PMC11482932 DOI: 10.1101/2024.10.10.617517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a progressive disorder marked by lipid accumulation, leading to metabolic dysfunction-associated steatohepatitis (MASH). A key feature of the transition to MASH involves oxidative stress resulting from defects in mitochondrial oxidative phosphorylation (OXPHOS). Here, we show that pathological alterations in the lipid composition of the inner mitochondrial membrane (IMM) directly instigate electron transfer inefficiency to promote oxidative stress. Specifically, mitochondrial cardiolipin (CL) was downregulated with MASLD/MASH in humans and in mice. Hepatocyte-specific CL synthase knockout (CLS-LKO) led to spontaneous and robust MASH with extensive steatotic and fibrotic phenotype. Loss of CL paradoxically increased mitochondrial respiratory capacity but also reduced the formation of I+III2+IV respiratory supercomplex, promoted electron leak primarily at sites IIIQO and IIF of the electron transport chain, and disrupted the propensity of coenzyme Q (CoQ) to become reduced. Thus, low mitochondrial CL disrupts electron transport chain to promote oxidative stress and contributes to pathogenesis of MASH.
Collapse
Affiliation(s)
- Marisa J. Brothwell
- Diabetes & Metabolism Research Center; University of Utah; Salt Lake City, UT; USA
- Department of Nutrition and Integrative Physiology; University of Utah; Salt Lake City, UT; USA
| | - Guoshen Cao
- Diabetes & Metabolism Research Center; University of Utah; Salt Lake City, UT; USA
- Department of Biochemistry; University of Utah; Salt Lake City, UT; USA
| | - J. Alan Maschek
- Diabetes & Metabolism Research Center; University of Utah; Salt Lake City, UT; USA
- Department of Nutrition and Integrative Physiology; University of Utah; Salt Lake City, UT; USA
- Metabolomics Core Research Facility; University of Utah; Salt Lake City, UT; USA
| | - Annelise M. Poss
- Diabetes & Metabolism Research Center; University of Utah; Salt Lake City, UT; USA
- Department of Nutrition and Integrative Physiology; University of Utah; Salt Lake City, UT; USA
| | - Alek D. Peterlin
- Diabetes & Metabolism Research Center; University of Utah; Salt Lake City, UT; USA
- Department of Nutrition and Integrative Physiology; University of Utah; Salt Lake City, UT; USA
| | - Liping Wang
- Diabetes & Metabolism Research Center; University of Utah; Salt Lake City, UT; USA
- Department of Nutrition and Integrative Physiology; University of Utah; Salt Lake City, UT; USA
| | - Talia B. Baker
- Huntsman Cancer Institute; University of Utah, Salt Lake City, UT; USA
- Division of Transplantation and Advanced Hepatobiliary Surgery, Department of Surgery; University of Utah; Salt Lake City, UT; USA
| | - Justin L. Shahtout
- Diabetes & Metabolism Research Center; University of Utah; Salt Lake City, UT; USA
- Department of Physical Therapy and Athletic Training; University of Utah; Salt Lake City, UT; USA
| | - Piyarat Siripoksup
- Diabetes & Metabolism Research Center; University of Utah; Salt Lake City, UT; USA
- Department of Physical Therapy and Athletic Training; University of Utah; Salt Lake City, UT; USA
| | - Quentinn J. Pearce
- Metabolomics Core Research Facility; University of Utah; Salt Lake City, UT; USA
| | - Jordan M. Johnson
- Diabetes & Metabolism Research Center; University of Utah; Salt Lake City, UT; USA
- Department of Nutrition and Integrative Physiology; University of Utah; Salt Lake City, UT; USA
| | - Fabian M. Finger
- Novo Nordisk Foundation Center for Basic Metabolic Research; University of Copenhagen; Copenhagen; DK
- Center for Adipocyte Signaling (ADIPOSIGN); University of Southern Denmark; Odense; DK
| | - Alexandre Prola
- Laboratory of Fundamental and Applied Bioenergetics; University of Grenoble Alpes, Inserm U1055; Grenoble; FR
| | - Sarah A. Pellizzari
- Department of Biochemistry; University of Utah; Salt Lake City, UT; USA
- Department of Pathology; University of Utah; Salt Lake City, UT; USA
| | - Gillian L. Hale
- Huntsman Cancer Institute; University of Utah, Salt Lake City, UT; USA
- Department of Pathology; University of Utah; Salt Lake City, UT; USA
| | - Allison M. Manuel
- Metabolomics Core Research Facility; University of Utah; Salt Lake City, UT; USA
| | - Shinya Watanabe
- Diabetes & Metabolism Research Center; University of Utah; Salt Lake City, UT; USA
- Department of Nutrition and Integrative Physiology; University of Utah; Salt Lake City, UT; USA
| | - Edwin R. Miranda
- Diabetes & Metabolism Research Center; University of Utah; Salt Lake City, UT; USA
- Department of Nutrition and Integrative Physiology; University of Utah; Salt Lake City, UT; USA
- Molecular Medicine Program; University of Utah; Salt Lake City, UT; USA
| | - Kajsa E. Affolter
- Huntsman Cancer Institute; University of Utah, Salt Lake City, UT; USA
- Laboratory of Fundamental and Applied Bioenergetics; University of Grenoble Alpes, Inserm U1055; Grenoble; FR
| | - Trevor S. Tippetts
- Diabetes & Metabolism Research Center; University of Utah; Salt Lake City, UT; USA
- Department of Nutrition and Integrative Physiology; University of Utah; Salt Lake City, UT; USA
| | - Linda S. Nikolova
- Electron Microscopy Core Facility; University of Utah; Salt Lake City, UT; USA
| | - Ran Hee Choi
- Diabetes & Metabolism Research Center; University of Utah; Salt Lake City, UT; USA
- Department of Nutrition and Integrative Physiology; University of Utah; Salt Lake City, UT; USA
- Molecular Medicine Program; University of Utah; Salt Lake City, UT; USA
| | - Stephen T. Decker
- Diabetes & Metabolism Research Center; University of Utah; Salt Lake City, UT; USA
- Department of Nutrition and Integrative Physiology; University of Utah; Salt Lake City, UT; USA
- Molecular Medicine Program; University of Utah; Salt Lake City, UT; USA
| | - Mallikarjun Patil
- Diabetes & Metabolism Research Center; University of Utah; Salt Lake City, UT; USA
- Department of Nutrition and Integrative Physiology; University of Utah; Salt Lake City, UT; USA
- Molecular Medicine Program; University of Utah; Salt Lake City, UT; USA
| | - J. Leon Catrow
- Metabolomics Core Research Facility; University of Utah; Salt Lake City, UT; USA
| | - William L. Holland
- Diabetes & Metabolism Research Center; University of Utah; Salt Lake City, UT; USA
- Department of Nutrition and Integrative Physiology; University of Utah; Salt Lake City, UT; USA
- Department of Biochemistry; University of Utah; Salt Lake City, UT; USA
- Molecular Medicine Program; University of Utah; Salt Lake City, UT; USA
| | - Sara M. Nowinski
- Department of Metabolism and Nutritional Programming; Van Andel Institute; Grand Rapids, MI; USA
| | - Daniel S. Lark
- College of Health and Human Sciences; Colorado State University; Fort Collins, CO; USA
- Columbine Health Systems Center for Healthy Aging; Colorado State University; Fort Collins, CO; USA
| | - Kelsey H. Fisher-Wellman
- Department of Cancer Biology, Wake Forest University School of Medicine; Atrium Health Wake Forest Baptist Comprehensive Cancer Center; Winston-Salem, NC; USA
| | - Patrice N. Mimche
- Departments of Dermatology and Medicine; Division of Gastroenterology and Hepatology, Indiana University School of Medicine; Indianapolis, IN; USA
| | - Kimberley J. Evason
- Huntsman Cancer Institute; University of Utah, Salt Lake City, UT; USA
- Department of Pathology; University of Utah; Salt Lake City, UT; USA
| | - James E. Cox
- Diabetes & Metabolism Research Center; University of Utah; Salt Lake City, UT; USA
- Department of Biochemistry; University of Utah; Salt Lake City, UT; USA
- Metabolomics Core Research Facility; University of Utah; Salt Lake City, UT; USA
| | - Scott A. Summers
- Diabetes & Metabolism Research Center; University of Utah; Salt Lake City, UT; USA
- Department of Nutrition and Integrative Physiology; University of Utah; Salt Lake City, UT; USA
- Department of Biochemistry; University of Utah; Salt Lake City, UT; USA
- Huntsman Cancer Institute; University of Utah, Salt Lake City, UT; USA
- Molecular Medicine Program; University of Utah; Salt Lake City, UT; USA
| | - Zach Gerhart-Hines
- Novo Nordisk Foundation Center for Basic Metabolic Research; University of Copenhagen; Copenhagen; DK
- Center for Adipocyte Signaling (ADIPOSIGN); University of Southern Denmark; Odense; DK
| | - Katsuhiko Funai
- Diabetes & Metabolism Research Center; University of Utah; Salt Lake City, UT; USA
- Department of Nutrition and Integrative Physiology; University of Utah; Salt Lake City, UT; USA
- Department of Biochemistry; University of Utah; Salt Lake City, UT; USA
- Huntsman Cancer Institute; University of Utah, Salt Lake City, UT; USA
- Department of Physical Therapy and Athletic Training; University of Utah; Salt Lake City, UT; USA
- Molecular Medicine Program; University of Utah; Salt Lake City, UT; USA
| |
Collapse
|
3
|
Kang P, Chen Q, Wu J, Zhang Q, Crump D, Su G. Novel Organophosphate Ester Tris(2,4-di- tert-butylphenyl)phosphate Alters Lipid Metabolism: Insights from Lipidomic Analysis and mRNA Expression. Chem Res Toxicol 2025; 38:448-457. [PMID: 39928661 DOI: 10.1021/acs.chemrestox.4c00460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2025]
Abstract
Tris(2,4-di-tert-butylphenyl)phosphate (TDTBPP), a novel organophosphate ester (OPE), has been extensively detected in various environmental and biological samples; however, its potential biological effects remain unexplored. In this study, we investigated biotransformation characteristics, alteration of lipid metabolism, and mRNA expression in primary mouse hepatocytes (PMHs) following exposure to TDTBPP. After 36-h exposure in PMHs, TDTBPP exhibited a high stability potential with no statistically significant degradation trend. Subsequently, we analyzed the disruption of lipid homeostasis in PMHs following exposure to 0-4.5 μM TDTBPP. Lipidomic analysis indicated that TDTBPP disrupted lipid homeostasis in PMHs, and several lipid classes were dysregulated, in particular, glycerolipids and glycerophospholipids. Additionally, three lipids were proposed as potential lipid biomarkers of TDTBPP exposure, including triglycerides (TGs) and phosphatidylcholines (PCs). These observations were further supported by transcriptional changes, with significant alteration observed in genes associated with lipid uptake, de novo lipogenesis, β-oxidation of fatty acids, glycerolipid metabolism, and lipid export. Overall, these findings highlight the detrimental effects of TDTBPP on lipid homeostasis, providing important insights for health risk assessments of this abundant OPE in the environment.
Collapse
Affiliation(s)
- Pingping Kang
- Key Laboratory of Environmental Remediation and Ecological Health, Ministry of Industry and Information Technology, School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing 210094, China
| | - Qianyu Chen
- Key Laboratory of Environmental Remediation and Ecological Health, Ministry of Industry and Information Technology, School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing 210094, China
| | - Jia Wu
- Key Laboratory of Environmental Remediation and Ecological Health, Ministry of Industry and Information Technology, School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing 210094, China
| | - Qi Zhang
- Key Laboratory of Environmental Remediation and Ecological Health, Ministry of Industry and Information Technology, School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing 210094, China
| | - Doug Crump
- National Wildlife Research Centre, Environment and Climate Change Canada, Ottawa, K1A0H3, Canada
| | - Guanyong Su
- Key Laboratory of Environmental Remediation and Ecological Health, Ministry of Industry and Information Technology, School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing 210094, China
| |
Collapse
|
4
|
Wang H, Ciccocioppo R, Terai S, Shoeibi S, Carnevale G, De Marchi G, Tsuchiya A, Ishii S, Tonouchi T, Furuyama K, Yang Y, Mito M, Abe H, Di Tinco R, Cardinale V. Targeted animal models for preclinical assessment of cellular and gene therapies in pancreatic and liver diseases: regulatory and practical insights. Cytotherapy 2025; 27:259-278. [PMID: 39755978 PMCID: PMC12068232 DOI: 10.1016/j.jcyt.2024.11.008] [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: 05/31/2024] [Revised: 11/08/2024] [Accepted: 11/10/2024] [Indexed: 01/07/2025]
Abstract
Cellular and gene therapy (CGT) products have emerged as a popular approach in regenerative medicine, showing promise in treating various pancreatic and liver diseases in numerous clinical trials. Before these therapies can be tested in human clinical trials, it is essential to evaluate their safety and efficacy in relevant animal models. Such preclinical testing is often required to obtain regulatory approval for investigational new drugs. However, there is a lack of detailed guidance on selecting appropriate animal models for CGT therapies targeting specific pancreatic and liver conditions, such as pancreatitis and chronic liver diseases. In this review, the gastrointestinal committee for the International Society for Cell and Gene Therapy provides a summary of current recommendations for animal species and disease model selection, as outlined by the US Food and Drug Administration, with references to EU EMA and Japan PMDA. We discuss a range of small and large animal models, as well as humanized models, that are suitable for preclinical testing of CGT products aimed at treating pancreatic and liver diseases. For each model, we cover the associated pathophysiology, commonly used metrics for assessing disease status, the pros and limitations of the models, and the relevance of these models to human conditions. We also summarize the use and application of humanized mouse and other animal models in evaluating the safety and efficacy of CGT products. This review aims to provide comprehensive guidance for selecting appropriate animal species and models to help bridge the gap between the preclinical research and clinical trials using CGT therapies for specific pancreatic and liver diseases.
Collapse
Affiliation(s)
- Hongjun Wang
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina, USA; Ralph H Johnson Veteran Medical Center, Charleston, South Carolina, USA.
| | - Rachele Ciccocioppo
- Department of Medicine, Gastroenterology Unit, Pancreas Institute, A.O.U.I. Policlinico G.B. Rossi & University of Verona, Verona, Italy
| | - Shuji Terai
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Sara Shoeibi
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Gianluca Carnevale
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Giulia De Marchi
- Department of Medicine, Gastroenterology Unit, Pancreas Institute, A.O.U.I. Policlinico G.B. Rossi & University of Verona, Verona, Italy
| | - Atsunori Tsuchiya
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Soichi Ishii
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Takafumi Tonouchi
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Kaito Furuyama
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Yuan Yang
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Masaki Mito
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Hiroyuki Abe
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Rosanna Di Tinco
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Vincenzo Cardinale
- Department of Translational and Precision Medicine, University of Rome, Rome, Italy.
| |
Collapse
|
5
|
Karin M, Kim JY. MASH as an emerging cause of hepatocellular carcinoma: current knowledge and future perspectives. Mol Oncol 2025; 19:275-294. [PMID: 38874196 PMCID: PMC11793012 DOI: 10.1002/1878-0261.13685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 04/15/2024] [Accepted: 06/04/2024] [Indexed: 06/15/2024] Open
Abstract
Hepatocellular carcinoma is one of the deadliest and fastest-growing cancers. Among HCC etiologies, metabolic dysfunction-associated fatty liver disease (MAFLD) has served as a major HCC driver due to its great potential for increasing cirrhosis. The obesogenic environment fosters a positive energy balance and results in a continuous rise of obesity and metabolic syndrome. However, it is difficult to understand how metabolic complications lead to the poor prognosis of liver diseases and which molecular mechanisms are underpinning MAFLD-driven HCC development. Thus, suitable preclinical models that recapitulate human etiologies are essentially required. Numerous preclinical models have been created but not many mimicked anthropometric measures and the course of disease progression shown in the patients. Here we review the literature on adipose tissues, liver-related HCC etiologies and recently discovered genetic mutation signatures found in MAFLD-driven HCC patients. We also critically review current rodent models suggested for MAFLD-driven HCC study.
Collapse
Affiliation(s)
- Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of MedicineUniversity of California San DiegoLa JollaCAUSA
| | - Ju Youn Kim
- Department of Molecular and Life ScienceHanyang University ERICAAnsanKorea
| |
Collapse
|
6
|
Zhong L, Yang Q, Shao Y, Hu S, Guo L. Helicobacter pylori promotes intestinal flora imbalance and hepatic metabolic disorders under arsenic stress. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 290:117512. [PMID: 39671763 DOI: 10.1016/j.ecoenv.2024.117512] [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: 05/16/2024] [Revised: 10/21/2024] [Accepted: 12/08/2024] [Indexed: 12/15/2024]
Abstract
Environmental arsenic contamination is a serious issue that cannot be ignored, since arsenic levels in drinking water frequently exceed safety standards, and there is an increased prevalence of Helicobacter pylori (H. pylori) infection. This results in an increasing population at risk of simultaneous exposure to both harmful agents, yet whether a synergistic interaction exists between them remains unclear. Therefore, this study aims to investigate the combined effects and underlying pathogenic mechanisms of concurrent exposure to these two hazardous factors by establishing a mouse model that is infected with H. pylori and exposed to inorganic arsenic through drinking water. Analysis of intestinal flora revealed significant alterations in the composition, relative abundance (Akkermansia, Faecalibaculum, Ilieibacterium, etc.), and metabolic potential of the intestinal microflora (amino acid metabolism and energy metabolism) in the combinatory exposure group. Non-targeted metabolomics analysis identified that the combinatory exposure group exhibited greater fluctuations in metabolite content, particularly in triacylglycerol, fatty-acid, peptide and amino acid. Moreover, H. pylori infection and arsenic exposure had increased levels of metabolites associated with the intestinal microbiota in their livers (4-Ethylphenyl sulfate and Phenylacetylglycine). Further analysis revealed significant correlations between changes in the intestinal flora and alterations in liver metabolic profiles. Herein, we hypothesize that H. pylori infection may exacerbate the intestinal flora imbalance and hepatic metabolic disturbances caused by arsenic exposure, which may disrupt enterohepatic homeostasis and potentially increase biological susceptibility to heavy metal toxicity.
Collapse
Affiliation(s)
- Linmin Zhong
- Dongguan Key Laboratory of Public Health Laboratory Science, The First Dongguan Affiliated Hospital, School of Public Health, Guangdong Medical University, Dongguan 523808, China
| | - Qiling Yang
- Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Yiming Shao
- Dongguan Key Laboratory of Sepsis Translational Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China
| | - Shanwen Hu
- Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Lianxian Guo
- Dongguan Key Laboratory of Public Health Laboratory Science, The First Dongguan Affiliated Hospital, School of Public Health, Guangdong Medical University, Dongguan 523808, China.
| |
Collapse
|
7
|
Gopinath V, Mariya Davis A, Menon TK, Raghavamenon AC. Alcohol promotes liver fibrosis in high fat diet induced diabetic rats. J Basic Clin Physiol Pharmacol 2024; 35:273-284. [PMID: 39023980 DOI: 10.1515/jbcpp-2024-0042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 06/18/2024] [Indexed: 07/20/2024]
Abstract
OBJECTIVES Type 2 diabetes (T2DM) and alcoholism are considered to be lifestyle-associated independent risk factors in fatty liver diseases (FLD) mediated cirrhosis and hepatocellular carcinoma (HCC). A combined effect of both these conditions may exacerbate the pathological changes and a pre-clinical exploration of this is expected to provide a mechanical detail of the pathophysiology. The present study aims to understand the effect of alcohol on pre- diabetic and type 2 diabetic female Wistar rats. METHODS In this experimental study, 12 Wistar rats (180-220 g) were randomly assigned into three groups: Normal (fed normal rat chow), alcohol (20 %) fed diabetic (HFD + STZ), and pre-diabetic rats (HFD alone). After, two months of the experimental period, blood and liver tissues were collected lipid metabolic alteration, liver injury, and fibrosis were determined following biochemical and histological methods. Data were analyzed using one-way ANOVA and Dunnett's Post Hoc test. RESULTS Significant dyslipidemia was observed in the liver tissues of diabetic and pre-diabetic rats following alcohol ingestion. A significant (p<0.05) increase in lipid peroxidation status, and hepatic marker enzyme activities (p<0.0001) were observed in diabetic animals. In corroborating with these observations, hematoxylin and eosin staining of hepatic tissue revealed the presence of sinusoidal dilation along with heavily damaged hepatocytes and inflammatory cell infiltration. Further, significantly (p<0.001) increased hepatic hydroxyproline content and extended picrosirius red stained areas of collagen in liver tissue indicated initiation of fibrosis in alcohol-fed diabetic rats. CONCLUSIONS Overall, the results indicate that alcohol consumption in T2DM conditions is more deleterious than pre diabetic conditions in progressing to hepatic fibrosis.
Collapse
Affiliation(s)
- Veena Gopinath
- Department of Biochemistry, Amala Cancer Research Center (Recognized Centre of the University of Calicut), Thrissur, Kerala, India
| | - Aleena Mariya Davis
- Department of Biochemistry, Amala Cancer Research Center (Recognized Centre of the University of Calicut), Thrissur, Kerala, India
| | - Thara K Menon
- Department of Biotechnology, University of Calicut, Thenhipalam, Kerala, India
| | - Achuthan C Raghavamenon
- Department of Biochemistry, Amala Cancer Research Center (Recognized Centre of the University of Calicut), Thrissur, Kerala, India
| |
Collapse
|
8
|
Vacca M, Kamzolas I, Harder LM, Oakley F, Trautwein C, Hatting M, Ross T, Bernardo B, Oldenburger A, Hjuler ST, Ksiazek I, Lindén D, Schuppan D, Rodriguez-Cuenca S, Tonini MM, Castañeda TR, Kannt A, Rodrigues CMP, Cockell S, Govaere O, Daly AK, Allison M, Honnens de Lichtenberg K, Kim YO, Lindblom A, Oldham S, Andréasson AC, Schlerman F, Marioneaux J, Sanyal A, Afonso MB, Younes R, Amano Y, Friedman SL, Wang S, Bhattacharya D, Simon E, Paradis V, Burt A, Grypari IM, Davies S, Driessen A, Yashiro H, Pors S, Worm Andersen M, Feigh M, Yunis C, Bedossa P, Stewart M, Cater HL, Wells S, Schattenberg JM, Anstee QM, Tiniakos D, Perfield JW, Petsalaki E, Davidsen P, Vidal-Puig A. An unbiased ranking of murine dietary models based on their proximity to human metabolic dysfunction-associated steatotic liver disease (MASLD). Nat Metab 2024; 6:1178-1196. [PMID: 38867022 PMCID: PMC11199145 DOI: 10.1038/s42255-024-01043-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 04/08/2024] [Indexed: 06/14/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), previously known as non-alcoholic fatty liver disease, encompasses steatosis and metabolic dysfunction-associated steatohepatitis (MASH), leading to cirrhosis and hepatocellular carcinoma. Preclinical MASLD research is mainly performed in rodents; however, the model that best recapitulates human disease is yet to be defined. We conducted a wide-ranging retrospective review (metabolic phenotype, liver histopathology, transcriptome benchmarked against humans) of murine models (mostly male) and ranked them using an unbiased MASLD 'human proximity score' to define their metabolic relevance and ability to induce MASH-fibrosis. Here, we show that Western diets align closely with human MASH; high cholesterol content, extended study duration and/or genetic manipulation of disease-promoting pathways are required to intensify liver damage and accelerate significant (F2+) fibrosis development. Choline-deficient models rapidly induce MASH-fibrosis while showing relatively poor translatability. Our ranking of commonly used MASLD models, based on their proximity to human MASLD, helps with the selection of appropriate in vivo models to accelerate preclinical research.
Collapse
Affiliation(s)
- Michele Vacca
- TVP Lab, WT/MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
- Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Bari, Italy.
- Laboratory of Liver Metabolism and MASLD, Roger Williams Institute of Hepatology, London, UK.
| | - Ioannis Kamzolas
- TVP Lab, WT/MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Lea Mørch Harder
- Research and Early Development, Novo Nordisk A/S, Måløv, Copenhagen, Denmark
| | - Fiona Oakley
- Newcastle Fibrosis Research Group, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Christian Trautwein
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Maximilian Hatting
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Trenton Ross
- Internal Medicine research Research Unit, Pfizer Worldwide Research and Development, Cambridge, MA, USA
| | - Barbara Bernardo
- Internal Medicine research Research Unit, Pfizer Worldwide Research and Development, Cambridge, MA, USA
| | - Anouk Oldenburger
- CardioMetabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| | | | - Iwona Ksiazek
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Daniel Lindén
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism (CVRM), AstraZeneca BioPharmaceuticals R&D, Gothenburg, Sweden
- Division of Endocrinology, Department of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Detlef Schuppan
- Institute of Translational Immunology and Research Center for Immunotherapy, Johannes Gutenberg University Medical Center, Mainz, Germany
| | | | - Maria Manuela Tonini
- Luxembourg Institute of Health, Translational Medicine Operations Hub, Dudelange, Luxembourg
| | - Tamara R Castañeda
- R&D Diabetes & Portfolio Innovation and Excellence, Sanofi-Aventis Deutschland GmbH, Industriepark Hoechst, Frankfurt, Germany
| | - Aimo Kannt
- R&D Diabetes, Sanofi-Aventis Deutschland GmbH, Industriepark Hoechst, Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Fraunhofer Innovation Center TheraNova and Goethe University, Frankfurt, Germany
| | - Cecília M P Rodrigues
- Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Simon Cockell
- Bioinformatics Support Unit, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Olivier Govaere
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Ann K Daly
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Michael Allison
- Liver Unit, Cambridge University Hospitals NHS Foundation Trust & Cambridge NIHR Biomedical Research Centre, Cambridge, UK
| | | | - Yong Ook Kim
- Institute of Translational Immunology and Research Center for Immunotherapy, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Anna Lindblom
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism (CVRM), AstraZeneca BioPharmaceuticals R&D, Gothenburg, Sweden
| | - Stephanie Oldham
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism (CVRM), AstraZeneca BioPharmaceuticals R&D, Gaithersburg, MD, USA
| | - Anne-Christine Andréasson
- Bioscience Cardiovascular, Research and Early Development Cardiovascular, Renal and Metabolism (CVRM), AstraZeneca BioPharmaceuticals R&D, Gothenburg, Sweden
| | - Franklin Schlerman
- Inflammation and Immunology Research Unit, Pfizer Worldwide Research and Development, Cambridge, MA, USA
| | | | - Arun Sanyal
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Marta B Afonso
- Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Ramy Younes
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- Boehringer Ingelheim International GmbH, Ingelheim am Rhein, Germany
| | - Yuichiro Amano
- Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Scott L Friedman
- Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Shuang Wang
- Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dipankar Bhattacharya
- Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eric Simon
- Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| | - Valérie Paradis
- Department of Imaging and Pathology, Université Paris Diderot and Hôpital Beaujon, Paris, France
| | - Alastair Burt
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- Newcastle NIHR Biomedical Research Centre, Newcastle upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, UK
| | - Ioanna Maria Grypari
- Department of Pathology, Aretaeion Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Susan Davies
- Department of Cellular Pathology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Ann Driessen
- Department of Pathology, Antwerp University Hospital, Edegem, Belgium
- Department of Molecular Imaging, Pathology, Radiotherapy, Oncology. Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk, Belgium
| | - Hiroaki Yashiro
- Research, Takeda Pharmaceuticals Company Limited, Cambridge, MA, USA
| | | | | | | | - Carla Yunis
- Pfizer, Inc.; Internal Medicine and Hospital, Pfizer Research and Development, Lake Mary, FL, USA
| | - Pierre Bedossa
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- LiverPat, Paris, France
| | | | | | - Sara Wells
- Mary Lyon Centre, MRC Harwell, Harwell Campus, Oxford, UK
| | - Jörn M Schattenberg
- Department of Internal Medicine II, Saarland University Medical Centre, Homburg, Germany
| | - Quentin M Anstee
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- Newcastle NIHR Biomedical Research Centre, Newcastle upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, UK
| | - Dina Tiniakos
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK.
- Department of Pathology, Aretaeion Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
| | - James W Perfield
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA.
| | - Evangelia Petsalaki
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, UK.
| | - Peter Davidsen
- Research and Early Development, Novo Nordisk A/S, Måløv, Copenhagen, Denmark.
- Ferring Pharmaceuticals A/S, International PharmaScience Center, Copenhagen, Denmark.
| | - Antonio Vidal-Puig
- TVP Lab, WT/MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
- Centro de Investigacion Principe Felipe, Valencia, Spain.
| |
Collapse
|
9
|
Gu TJ, Liu PK, Wang YW, Flowers MT, Xu S, Liu Y, Davis DB, Li L. Diazobutanone-assisted isobaric labelling of phospholipids and sulfated glycolipids enables multiplexed quantitative lipidomics using tandem mass spectrometry. Nat Chem 2024; 16:762-770. [PMID: 38365942 DOI: 10.1038/s41557-023-01436-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 12/21/2023] [Indexed: 02/18/2024]
Abstract
Mass spectrometry-based quantitative lipidomics is an emerging field aiming to uncover the intricate relationships between lipidomes and disease development. However, quantifying lipidomes comprehensively in a high-throughput manner remains challenging owing to the diverse lipid structures. Here we propose a diazobutanone-assisted isobaric labelling strategy as a rapid and robust platform for multiplexed quantitative lipidomics across a broad range of lipid classes, including various phospholipids and glycolipids. The diazobutanone reagent is designed to conjugate with phosphodiester or sulfate groups, while accommodating various functional groups on different lipid classes, enabling subsequent isobaric labelling for high-throughput multiplexed quantitation. Our method demonstrates excellent performance in terms of labelling efficiency, detection sensitivity, quantitative accuracy and broad applicability to various biological samples. Finally, we performed a six-plex quantification analysis of lipid extracts from lean and obese mouse livers. In total, we identified and quantified 246 phospholipids in a high-throughput manner, revealing lipidomic changes that may be associated with obesity in mice.
Collapse
Affiliation(s)
- Ting-Jia Gu
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA
| | - Peng-Kai Liu
- Biophysics Graduate program, University of Wisconsin-Madison, Madison, WI, USA
| | - Yen-Wen Wang
- Department of Biostatics, Yale University, New Haven, CT, USA
| | - Matthew T Flowers
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Shuling Xu
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA
| | - Yuan Liu
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA
| | - Dawn B Davis
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Lingjun Li
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA.
- Biophysics Graduate program, University of Wisconsin-Madison, Madison, WI, USA.
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
10
|
Xu Y, Su T, Mishra H, Ando R, Furutani Y, Lu J, Cai M, Suzuki H, Yu W, Qin XY. Corn Oligopeptide Alleviates Nonalcoholic Fatty Liver Disease by Regulating the Sirtuin Signaling Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:6360-6371. [PMID: 38489847 DOI: 10.1021/acs.jafc.3c09058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/17/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD) represents the most prevalent type of chronic liver disease, spanning from simple steatosis to nonalcoholic steatohepatitis (NASH). Corn oligopeptide (CP) is a functional peptide known for its diverse pharmacological effects on metabolism. In this study, we evaluated the protective activity of CP against fatty liver disease. Oral administration of CP significantly reduced body weight gain by 2.95%, serum cholesterol by 22.54%, and liver injury, as evidenced by a reduction of 32.19% in serum aspartate aminotransferase (AST) and 49.10% in alanine aminotransferase (ALT) levels in mice subjected to a high-fat diet (HFD). In a streptozotocin/HFD-induced NASH mouse model, CP attenuated body weight gain by 5.11%, liver injury (with a 34.15% decrease in AST and 11.43% decrease in ALT), and, to some extent, liver inflammation and fibrosis. Proteomic analysis revealed the modulation of oxidative phosphorylation and sirtuin (SIRT) signaling pathways by CP. Remarkably, CP selectively inhibited the hepatic expression of mitochondrial SIRT3 and SIRT5 in both HFD and NASH models. In summary, CP demonstrates a preventive effect against metabolic-stress-induced NAFLD progression by modulating oxidative stress and the SIRT signaling pathway, suggesting the potential of CP as a therapeutic agent for the treatment of NAFLD and advanced-stage NASH.
Collapse
Affiliation(s)
- Yali Xu
- Laboratory for Cellular Function Conversion Technology, RIKEN Center for Integrative Medical Sciences, Yokohama 2300045, Japan
- Department of Intensive Care Unit, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China
| | - Ting Su
- Department of Intensive Care Unit, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China
| | - Hricha Mishra
- Laboratory for Cellular Function Conversion Technology, RIKEN Center for Integrative Medical Sciences, Yokohama 2300045, Japan
| | - Reiko Ando
- Support Unit for Bio-Material Analysis, Research Resources Division, RIKEN Center for Brain Science, Wako 3510106, Japan
| | - Yutaka Furutani
- Department of Laboratory Medicine, The Jikei University School of Medicine, Tokyo 1058461, Japan
| | - Jun Lu
- China National Research Institute of Food and Fermentation Industries, Beijing 100016, China
| | - Muyi Cai
- China National Research Institute of Food and Fermentation Industries, Beijing 100016, China
| | - Harukazu Suzuki
- Laboratory for Cellular Function Conversion Technology, RIKEN Center for Integrative Medical Sciences, Yokohama 2300045, Japan
| | - Wenkui Yu
- Department of Intensive Care Unit, The Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210008, China
| | - Xian-Yang Qin
- Laboratory for Cellular Function Conversion Technology, RIKEN Center for Integrative Medical Sciences, Yokohama 2300045, Japan
| |
Collapse
|
11
|
Yang J, Dai M, Wang Y, Yan Z, Mao S, Liu A, Lu C. A CDAHFD-induced mouse model mimicking human NASH in the metabolism of hepatic phosphatidylcholines and acyl carnitines. Food Funct 2024; 15:2982-2995. [PMID: 38411344 DOI: 10.1039/d3fo05111k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Non-alcoholic steatohepatitis (NASH) is the hepatic manifestation of a cluster of conditions associated with lipid metabolism disorders. Ideal animal models mimicking the human NASH need to be explored to better understand the pathogenesis. A choline-deficient, L-amino acid-defined, high-fat diet (CDAHFD) has recently been used to induce the NASH model, but the advantages are not established. NASH models were induced using the well-known traditional methionine- and choline-deficient (MCD) diet for 5 weeks and the recently used CDAHFD for 3 weeks. Liver phenotypes were analyzed to evaluate the differences in markers related to NASH. Lipidomics and metabolism analyses were used to investigate the effects of dietary regimens on the lipidome of the liver. The CDAHFD induced stronger NASH responses than the MCD, including lipid deposition, liver injury, inflammation, bile acid overload and hepatocyte proliferation. A significant difference in the hepatic lipidome was revealed between the CDAHFD and MCD-induced NASH models. In particular, the CDAHFD reduced the hepatic levels of phosphatidylcholines (PCs) and acylcarnitines (ACs), which was supported by the metabolism analysis and in line with the tendency of human NASH. Pathologically, the CDAHFD could effectively induce a more human-like NASH model over the traditional MCD. The hepatic PCs, ACs and their metabolism in CDAHFD-treated mice were down-regulated, similar to those in human NASH.
Collapse
Affiliation(s)
- Jie Yang
- Department of Hepatopancreatobiliary Surgery, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, 315040, China.
| | - Manyun Dai
- Zhejiang Key Laboratory of Pathophysiology, Department of Physiology and Pharmacology, Health Science Centre, Ningbo University, Ningbo, Zhejiang, 315211, China.
| | - Ying Wang
- Zhejiang Key Laboratory of Pathophysiology, Department of Physiology and Pharmacology, Health Science Centre, Ningbo University, Ningbo, Zhejiang, 315211, China.
| | - Zheng Yan
- Zhejiang Key Laboratory of Pathophysiology, Department of Physiology and Pharmacology, Health Science Centre, Ningbo University, Ningbo, Zhejiang, 315211, China.
| | - Shuqi Mao
- Department of Hepatopancreatobiliary Surgery, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, 315040, China.
| | - Aiming Liu
- Zhejiang Key Laboratory of Pathophysiology, Department of Physiology and Pharmacology, Health Science Centre, Ningbo University, Ningbo, Zhejiang, 315211, China.
| | - Caide Lu
- Department of Hepatopancreatobiliary Surgery, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang, 315040, China.
| |
Collapse
|
12
|
Wu Y, Yin W, Hao P, Chen Y, Yu L, Yu X, Wu Y, Li X, Wang W, Zhou H, Yuan Y, Quan X, Yu Y, Hu B, Chen S, Zhou Z, Sun W. Polysaccharide from Panax japonicus C.A. Mey prevents non-alcoholic fatty liver disease development based on regulating liver metabolism and gut microbiota in mice. Int J Biol Macromol 2024; 260:129430. [PMID: 38228199 DOI: 10.1016/j.ijbiomac.2024.129430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/13/2023] [Accepted: 01/09/2024] [Indexed: 01/18/2024]
Abstract
In this study, a new polysaccharide (PSPJ) with specific molecular weight and monosaccharide compositions was isolated and purified from the water extract of Panacis Japonici Rhizoma (PJR). 16S rRNA analysis and untargeted metabolomic analysis were used to assess PSPJ's efficacy in averting non-alcoholic fatty liver disease (NAFLD). This study indicated that PSPJ significantly reduced liver fat accumulation, the increase in blood lipids and ALT caused by HFD, indicating that PSPJ can prevent NAFLD. We demonstrated through cell experiments that PSPJ does not directly affect liver cells. The gut microbiota disorder and alterations in short-chain fatty acids (SCFAs) induced by the high-fat diet (HFD) were ameliorated by PSPJ, as evidenced by the analysis of 16S rRNA. In particular, supplementing PSPJ reduced the abundance of Turicibacter, Dubosiella, and Staphylococcus, and increased the abundance of Bacteroides, Blautia, and Lactobacillus. Untargeted metabolomic analysis shows that PSPJ improves liver metabolic disorders by regulating arachidonic acid metabolism, carbohydrate digestion and absorption, fatty acid biosynthesis, fatty acid metabolism and retinol metabolism. The findings of our investigation indicate that PSPJ has the potential to modulate liver metabolism through alterations in the composition of intestinal bacteria, hence preventing NAFLD.
Collapse
Affiliation(s)
- Yi Wu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Wen Yin
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Ping Hao
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Yueru Chen
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Lingyun Yu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Xingjian Yu
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento 95817, CA, United States of America
| | - Yu Wu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; Jiangsu Key Laboratory of Pathogen Biology, Department of Pathogen Biology and Immunology, Center for Global Health, Nanjing Medical University, Nanjing 211166, China
| | - Xiaocong Li
- College of Medicine, Hubei Three Gorges Polytechnic, No.31 Stadium Road, Yichang 443000, China
| | - Wenjia Wang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; College of Animal Science and Technology, Ningxia University, China
| | - Hui Zhou
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Yuan Yuan
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiaoyu Quan
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Yue Yu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Bing Hu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Shouhai Chen
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Zhenlei Zhou
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China.
| | - Wenjing Sun
- Guangxi Key Laboratory of Agricultural Resources Chemistry and Biotechnology, College of Biology & Pharmacy, Yulin Normal University, No. 1303 Jiaoyu East Road, Yulin 537000, Guangxi, China.
| |
Collapse
|
13
|
Fernández-Ginés R, Encinar JA, Escoll M, Carnicero-Senabre D, Jiménez-Villegas J, García-Yagüe ÁJ, González-Rodríguez Á, Garcia-Martinez I, Valverde ÁM, Rojo AI, Cuadrado A. Specific targeting of the NRF2/β-TrCP axis promotes beneficial effects in NASH. Redox Biol 2024; 69:103027. [PMID: 38184999 PMCID: PMC10808969 DOI: 10.1016/j.redox.2024.103027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/18/2023] [Accepted: 01/02/2024] [Indexed: 01/09/2024] Open
Abstract
Non-alcoholic steatohepatitis (NASH) is a common chronic liver disease that compromises liver function, for which there is not a specifically approved medicine. Recent research has identified transcription factor NRF2 as a potential therapeutic target. However, current NRF2 activators, designed to inhibit its repressor KEAP1, exhibit unwanted side effects. Alternatively, we previously introduced PHAR, a protein-protein interaction inhibitor of NRF2/β-TrCP, which induces a mild NRF2 activation and selectively activates NRF2 in the liver, close to normal physiological levels. Herein, we assessed the effect of PHAR in protection against NASH and its progression to fibrosis. We conducted experiments to demonstrate that PHAR effectively activated NRF2 in hepatocytes, Kupffer cells, and stellate cells. Then, we used the STAM mouse model of NASH, based on partial damage of endocrine pancreas and insulin secretion impairment, followed by a high fat diet. Non-invasive analysis using MRI revealed that PHAR protects against liver fat accumulation. Moreover, PHAR attenuated key markers of NASH progression, including liver steatosis, hepatocellular ballooning, inflammation, and fibrosis. Notably, transcriptomic data indicate that PHAR led to upregulation of 3 anti-fibrotic genes (Plg, Serpina1a, and Bmp7) and downregulation of 6 pro-fibrotic (including Acta2 and Col3a1), 11 extracellular matrix remodeling, and 8 inflammatory genes. Overall, our study suggests that the mild activation of NRF2 via the protein-protein interaction inhibitor PHAR holds promise as a strategy for addressing NASH and its progression to liver fibrosis.
Collapse
Affiliation(s)
- Raquel Fernández-Ginés
- Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-UAM, Instituto de Investigación Sanitaria La Paz (IdiPaz) and Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, Spain
| | - José Antonio Encinar
- Institute of Research, Development and Innovation in Biotechnology of Elche (IDiBE) and Molecular and Cell Biology Institute (IBMC), Miguel Hernández University (UMH), 03202, Elche, Alicante, Spain
| | - Maribel Escoll
- Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-UAM, Instituto de Investigación Sanitaria La Paz (IdiPaz) and Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, Spain
| | - Daniel Carnicero-Senabre
- Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-UAM, Instituto de Investigación Sanitaria La Paz (IdiPaz) and Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, Spain
| | - José Jiménez-Villegas
- Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-UAM, Instituto de Investigación Sanitaria La Paz (IdiPaz) and Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, Spain
| | - Ángel J García-Yagüe
- Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-UAM, Instituto de Investigación Sanitaria La Paz (IdiPaz) and Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, Spain
| | - Águeda González-Rodríguez
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-UAM, Madrid, Spain
| | - Irma Garcia-Martinez
- Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-UAM, Instituto de Investigación Sanitaria La Paz (IdiPaz), Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Madrid, Spain
| | - Ángela M Valverde
- Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-UAM, Instituto de Investigación Sanitaria La Paz (IdiPaz), Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Madrid, Spain
| | - Ana I Rojo
- Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-UAM, Instituto de Investigación Sanitaria La Paz (IdiPaz) and Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, Spain
| | - Antonio Cuadrado
- Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-UAM, Instituto de Investigación Sanitaria La Paz (IdiPaz) and Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, Madrid, Spain.
| |
Collapse
|
14
|
Kumari D, Gautam J, Sharma V, Gupta SK, Sarkar S, Jana P, Singhal V, Babele P, Kamboj P, Bajpai S, Tandon R, Kumar Y, Dikshit M. Effect of herbal extracts and Saroglitazar on high-fat diet-induced obesity, insulin resistance, dyslipidemia, and hepatic lipidome in C57BL/6J mice. Heliyon 2023; 9:e22051. [PMID: 38027691 PMCID: PMC10663915 DOI: 10.1016/j.heliyon.2023.e22051] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 11/02/2023] [Accepted: 11/02/2023] [Indexed: 12/01/2023] Open
Abstract
We evaluated the effects of select herbal extracts (Tinospora cordifolia [TC], Tinospora cordifolia with Piper longum [TC + PL], Withania somnifera [WS], Glycyrrhiza glabra [GG], AYUSH-64 [AY-64], and Saroglitazar [S]) on various parameters in a diet-induced obesity mouse model. After 12 weeks of oral administration of the herbal extracts in high-fat diet (HFD)-fed C57BL/6J mice, we analyzed plasma biochemical parameters, insulin resistance (IR), liver histology, and the expression of inflammatory and fibrosis markers, along with hepatic lipidome. We also used a 3D hepatic spheroid model to assess their impact on profibrotic gene expression. Among the extracts, TC + PL showed a significant reduction in IR, liver weight, TNF-α, IL4, IL10 expression, and hepatic lipid levels (saturated triglycerides, ceramides, lysophosphocholines, acylcarnitines, diglycerides, and phosphatidylinositol levels). Saroglitazar reversed changes in body weight, IR, plasma triglycerides, glucose, insulin, and various hepatic lipid species (fatty acids, phospholipids, glycerophospholipids, sphingolipids, and triglycerides). With the exception of GG, Saroglitazar, and other extracts protected against palmitic acid-induced fibrosis marker gene expression in the 3D spheroids. TC + PL and Saroglitazar also effectively prevented HFD-induced insulin resistance, inflammation, and specific harmful lipid species in the liver.
Collapse
Affiliation(s)
- Deepika Kumari
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad, 121001, Haryana, India
| | - Jyoti Gautam
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad, 121001, Haryana, India
| | - Vipin Sharma
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad, 121001, Haryana, India
| | - Sonu Kumar Gupta
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad, 121001, Haryana, India
| | - Soumalya Sarkar
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad, 121001, Haryana, India
| | - Pradipta Jana
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad, 121001, Haryana, India
| | - Vikas Singhal
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad, 121001, Haryana, India
| | - Prabhakar Babele
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad, 121001, Haryana, India
| | - Parul Kamboj
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad, 121001, Haryana, India
| | - Sneh Bajpai
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad, 121001, Haryana, India
| | | | - Yashwant Kumar
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad, 121001, Haryana, India
| | - Madhu Dikshit
- Central Drug Research Institute, Sitapur Rd, Sector 10, Jankipuram Extension, Lucknow, Uttar Pradesh, 226031, India.
| |
Collapse
|
15
|
Lee SH, Park H, Yang EK, Lee BR, Jung IH, Kim TH, Goo MJ, Chae Y, Kim MK. GPR119 activation by DA-1241 alleviates hepatic and systemic inflammation in MASH mice through inhibition of NFκB signaling. Biomed Pharmacother 2023; 166:115345. [PMID: 37657264 DOI: 10.1016/j.biopha.2023.115345] [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: 06/22/2023] [Revised: 08/14/2023] [Accepted: 08/19/2023] [Indexed: 09/03/2023] Open
Abstract
BACKGROUND AND PURPOSE GPR119 activation has been suggested to improve hyperglycemia, dyslipidemia and hepatic steatosis. But its therapeutic potential for metabolic dysfunction-associated steatohepatitis (MASH) are underexplored. Here, we investigated the effects of DA-1241, a novel GPR119 agonist, on MASH and explored its underlying mechanism of anti-inflammatory effects. EXPERIMENTAL APPROACH The in vivo anti-MASH effect was assessed by examining the preventive effect in MS-MASH and Ob-MASH mice and the therapeutic effect in MASH with severe hyperglycemia and diet-induced obese (DIO)-MASH mice. Histological and biochemical changes in liver tissue were assessed. Both plasma and hepatic biomarkers related to inflammation and fibrosis were comprehensively analyzed. To understand its mode of action, changes in NFκB signaling were determined in HepG2 and THP-1 cells. KEY RESULTS DA-1241 attenuated MASH progression and alleviated the MASH phenotypes in MASH mouse models with different etiologies, regardless of glucose-lowering activity. In DIO-MASH mice, DA-1241 significantly reduced biochemical parameters related to steatosis, inflammation and fibrosis in the liver with reduced plasma liver enzymes. When used in combination with a dipeptidyl peptidase 4 (DPP4) inhibitor, DA-1241 further improved the MASH phenotype by increasing endogenous glucagon-like peptide-1 effect. Notably, DA-1241 alone and in combination reduced liver inflammation and restored inflammation-related hepatic gene expression, leading to remission of systemic inflammation as assessed by plasma inflammatory cytokines and chemokines. We demonstrated that DA-1241 reduces macrophage differentiation through downregulation of NFκB signaling by activating GPR119. CONCLUSION Our data suggest the therapeutic potential of DA-1241, alone and in combination with a DPP4 inhibitor, for MASH.
Collapse
Affiliation(s)
- Seung-Ho Lee
- Research Headquarter, Dong-A ST Co., Ltd., Yongin 17073, Republic of Korea
| | - Hansu Park
- Research Headquarter, Dong-A ST Co., Ltd., Yongin 17073, Republic of Korea
| | - Eun-Kyoung Yang
- Research Headquarter, Dong-A ST Co., Ltd., Yongin 17073, Republic of Korea
| | - Bo Ram Lee
- Research Headquarter, Dong-A ST Co., Ltd., Yongin 17073, Republic of Korea
| | - Il-Hoon Jung
- Research Headquarter, Dong-A ST Co., Ltd., Yongin 17073, Republic of Korea
| | - Tae-Hyoung Kim
- Research Headquarter, Dong-A ST Co., Ltd., Yongin 17073, Republic of Korea
| | - Moon Jung Goo
- Research Headquarter, Dong-A ST Co., Ltd., Yongin 17073, Republic of Korea
| | - Yuna Chae
- Research Headquarter, Dong-A ST Co., Ltd., Yongin 17073, Republic of Korea
| | - Mi-Kyung Kim
- Research Headquarter, Dong-A ST Co., Ltd., Yongin 17073, Republic of Korea.
| |
Collapse
|
16
|
Miao G, Wang Y, Wang B, Yu H, Liu J, Pan R, Zhou C, Ning J, Zheng Y, Zhang R, Jin X. Multi-omics analysis reveals hepatic lipid metabolism profiles and serum lipid biomarkers upon indoor relevant VOC exposure. ENVIRONMENT INTERNATIONAL 2023; 180:108221. [PMID: 37742460 DOI: 10.1016/j.envint.2023.108221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 08/01/2023] [Accepted: 09/18/2023] [Indexed: 09/26/2023]
Abstract
As a widespread indoor air pollutant, volatile organic compound (VOC) caused various adverse health effects, especial the damage to liver, which has become a growing public concern. However, the current toxic data are intrinsically restricted in the single or major VOC species. Limited knowledge is available regarding toxic effects, biomarkers and underlying mechanisms of real indoor VOC-caused liver damage. Herein, an indoor relevant VOC exposure model was established to evaluate the hepatic adverse outcomes. Machine learning and multi-omics approaches, including liver lipidomic, serum lipidomic and liver transcriptomic, were utilized to uncover the characteristics of liver damage, serum lipid biomarkers, and involved mechanism stimulated by VOC exposure. The result showed that indoor relevant VOC led to the abnormal hepatic lipid metabolism, mainly manifested as a decrease in triacylglycerol (TG) and its precursor substance diacylglycerol (DG), which could be contributed to the occurrence of hepatic adverse outcomes. In terms of serum lipid biomarkers, five lipid biomarkers in serum were uncovered using machine learning to reflect the hepatic lipid disorders induced by VOC. Multi-omics approaches revealed that the upregulated Dgkq disturbed the interconversion of DG and phosphatidic acid (PA), leading to a TG downregulation. The in-depth analysis revealed that VOC down-regulated FoxO transcription factor, contributing to the upregulation of Dgkq. Hence, this study can provide valuable insights into the understanding of liver damage caused by indoor relevant VOC exposure model VOC exposure, from the perspective of multi-omics analysis.
Collapse
Affiliation(s)
- Gan Miao
- Department of Occupational Health and Environmental Health, School of Public Health, Qingdao University, Qingdao, China
| | - Yu Wang
- Department of Occupational Health and Environmental Health, School of Public Health, Qingdao University, Qingdao, China
| | - Baoqiang Wang
- Department of Occupational Health and Environmental Health, School of Public Health, Qingdao University, Qingdao, China
| | - Hongyan Yu
- Department of Occupational Health and Environmental Health, School of Public Health, Qingdao University, Qingdao, China
| | - Jing Liu
- Department of Occupational Health and Environmental Health, School of Public Health, Qingdao University, Qingdao, China
| | - Ruonan Pan
- Department of Occupational Health and Environmental Health, School of Public Health, Qingdao University, Qingdao, China
| | - Chengying Zhou
- Department of Occupational Health and Environmental Health, School of Public Health, Qingdao University, Qingdao, China
| | - Jie Ning
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang, China
| | - Yuxin Zheng
- Department of Occupational Health and Environmental Health, School of Public Health, Qingdao University, Qingdao, China
| | - Rong Zhang
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang, China.
| | - Xiaoting Jin
- Department of Occupational Health and Environmental Health, School of Public Health, Qingdao University, Qingdao, China.
| |
Collapse
|
17
|
Takezaki D, Morizane S, Ikeda K, Iseki M, Sakamoto Y, Kawakami Y, Hashiguchi T, Shirakata Y, Nishina S, Mukai T. Co-occurrence of non-alcoholic steatohepatitis exacerbates psoriasis associated with decreased adiponectin expression in a murine model. Front Immunol 2023; 14:1214623. [PMID: 37646025 PMCID: PMC10461570 DOI: 10.3389/fimmu.2023.1214623] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 07/24/2023] [Indexed: 09/01/2023] Open
Abstract
Introduction Clinical studies have suggested a bidirectional association between non-alcoholic steatohepatitis (NASH) and psoriasis, affecting each other's development and severity. Here, we explored bidirectional causal linkages between NASH and psoriasis using a murine model. Methods NASH was induced in mice by streptozotocin injection at 2 days of age and by high-fat diet feeding (STAM™ model). Psoriasis was induced by topical application of imiquimod (IMQ) on the ear. The severities of liver damage and psoriatic skin changes were determined using histological analysis. Gene expression in the skin tissues was evaluated using quantitative PCR analysis. Serum cytokine levels were determined using enzyme-linked immunosorbent assay. To examine the innate immune responses of normal human epidermal keratinocytes (NHEKs), the cells were treated with interleukin (IL)-17A, tumor necrosis factor (TNF)-α, and AdipoRon, an adiponectin receptor agonist. Results and Discussion There were no differences in the degree of liver tissue damage (fat deposition, inflammation, and fibrosis) between NASH mice with and those without psoriasis. Conversely, the co-occurrence of NASH significantly augmented psoriatic skin changes, represented by epidermal hyperplasia, in psoriatic mice. Pro-inflammatory cytokines were expressed in the inflamed skin of psoriatic mice, and the expression of genes, especially Il23a, Il1b, Il36g, and Mip2, was significantly upregulated by the co-occurrence of NASH. The expression of keratinocyte activation marker genes Defb4b and Krt16 was also upregulated by the co-occurrence of NASH. The serum TNF-α and IL-17 levels were increased by the co-occurrence of NASH and psoriasis. The serum adiponectin levels decreased in NASH mice compared with that in non-NASH mice. In NHEK culture, TNF-α and IL-17A synergistically upregulated CXCL1, CXCL8, and IL1B expression. The upregulated pro-inflammatory gene expression was suppressed by AdipoRon treatment, reflecting the anti-inflammatory capacity of adiponectin. Conclusion The co-occurrence of NASH exacerbated psoriatic skin changes associated with increased serum inflammatory cytokine levels and decreased serum adiponectin levels. Combined with in vitro findings, increased inflammatory cytokine levels and decreased adiponectin levels likely promote innate immune responses in epidermal keratinocytes in psoriatic skin lesions. Overall, therapeutic intervention for co-occurring NASH is essential to achieve a favorable prognosis of psoriasis in clinical practice.
Collapse
Affiliation(s)
- Daiki Takezaki
- Department of Dermatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
- Department of Immunology and Molecular Genetics, Kawasaki Medical School, Okayama, Japan
| | - Shin Morizane
- Department of Dermatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kenta Ikeda
- Department of Dermatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
- Department of Dermatology, National Hospital Organization Iwakuni Clinical Center, Yamaguchi, Japan
| | - Masanori Iseki
- Department of Immunology and Molecular Genetics, Kawasaki Medical School, Okayama, Japan
| | - Yuma Sakamoto
- Department of Immunology and Molecular Genetics, Kawasaki Medical School, Okayama, Japan
| | - Yoshio Kawakami
- Department of Dermatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | | | | | - Sohji Nishina
- Department of Gastroenterology and Hepatology, Kawasaki Medical School, Okayama, Japan
| | - Tomoyuki Mukai
- Department of Immunology and Molecular Genetics, Kawasaki Medical School, Okayama, Japan
| |
Collapse
|
18
|
Zhang B, Zhang B, Lai RC, Sim WK, Lam KP, Lim SK. MSC-sEV Treatment Polarizes Pro-Fibrotic M2 Macrophages without Exacerbating Liver Fibrosis in NASH. Int J Mol Sci 2023; 24:ijms24098092. [PMID: 37175803 PMCID: PMC10179074 DOI: 10.3390/ijms24098092] [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: 03/03/2023] [Revised: 04/20/2023] [Accepted: 04/21/2023] [Indexed: 05/15/2023] Open
Abstract
Mesenchymal stem/stromal cell small extracellular vesicles (MSC-sEVs) have shown promise in treating a wide range of animal models of various human diseases, which has led to their consideration for clinical translation. However, the possibility of contraindication for MSC-sEV use is an important consideration. One concern is that MSC-sEVs have been shown to induce M2 macrophage polarization, which is known to be pro-fibrotic, potentially indicating contraindication in fibrotic diseases such as liver fibrosis. Despite this concern, previous studies have shown that MSC-sEVs alleviate high-fat diet (HFD)-induced non-alcoholic steatohepatitis (NASH). To assess whether the pro-fibrotic M2 macrophage polarization induced by MSC-sEVs could worsen liver fibrosis, we first verified that our MSC-sEV preparations could promote M2 polarization in vitro prior to their administration in a mouse model of NASH. Our results showed that treatment with MSC-sEVs reduced or had comparable NAFLD Activity Scores and liver fibrosis compared to vehicle- and Telmisartan-treated animals, respectively. Although CD163+ M2 macrophages were increased in the liver, and serum IL-6 levels were reduced in MSC-sEV treated animals, our data suggests that MSC-sEV treatment was efficacious in reducing liver fibrosis in a mouse model of NASH despite an increase in pro-fibrotic M2 macrophage polarization.
Collapse
Affiliation(s)
- Bin Zhang
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Biyan Zhang
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, Immunos, Singapore 138648, Singapore
| | - Ruenn Chai Lai
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Wei Kian Sim
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Kong Peng Lam
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, Immunos, Singapore 138648, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 5 Science Drive 2, Singapore 117545, Singapore
- School of Biological Sciences, College of Science, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Sai Kiang Lim
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
- Department of Surgery, YLL School of Medicine, NUS, 5 Lower Kent Ridge Road, Singapore 119074, Singapore
| |
Collapse
|
19
|
Shin JH, Lee Y, Song EJ, Lee D, Jang SY, Byeon HR, Hong MG, Lee SN, Kim HJ, Seo JG, Jun DW, Nam YD. Faecalibacterium prausnitzii prevents hepatic damage in a mouse model of NASH induced by a high-fructose high-fat diet. Front Microbiol 2023; 14:1123547. [PMID: 37007480 PMCID: PMC10060964 DOI: 10.3389/fmicb.2023.1123547] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 02/20/2023] [Indexed: 03/18/2023] Open
Abstract
IntroductionNonalcoholic steatohepatitis (NASH) is an advanced nonalcoholic fatty liver disease characterized by chronic inflammation and fibrosis. A dysbiosis of the gut microbiota has been associated with the pathophysiology of NASH, and probiotics have proven helpful in its treatment and prevention. Although both traditional and next-generation probiotics have the potential to alleviate various diseases, studies that observe the therapeutic effect of next-generation probiotics on NASH are lacking. Therefore, we investigated whether a next-generation probiotic candidate, Faecalibacterium prausnitzii, contributed to the mitigation of NASH.MethodsIn this study, we conducted 16S rRNA sequencing analyses in patients with NASH and healthy controls. To test F. prausnitzii could alleviate NASH symptoms, we isolated four F. prausnitzii strains (EB-FPDK3, EB-FPDK9, EB-FPDK11, and EB-FPYYK1) from fecal samples collected from four healthy individuals. Mice were maintained on a high-fructose high-fat diet for 16 weeks to induce a NASH model and received oral administration of the bacterial strains. Changes in characteristic NASH phenotypes were assessed via oral glucose tolerance tests, biochemical assays, and histological analyses.Results16S rRNA sequencing analyses confirmed that the relative abundance of F. prausnitzii reduced significantly in patients with NASH compared to healthy controls (p < 0.05). In the NASH mice, F. prausnitzii supplementation improved glucose homeostasis, prevented hepatic lipid accumulation, curbed liver damage and fibrosis, restored damaged gut barrier functions, and alleviated hepatic steatosis and liver inflammation. Furthermore, real-time PCR assays documented that the four F. prausnitzii strains regulated the expression of genes related to hepatic steatosis in these mice.DiscussionOur study, therefore, confirms that the administration of F. prausnitzii bacteria can alleviate NASH symptoms. We propose that F. prausnitzii has the potential to contribute to the next-generation probiotic treatment of NASH.
Collapse
Affiliation(s)
- Ji-Hee Shin
- Research Group of Personalized Diet, Korea Food Research Institute, Wanju-gun, Jeollabuk-do, Republic of Korea
| | - Yoonmi Lee
- R&D Center, Enterobiome Inc., Goyang-si, Republic of Korea
| | - Eun-Ji Song
- Research Group of Personalized Diet, Korea Food Research Institute, Wanju-gun, Jeollabuk-do, Republic of Korea
| | - Dokyung Lee
- R&D Center, Enterobiome Inc., Goyang-si, Republic of Korea
| | - Seo-Yul Jang
- R&D Center, Enterobiome Inc., Goyang-si, Republic of Korea
| | - Hye Rim Byeon
- R&D Center, Enterobiome Inc., Goyang-si, Republic of Korea
| | - Moon-Gi Hong
- R&D Center, Enterobiome Inc., Goyang-si, Republic of Korea
| | - Sang-Nam Lee
- R&D Center, Enterobiome Inc., Goyang-si, Republic of Korea
| | - Hyun-Jin Kim
- Division of Applied Life Science (BK21 Four), Institute of Agriculture and Life Science, Gyeongsang National University, Jinju-si, Republic of Korea
| | - Jae-Gu Seo
- R&D Center, Enterobiome Inc., Goyang-si, Republic of Korea
- *Correspondence: Jae-Gu Seo,
| | - Dae Won Jun
- Department of Internal Medicine, Hanyang University, College of Medicine, Seoul, Republic of Korea
- Dae Won Jun,
| | - Young-Do Nam
- Research Group of Personalized Diet, Korea Food Research Institute, Wanju-gun, Jeollabuk-do, Republic of Korea
- Young-Do Nam,
| |
Collapse
|
20
|
Green CD, Spiegel S. Preclinical models of non-alcoholic steatohepatitis leading to hepatocellular carcinoma. Adv Biol Regul 2023; 87:100925. [PMID: 36706611 DOI: 10.1016/j.jbior.2022.100925] [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: 10/16/2022] [Accepted: 10/20/2022] [Indexed: 01/29/2023]
Abstract
Hepatocellular carcinoma (HCC) is the third leading cause of cancer related deaths worldwide and its incidence is increasing due to endemic obesity and the growing burden of non-alcoholic steatohepatitis (NASH) associated liver cancer. Although much is known about the clinical and histological pathology of NASH-driven HCC in humans, its etiology remains unclear and there is a lack of reliable biomarkers and limited effective therapies. Progress has been hampered by the scarcity of standardized animal models that recapitulate the gradual progression of NASH towards HCC observed in humans. Here we review existing mouse models and their suitability for studying NASH-driven HCC with special emphasis on a preclinical model that we recently developed that faithfully mimics all the clinical endpoints of progression of the human disease. Moreover, it is highly translatable, allows the use of gene-targeted mice, and is suitable for gaining knowledge of how NASH progresses to HCC and development of new targets for treatment.
Collapse
Affiliation(s)
- Christopher D Green
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA.
| | - Sarah Spiegel
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA.
| |
Collapse
|
21
|
Ishizawa S, Nishi A, Kaifuchi N, Shimobori C, Nahata M, Yamada C, Iizuka S, Ohbuchi K, Nishiyama M, Fujitsuka N, Kono T, Yamamoto M. Integrated analysis of effect of daisaikoto, a traditional Japanese medicine, on the metabolome and gut microbiome in a mouse model of nonalcoholic fatty liver disease. Gene X 2022; 846:146856. [PMID: 36067864 DOI: 10.1016/j.gene.2022.146856] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 08/26/2022] [Accepted: 08/29/2022] [Indexed: 11/04/2022] Open
Abstract
Dysregulation of lipid metabolism and diabetes are risk factors for nonalcoholic fatty liver disease (NAFLD), and the gut-liver axis and intestinal microbiome are known to be highly associated with the pathogenesis of this disease. In Japan, the traditional medicine daisaikoto (DST) is prescribed for individuals affected by hepatic dysfunction. Herein, we evaluated the therapeutic potential of DST for treating NAFLD through modification of the liver and stool metabolome and microbiome by using STAM mice as a model of NAFLD. STAM mice were fed a high-fat diet with or without 3 % DST for 3 weeks. Plasma and liver of STAM, STAM with DST, and C57BL/6J ("Normal") mice were collected at 9 weeks, and stools at 4, 6, and 9 weeks of age. The liver pathology, metabolome and stool microbiome were analyzed. DST ameliorated the NAFLD activity score of STAM mice and decreased the levels of several liver lipid mediators such as arachidonic acid and its derivatives. In normal mice, nine kinds of family accounted for 94.1 % of microbiome composition; the total percentage of these family was significantly decreased in STAM mice (45.6 %), and DST administration improved this imbalance in microbiome composition (65.2 %). In stool samples, DST increased ursodeoxycholic acid content and altered several amino acids, which were correlated with changes in the gut microbiome and liver metabolites. In summary, DST ameliorates NAFLD by decreasing arachidonic acid metabolism in the liver; this amelioration seems to be associated with crosstalk among components of the liver, intestinal environment, and microbiome.
Collapse
Affiliation(s)
- Shiori Ishizawa
- Tsumura Advanced Technology Research Laboratories, Tsumura & Co., Ami, Ibaraki 300-1192, Japan
| | - Akinori Nishi
- Tsumura Advanced Technology Research Laboratories, Tsumura & Co., Ami, Ibaraki 300-1192, Japan.
| | - Noriko Kaifuchi
- Tsumura Advanced Technology Research Laboratories, Tsumura & Co., Ami, Ibaraki 300-1192, Japan
| | - Chika Shimobori
- Tsumura Advanced Technology Research Laboratories, Tsumura & Co., Ami, Ibaraki 300-1192, Japan
| | - Miwa Nahata
- Tsumura Kampo Research Laboratories, Tsumura & Co., Ami, Ibaraki 300-1192, Japan
| | - Chihiro Yamada
- Tsumura Kampo Research Laboratories, Tsumura & Co., Ami, Ibaraki 300-1192, Japan
| | - Seiichi Iizuka
- Tsumura Advanced Technology Research Laboratories, Tsumura & Co., Ami, Ibaraki 300-1192, Japan
| | - Katsuya Ohbuchi
- Tsumura Advanced Technology Research Laboratories, Tsumura & Co., Ami, Ibaraki 300-1192, Japan
| | - Mitsue Nishiyama
- Tsumura Advanced Technology Research Laboratories, Tsumura & Co., Ami, Ibaraki 300-1192, Japan
| | - Naoki Fujitsuka
- Tsumura Kampo Research Laboratories, Tsumura & Co., Ami, Ibaraki 300-1192, Japan
| | - Toru Kono
- Department of Gastroenterological Surgery I, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido 060-8638, Japan; Center for Clinical and Biomedical Research, Sapporo Higashi Tokushukai Hospital, Sapporo, Hokkaido 065-0033, Japan
| | - Masahiro Yamamoto
- Tsumura Advanced Technology Research Laboratories, Tsumura & Co., Ami, Ibaraki 300-1192, Japan
| |
Collapse
|
22
|
Flessa CM, Nasiri-Ansari N, Kyrou I, Leca BM, Lianou M, Chatzigeorgiou A, Kaltsas G, Kassi E, Randeva HS. Genetic and Diet-Induced Animal Models for Non-Alcoholic Fatty Liver Disease (NAFLD) Research. Int J Mol Sci 2022; 23:15791. [PMID: 36555433 PMCID: PMC9780957 DOI: 10.3390/ijms232415791] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/05/2022] [Accepted: 12/10/2022] [Indexed: 12/15/2022] Open
Abstract
A rapidly increasing incidence of non-alcoholic fatty liver disease (NAFLD) is noted worldwide due to the adoption of western-type lifestyles and eating habits. This makes the understanding of the molecular mechanisms that drive the pathogenesis of this chronic disease and the development of newly approved treatments of utmost necessity. Animal models are indispensable tools for achieving these ends. Although the ideal mouse model for human NAFLD does not exist yet, several models have arisen with the combination of dietary interventions, genetic manipulations and/or administration of chemical substances. Herein, we present the most common mouse models used in the research of NAFLD, either for the whole disease spectrum or for a particular disease stage (e.g., non-alcoholic steatohepatitis). We also discuss the advantages and disadvantages of each model, along with the challenges facing the researchers who aim to develop and use animal models for translational research in NAFLD. Based on these characteristics and the specific study aims/needs, researchers should select the most appropriate model with caution when translating results from animal to human.
Collapse
Affiliation(s)
- Christina-Maria Flessa
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
| | - Narjes Nasiri-Ansari
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Ioannis Kyrou
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
- Research Institute for Health and Wellbeing, Coventry University, Coventry CV1 5FB, UK
- Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham B4 7ET, UK
- Laboratory of Dietetics and Quality of Life, Department of Food Science and Human Nutrition, School of Food and Nutritional Sciences, Agricultural University of Athens, 11855 Athens, Greece
| | - Bianca M. Leca
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
| | - Maria Lianou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Antonios Chatzigeorgiou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Gregory Kaltsas
- Endocrine Unit, 1st Department of Propaedeutic Internal Medicine, Laiko Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Eva Kassi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Endocrine Unit, 1st Department of Propaedeutic Internal Medicine, Laiko Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Harpal S. Randeva
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
| |
Collapse
|
23
|
Huang CY, Chen HW, Lo CW, Wang YR, Li CC, Liu KL, Lii CK. Luteolin ameliorates palmitate-induced lipotoxicity in hepatocytes by mediating endoplasmic reticulum stress and autophagy. Food Chem Toxicol 2022; 171:113554. [PMID: 36509263 DOI: 10.1016/j.fct.2022.113554] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 11/21/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022]
Abstract
Abnormal accumulation of lipids in liver leads to uncontrolled endoplasmic reticulum (ER) stress and autophagy. Luteolin is known to have antioxidant, anti-inflammatory, and anti-cancer properties, but whether it protects against lipotoxicity in liver remains unclear. In this study, we challenged AML12 liver cells and mouse primary hepatocytes with palmitic acid (PA) with or without luteolin pretreatment. In the presence of PA, reactive oxygen species (ROS) production was increased at 3 h, followed by enhancement of expression of p-PERK, ATF4, p-eIF2α, CHOP, and TXNIP (ER stress markers) and p-p62 and LC3II/LC3I ratio (autophagy markers), in both primary hepatocytes and AML12 cells. When PA treatment was extended up to 24 h, apoptosis was induced as evidenced by an increase in caspase-3 activation. RFP-GFP-LC3B transfection further revealed that the fusion of autophagosomes with lysosomes was damaged by PA. With luteolin treatment, the expression of antioxidant enzymes, i.e., heme oxygenase-1 and glutathione peroxidase, was upregulated, and PA-induced ROS production, ER stress, and cell death were dose-dependently ameliorated. Luteolin could also reverse the damage caused to autophagic flux. These results indicate that luteolin protects hepatocytes against PA assault by enhancing antioxidant defense, which can attenuate ER stress and autophagy as well as promote autophagic flux.
Collapse
Affiliation(s)
- Chun-Yin Huang
- Department of Nutrition, China Medical University, Taichung, Taiwan
| | - Haw-Wen Chen
- Department of Nutrition, China Medical University, Taichung, Taiwan
| | - Chia-Wen Lo
- Department of Nutrition, China Medical University, Taichung, Taiwan
| | - Yu-Ru Wang
- Department of Nutrition, China Medical University, Taichung, Taiwan
| | - Chien-Chun Li
- Department of Nutrition, Chung Shan Medical University, Taichung, Taiwan; Department of Nutrition, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Kai-Li Liu
- Department of Nutrition, Chung Shan Medical University, Taichung, Taiwan; Department of Nutrition, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chong-Kuei Lii
- Department of Nutrition, China Medical University, Taichung, Taiwan; Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan.
| |
Collapse
|
24
|
Roh PR, Kim SM, Kang BY, Mun KD, Park JG, Kang MW, Hur W, Han JW, Nam H, Yoon SK, Sung PS. Tenofovir alafenamide alleviates nonalcoholic steatohepatitis in mice by blocking the phosphorylation of AKT in intrahepatic mononuclear phagocytes. Biomed Pharmacother 2022; 156:113952. [PMID: 36411662 DOI: 10.1016/j.biopha.2022.113952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/25/2022] [Accepted: 11/01/2022] [Indexed: 11/06/2022] Open
|
25
|
Phung HH, Lee CH. Mouse models of nonalcoholic steatohepatitis and their application to new drug development. Arch Pharm Res 2022; 45:761-794. [DOI: 10.1007/s12272-022-01410-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 10/10/2022] [Indexed: 11/05/2022]
|
26
|
Najimi M, Michel S, Binda MM, Gellynck K, Belmonte N, Mazza G, Gordillo N, Vainilovich Y, Sokal E. Human Allogeneic Liver-Derived Progenitor Cells Significantly Improve NAFLD Activity Score and Fibrosis in Late-Stage NASH Animal Model. Cells 2022; 11:cells11182854. [PMID: 36139429 PMCID: PMC9497074 DOI: 10.3390/cells11182854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 09/05/2022] [Accepted: 09/06/2022] [Indexed: 11/26/2022] Open
Abstract
Accumulated experimental and clinical evidence supports the development of human allogeneic liver-derived progenitor cells (HALPCs) to treat fibro-inflammatory liver diseases. The aim of the present study was to evaluate their therapeutic effect in a non-alcoholic steatohepatitis (NASH)-STAM mouse model. The immune signaling characteristics of HALPCs were first assessed in vitro. Upon inflammation treatment, HALPCs secreted large amounts of potent bioactive prostaglandin E2 and indoleamine 2,3-dioxygenase, which significantly reduced CD4+ T-lymphocyte proliferation and secretion of proinflammatory cytokines. In vivo, HALPCs were intravenously administered as single or triple shots (of a dose of 12.5 × 106 cells/kg BW) in STAM mice. Transplantation of HALPCs was associated with a significant decrease in the NAFLD activity score at an early stage and in both inflammation and hepatocyte ballooning scores in late-stage NASH. Sirius red staining analyses revealed decreased collagen deposition in the pericentral region at both stages of NASH. Altogether, these findings showed the anti-inflammatory and anti-fibrotic features of HALPCs in an in vivo NASH model, which suggests their potential to reverse the progression of this chronic fibro-inflammatory disease.
Collapse
Affiliation(s)
- Mustapha Najimi
- Cellaïon, 1435 Mont-Saint-Guibert, Belgium
- UCLouvain, Laboratory of Pediatric Hepatology and Cell Therapy (PEDI), Institute of Experimental and Clinical Research (IREC), 1200 Brussels, Belgium
- Correspondence: (M.N.); (E.S.); Tel.: +32-10-39-43-00 (M.N.)
| | | | | | | | | | | | | | | | - Etienne Sokal
- Cellaïon, 1435 Mont-Saint-Guibert, Belgium
- UCLouvain, Laboratory of Pediatric Hepatology and Cell Therapy (PEDI), Institute of Experimental and Clinical Research (IREC), 1200 Brussels, Belgium
- Correspondence: (M.N.); (E.S.); Tel.: +32-10-39-43-00 (M.N.)
| |
Collapse
|
27
|
Age- and Diet-Dependent Changes in Hepatic Lipidomic Profiles of Phospholipids in Male Mice: Age Acceleration in Cyp2b-Null Mice. J Lipids 2022; 2022:7122738. [PMID: 35391786 PMCID: PMC8983274 DOI: 10.1155/2022/7122738] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 03/03/2022] [Indexed: 11/17/2022] Open
Abstract
Increases in traditional serum lipid profiles are associated with obesity, cancer, and cardiovascular disease. Recent lipidomic analysis has indicated changes in serum lipidome profiles, especially in regard to specific phosphatidylcholines, associated with obesity. However, little work has evaluated murine hepatic liver lipidomic profiles nor compared these profiles across age, high-fat diet, or specific genotypes, in this case the lack of hepatic Cyp2b enzymes. In this study, the effects of age (9 months old), high-fat diet (4.5 months old), and the loss of three primarily hepatic xeno- and endobiotic metabolizing cytochrome P450 (Cyp) enzymes, Cyp2b9, Cyp2b10, and Cyp2b13 (Cyp2b-null mice), on the male murine hepatic lipidome were compared. Hierarchical clustering and principal component analysis show that age perturbs hepatic phospholipid profiles and serum lipid markers the most compared to young mice, followed by a high-fat diet and then loss of Cyp2b. Several lipid biomarkers such as PC/PE ratios, PE 38 : 6, and LPC concentrations indicate greater potential for NAFLD and hypertension with mixed effects in Cyp2b-null mice(less NAFLD and greater hypertension-associated markers). Lipid profiles from older mice contain greater total and n-6 fatty acids than normal diet (ND)-fed young mice; however, surprisingly, young Cyp2b-null mice contain high n-6 : n-3 ratios. Overall, the lack of Cyp2b typically enhanced adverse physiological parameters observed in the older (9 mo) mice with increased weight gain combined with a deteriorating cholesterol profile, but not necessarily all phospholipid profiles were adversely perturbed.
Collapse
|
28
|
Manifold Roles of Ceramide Metabolism in Non-Alcoholic Fatty Liver Disease and Liver Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1372:157-168. [DOI: 10.1007/978-981-19-0394-6_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
29
|
Gu CY, Lee TKW. Preclinical mouse models of hepatocellular carcinoma: An overview and update. Exp Cell Res 2022; 412:113042. [PMID: 35101391 DOI: 10.1016/j.yexcr.2022.113042] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 01/15/2022] [Accepted: 01/19/2022] [Indexed: 11/29/2022]
|
30
|
Newberry EP, Hall Z, Xie Y, Molitor EA, Bayguinov PO, Strout GW, Fitzpatrick JA, Brunt EM, Griffin JL, Davidson NO. Liver-Specific Deletion of Mouse Tm6sf2 Promotes Steatosis, Fibrosis, and Hepatocellular Cancer. Hepatology 2021; 74:1203-1219. [PMID: 33638902 PMCID: PMC8390580 DOI: 10.1002/hep.31771] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 12/17/2020] [Accepted: 01/13/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIMS Human transmembrane 6 superfamily 2 (TM6SF2) variant rs58542926 is associated with NAFLD and HCC. However, conflicting reports in germline Tm6sf2 knockout mice suggest no change or decreased very low density lipoprotein (VLDL) secretion and either unchanged or increased hepatic steatosis, with no increased fibrosis. We generated liver-specific Tm6Sf2 knockout mice (Tm6 LKO) to study VLDL secretion and the impact on development and progression of NAFLD. APPROACH AND RESULTS Two independent lines of Tm6 LKO mice exhibited spontaneous hepatic steatosis. Targeted lipidomic analyses showed increased triglyceride species whose distribution and abundance phenocopied findings in mice with liver-specific deletion of microsomal triglyceride transfer protein. The VLDL triglyceride secretion was reduced with small, underlipidated particles and unchanged or increased apolipoprotein B. Liver-specific adeno-associated viral, serotype 8 (AAV8) rescue using either wild-type or mutant E167K-Tm6 reduced hepatic steatosis and improved VLDL secretion. The Tm6 LKO mice fed a high milk-fat diet for 3 weeks exhibited increased steatosis and fibrosis, and those phenotypes were further exacerbated when mice were fed fibrogenic, high fat/fructose diets for 20 weeks. In two models of HCC, either neonatal mice injected with streptozotocin (NASH/STAM) and high-fat fed or with diethylnitrosamine injection plus fibrogenic diet feeding, Tm6 LKO mice exhibited increased steatosis, greater tumor burden, and increased tumor area versus Tm6 flox controls. Additionally, diethylnitrosamine-injected and fibrogenic diet-fed Tm6 LKO mice administered wild-type Tm6 or E167K-mutant Tm6 AAV8 revealed significant tumor attenuation, with tumor burden inversely correlated with Tm6 protein levels. CONCLUSIONS Liver-specific Tm6sf2 deletion impairs VLDL secretion, promoting hepatic steatosis, fibrosis, and accelerated development of HCC, which was mitigated with AAV8- mediated rescue.
Collapse
Affiliation(s)
- Elizabeth P. Newberry
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Zoe Hall
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge, United Kingdom
- Biomolecular Medicine, Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Yan Xie
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Elizabeth A. Molitor
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Peter O. Bayguinov
- Washington University Center for Cellular Imaging, Washington University in Saint Louis, St. Louis, MO 63130
| | - Gregory W. Strout
- Washington University Center for Cellular Imaging, Washington University in Saint Louis, St. Louis, MO 63130
| | - James A.J. Fitzpatrick
- Washington University Center for Cellular Imaging, Washington University in Saint Louis, St. Louis, MO 63130
- Departments of Cell Biology & Physiology and Neuroscience, Washington University School of Medicine, Louis, St. Louis, MO 63130
- Department of Biomedical Engineering, Washington University in Saint Louis, St. Louis, MO 63130
| | - Elizabeth M. Brunt
- Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Julian L. Griffin
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge, United Kingdom
- Biomolecular Medicine, Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Nicholas O. Davidson
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
31
|
Takihata Y, Kawauchi S, Ogata S, Nishidate I, Sato S, Yamamoto J, Kishi Y. In vivo diffuse reflectance spectroscopic analysis of fatty liver with inflammation in mice. Surg Open Sci 2021; 6:21-28. [PMID: 34458710 PMCID: PMC8379345 DOI: 10.1016/j.sopen.2021.07.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 06/13/2021] [Accepted: 07/01/2021] [Indexed: 11/25/2022] Open
Abstract
Background Nonalcoholic steatohepatitis is a progressive liver disease that can lead to cirrhosis, hepatocellular carcinoma, and hepatic failure. Thus, the diagnosis of nonalcoholic steatohepatitis, especially discrimination from nonalcoholic fatty liver, is crucial, but reliable methods other than invasive biopsy have not been established yet. In this study, we investigated the usefulness of diffuse reflectance spectroscopy, which does not require tissue collection, to evaluate the pathological states of fatty liver with inflammation. Materials and Methods We performed in vivo optical fiber-based diffuse reflectance spectroscopy in both the near-infrared and visible spectral regions for livers in STAM mice, which typically show steatosis at 6 weeks, steatohepatitis at 8 weeks, and fibrosis at 12 weeks of age. After diffuse reflectance spectroscopy, all of the liver tissues were histologically analyzed and scored on the basis of the rodent nonalcoholic fatty liver disease scoring system. We examined correlations between the diffuse reflectance spectra and scores associated with steatosis and inflammation. Results and Conclusion The results showed that the second derivative values of reflectance at 1204 nm, the lipid absorption peak in the near-infrared region, were strongly correlated with steatosis scores (r = 0.9172, P < .0001, n = 20) and that the differences of the first derivative values of reflectance in the visible region (570 nm − 550 nm) that reflect hemoglobin deoxygenation were significantly correlated with inflammation scores (r = 0.5260, P = .0172, n = 20). These results suggest that our diffuse reflectance spectroscopy method is useful for diagnosis of the states of steatosis with inflammation in livers and hence nonalcoholic steatohepatitis. Optical reflectance signals are used to diagnose nonalcoholic steatohepatitis in mice. The near-infrared reflectance signals are strongly correlated with steatosis scores. The visible reflectance signals are significantly correlated with inflammation scores. Nonalcoholic steatohepatitis can be noninvasively detected by reflectance signals.
Collapse
Affiliation(s)
- Yasuhiro Takihata
- Department of Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan.,Division of Bioinformation and Therapeutic Systems, National Defense Medical College Research Institute, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan
| | - Satoko Kawauchi
- Division of Bioinformation and Therapeutic Systems, National Defense Medical College Research Institute, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan
| | - Sho Ogata
- Department of Pathology and Laboratory Medicine, National Defense Medical College Hospital, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan
| | - Izumi Nishidate
- Graduate School of Bio-Applications & Systems Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo 184-8588, Japan
| | - Shunichi Sato
- Division of Bioinformation and Therapeutic Systems, National Defense Medical College Research Institute, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan
| | - Junji Yamamoto
- Department of Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan
| | - Yoji Kishi
- Department of Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan
| |
Collapse
|
32
|
Ma P, Wang Z, Wang Y, Hou B, Sun J, Tian H, Li B, Shui G, Yang X, Yang X, Qiang G, Liew CW, Du G. Integration of Metabolomics and Transcriptomics Reveals Ketone Body and Lipid Metabolism Disturbance Related to ER Stress in the Liver. J Proteome Res 2021; 20:3875-3888. [PMID: 34270263 DOI: 10.1021/acs.jproteome.1c00167] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Once protein synthesis is excessive or misfolded protein becomes aggregated, which eventually overwhelms the capacity of the endoplasmic reticulum (ER), a state named ER stress would be reached. ER stress could affect many tissues, especially the liver, in which nonalcoholic fatty liver disease, liver steatosis, etc. have been reported relative. However, there is still a lack of systematic insight into ER stress in the liver, which can be obtained by integrating metabolomics and transcriptomics of the tissue. Here, tunicamycin was utilized to induce ER stress in C57BL/6N mice. Microarray and untargeted metabolomics were performed to identify the genes and metabolites significantly altered in liver tissues. Surprisingly, apart from the predictable unfolded protein response, liver lipid, arginine, and proline metabolisms were affirmed to be related to ER stress. Also, the ketone body metabolism changed most prominently in response to ER stress, with few studies backing. What is more, succinate receptor 1 (Sucnr1) may be a novel marker and therapeutical target of liver ER stress. In this study, the combination of the metabolome and transcriptome provided reliable information about liver pathological processes, including key relative pathways, potential markers, and targets involved in ER stress of the liver.
Collapse
Affiliation(s)
- Peng Ma
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College and Beijing Key Laboratory of Drug Target and Screening Research, Beijing 100050, China
| | - Zijing Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College and Beijing Key Laboratory of Drug Target and Screening Research, Beijing 100050, China
| | - Yisa Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College and Beijing Key Laboratory of Drug Target and Screening Research, Beijing 100050, China.,College of Pharmacy, Harbin University of Commerce, Harbin 510006, China
| | - Biyu Hou
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College and Beijing Key Laboratory of Drug Target and Screening Research, Beijing 100050, China
| | - Jialin Sun
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qindao 266000, China
| | - He Tian
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Bowen Li
- LipidALL Technologies Ltd., Changzhou 213125, China
| | - Guanghou Shui
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xiuying Yang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College and Beijing Key Laboratory of Drug Target and Screening Research, Beijing 100050, China
| | - Xinyu Yang
- Department of Pharmaceutical Analysis, College of Chemistry and Chemical Engineering, Queen's University Belfast, Belfast BT7 1NN, U.K
| | - Guifen Qiang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College and Beijing Key Laboratory of Drug Target and Screening Research, Beijing 100050, China
| | - Chong Wee Liew
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, 60612 Illinois, United States
| | - Guanhua Du
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College and Beijing Key Laboratory of Drug Target and Screening Research, Beijing 100050, China
| |
Collapse
|
33
|
Abstract
A growing appreciation of the importance of cellular metabolism and revelations concerning the extent of cell-cell heterogeneity demand metabolic characterization of individual cells. We present SpaceM, an open-source method for in situ single-cell metabolomics that detects >100 metabolites from >1,000 individual cells per hour, together with a fluorescence-based readout and retention of morpho-spatial features. We validated SpaceM by predicting the cell types of cocultured human epithelial cells and mouse fibroblasts. We used SpaceM to show that stimulating human hepatocytes with fatty acids leads to the emergence of two coexisting subpopulations outlined by distinct cellular metabolic states. Inducing inflammation with the cytokine interleukin-17A perturbs the balance of these states in a process dependent on NF-κB signaling. The metabolic state markers were reproduced in a murine model of nonalcoholic steatohepatitis. We anticipate SpaceM to be broadly applicable for investigations of diverse cellular models and to democratize single-cell metabolomics.
Collapse
|
34
|
M Onorato A, Fiore E, Bayo J, Casali C, Fernandez-Tomé M, Rodríguez M, Domínguez L, Argemi J, Hidalgo F, Favre C, García M, Atorrasagasti C, Mazzolini GD. SPARC inhibition accelerates NAFLD-associated hepatocellular carcinoma development by dysregulating hepatic lipid metabolism. Liver Int 2021; 41:1677-1693. [PMID: 33641248 DOI: 10.1111/liv.14857] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/27/2021] [Accepted: 02/15/2021] [Indexed: 02/13/2023]
Abstract
BACKGROUND AND AIMS Non-alcoholic fatty liver (NAFLD) and its more serious form non-alcoholic steatohepatitis increase risk of hepatocellular carcinoma (HCC). Lipid metabolic alterations and its role in HCC development remain unclear. SPARC (Secreted Protein, Acidic and Rich in Cysteine) is involved in lipid metabolism, NAFLD and diabetes, but the effects on hepatic lipid metabolism and HCC development is unknown. The aim of this study was to evaluate the role of SPARC in HCC development in the context of NAFLD. METHODS Primary hepatocyte cultures from knockout (SPARC-/- ) or wild-type (SPARC+/+ ) mice, and HepG2 cells were used to assess the effects of free fatty acids on lipid accumulation, expression of lipogenic genes and de novo triglyceride (TG) synthesis. A NAFLD-HCC model was stabilized on SPARC-/- or SPARC+/+ mice. Correlations among SPARC, lipid metabolism-related gene expression patterns and clinical prognosis were studied using HCC gene expression dataset. RESULTS SPARC-/- mice increases hepatic lipid deposits over time. Hepatocytes from SPARC-/- mice or inhibition of SPARC by an antisense adenovirus in HepG2 cells resulted in increased TG deposit, expression of lipid-related genes and nuclear translocation of SREBP1c. Human HCC database analysis revealed that SPARC negatively correlated with genes involved in lipid metabolism, and with poor survival. In NAFLD-HCC murine model, the absence of SPARC accelerates HCC development. RNA-seq study revealed that pathways related to lipid metabolism, cellular detoxification and proliferation were upregulated in SPARC-/- tumour-bearing mice. CONCLUSIONS The absence of SPARC is associated with an altered hepatic lipid metabolism, and an accelerated NAFLD-related HCC development.
Collapse
Affiliation(s)
- Agostina M Onorato
- Gene Therapy Laboratory, Instituto de Investigaciones en Medicina Traslacional, Facultad de Ciencias Biomédicas, CONICET- Universidad Austral, Buenos Aires, Argentina
| | - Esteban Fiore
- Gene Therapy Laboratory, Instituto de Investigaciones en Medicina Traslacional, Facultad de Ciencias Biomédicas, CONICET- Universidad Austral, Buenos Aires, Argentina
| | - Juan Bayo
- Gene Therapy Laboratory, Instituto de Investigaciones en Medicina Traslacional, Facultad de Ciencias Biomédicas, CONICET- Universidad Austral, Buenos Aires, Argentina
| | - Cecilia Casali
- Departamento de Ciencias Biológicas, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Cátedra de Biología Celular y Molecular, Buenos Aires, Argentina.,Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Química y Fisicoquímica Biológicas Prof. Dr. Alejandro C. Paladini (IQUIFIB), Buenos Aires, Argentina
| | - María Fernandez-Tomé
- Departamento de Ciencias Biológicas, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Cátedra de Biología Celular y Molecular, Buenos Aires, Argentina.,Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Química y Fisicoquímica Biológicas Prof. Dr. Alejandro C. Paladini (IQUIFIB), Buenos Aires, Argentina
| | - Marcelo Rodríguez
- Gene Therapy Laboratory, Instituto de Investigaciones en Medicina Traslacional, Facultad de Ciencias Biomédicas, CONICET- Universidad Austral, Buenos Aires, Argentina
| | - Luciana Domínguez
- Gene Therapy Laboratory, Instituto de Investigaciones en Medicina Traslacional, Facultad de Ciencias Biomédicas, CONICET- Universidad Austral, Buenos Aires, Argentina
| | - Josepmaría Argemi
- Josepmaria Argemi, CIMA and Clinica Universidad de Navarra, Pamplona, Spain
| | - Florencia Hidalgo
- Institute of Experimental Physiology, CONICET, School of Biochemical Sciences, University of Rosario, Rosario, Argentina
| | - Cristian Favre
- Institute of Experimental Physiology, CONICET, School of Biochemical Sciences, University of Rosario, Rosario, Argentina
| | - Mariana García
- Gene Therapy Laboratory, Instituto de Investigaciones en Medicina Traslacional, Facultad de Ciencias Biomédicas, CONICET- Universidad Austral, Buenos Aires, Argentina
| | - Catalina Atorrasagasti
- Gene Therapy Laboratory, Instituto de Investigaciones en Medicina Traslacional, Facultad de Ciencias Biomédicas, CONICET- Universidad Austral, Buenos Aires, Argentina
| | - Guillermo D Mazzolini
- Gene Therapy Laboratory, Instituto de Investigaciones en Medicina Traslacional, Facultad de Ciencias Biomédicas, CONICET- Universidad Austral, Buenos Aires, Argentina.,Liver Unit, Hospital Universitario Austral, Universidad Austral, Buenos Aires, Argentina
| |
Collapse
|
35
|
Krause BC, Kriegel FL, Tartz V, Jungnickel H, Reichardt P, Singh AV, Laux P, Shemis M, Luch A. Combinatory Effects of Cerium Dioxide Nanoparticles and Acetaminophen on the Liver-A Case Study of Low-Dose Interactions in Human HuH-7 Cells. Int J Mol Sci 2021; 22:6866. [PMID: 34202329 PMCID: PMC8268126 DOI: 10.3390/ijms22136866] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 06/18/2021] [Accepted: 06/23/2021] [Indexed: 12/12/2022] Open
Abstract
The interactions between pharmaceuticals and nanomaterials and its potentially resulting toxicological effects in living systems are only insufficiently investigated. In this study, two model compounds, acetaminophen, a pharmaceutical, and cerium dioxide, a manufactured nanomaterial, were investigated in combination and individually. Upon inhalation, cerium dioxide nanomaterials were shown to systemically translocate into other organs, such as the liver. Therefore we picked the human liver cell line HuH-7 cells as an in vitro system to investigate liver toxicity. Possible synergistic or antagonistic metabolic changes after co-exposure scenarios were investigated. Toxicological data of the water soluble tetrazolium (WST-1) assay for cell proliferation and genotoxicity assessment using the Comet assay were combined with an untargeted as well as a targeted lipidomics approach. We found an attenuated cytotoxicity and an altered metabolic profile in co-exposure experiments with cerium dioxide, indicating an interaction of both compounds at these endpoints. Single exposure against cerium dioxide showed a genotoxic effect in the Comet assay. Conversely, acetaminophen exhibited no genotoxic effect. Comet assay data do not indicate an enhancement of genotoxicity after co-exposure. The results obtained in this study highlight the advantage of investigating co-exposure scenarios, especially for bioactive substances.
Collapse
Affiliation(s)
- Benjamin C. Krause
- Department of Chemical & Product Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Straße 8-10, 10589 Berlin, Germany; (F.L.K.); (V.T.); (H.J.); (P.R.); (A.V.S.); (P.L.); (A.L.)
| | - Fabian L. Kriegel
- Department of Chemical & Product Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Straße 8-10, 10589 Berlin, Germany; (F.L.K.); (V.T.); (H.J.); (P.R.); (A.V.S.); (P.L.); (A.L.)
- NUVISAN ICB GmbH, Preclinical Compound Profiling, Muellerstrasse 178, 13353 Berlin, Germany
| | - Victoria Tartz
- Department of Chemical & Product Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Straße 8-10, 10589 Berlin, Germany; (F.L.K.); (V.T.); (H.J.); (P.R.); (A.V.S.); (P.L.); (A.L.)
| | - Harald Jungnickel
- Department of Chemical & Product Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Straße 8-10, 10589 Berlin, Germany; (F.L.K.); (V.T.); (H.J.); (P.R.); (A.V.S.); (P.L.); (A.L.)
| | - Philipp Reichardt
- Department of Chemical & Product Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Straße 8-10, 10589 Berlin, Germany; (F.L.K.); (V.T.); (H.J.); (P.R.); (A.V.S.); (P.L.); (A.L.)
| | - Ajay Vikram Singh
- Department of Chemical & Product Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Straße 8-10, 10589 Berlin, Germany; (F.L.K.); (V.T.); (H.J.); (P.R.); (A.V.S.); (P.L.); (A.L.)
| | - Peter Laux
- Department of Chemical & Product Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Straße 8-10, 10589 Berlin, Germany; (F.L.K.); (V.T.); (H.J.); (P.R.); (A.V.S.); (P.L.); (A.L.)
| | - Mohamed Shemis
- Department of Biochemistry & Molecular Biology, Theodor Bilharz Research Institute, Warak El-Hadar, Kornish El-Nile, P.O. Box 30 Imbaba, Giza 12411, Egypt;
| | - Andreas Luch
- Department of Chemical & Product Safety, German Federal Institute for Risk Assessment, Max-Dohrn-Straße 8-10, 10589 Berlin, Germany; (F.L.K.); (V.T.); (H.J.); (P.R.); (A.V.S.); (P.L.); (A.L.)
| |
Collapse
|
36
|
Tsuji T, Morita SY, Nakamura Y, Ikeda Y, Kambe T, Terada T. Alterations in cellular and organellar phospholipid compositions of HepG2 cells during cell growth. Sci Rep 2021; 11:2731. [PMID: 33526799 PMCID: PMC7851136 DOI: 10.1038/s41598-021-81733-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 01/11/2021] [Indexed: 01/30/2023] Open
Abstract
The human hepatoblastoma cell line, HepG2, has been used for investigating a wide variety of physiological and pathophysiological processes. However, less information is available about the phospholipid metabolism in HepG2 cells. In the present report, to clarify the relationship between cell growth and phospholipid metabolism in HepG2 cells, we examined the phospholipid class compositions of the cells and their intracellular organelles by using enzymatic fluorometric methods. In HepG2 cells, the ratios of all phospholipid classes, but not the ratio of cholesterol, markedly changed with cell growth. Of note, depending on cell growth, the phosphatidic acid (PA) ratio increased and phosphatidylcholine (PC) ratio decreased in the nuclear membranes, the sphingomyelin (SM) ratio increased in the microsomal membranes, and the phosphatidylethanolamine (PE) ratio increased and the phosphatidylserine (PS) ratio decreased in the mitochondrial membranes. Moreover, the mRNA expression levels of enzymes related to PC, PE, PS, PA, SM and cardiolipin syntheses changed during cell growth. We suggest that the phospholipid class compositions of organellar membranes are tightly regulated by cell growth. These findings provide a basis for future investigations of cancer cell growth and lipid metabolism.
Collapse
Affiliation(s)
- Tokuji Tsuji
- grid.472014.4Department of Pharmacy, Shiga University of Medical Science Hospital, Otsu, Shiga 520-2192 Japan
| | - Shin-ya Morita
- grid.472014.4Department of Pharmacy, Shiga University of Medical Science Hospital, Otsu, Shiga 520-2192 Japan
| | - Yoshinobu Nakamura
- grid.472014.4Department of Pharmacy, Shiga University of Medical Science Hospital, Otsu, Shiga 520-2192 Japan
| | - Yoshito Ikeda
- grid.472014.4Department of Pharmacy, Shiga University of Medical Science Hospital, Otsu, Shiga 520-2192 Japan
| | - Taiho Kambe
- grid.258799.80000 0004 0372 2033Graduate School of Biostudies, Kyoto University, Kyoto, 606-8502 Japan
| | - Tomohiro Terada
- grid.472014.4Department of Pharmacy, Shiga University of Medical Science Hospital, Otsu, Shiga 520-2192 Japan
| |
Collapse
|
37
|
Soret PA, Magusto J, Housset C, Gautheron J. In Vitro and In Vivo Models of Non-Alcoholic Fatty Liver Disease: A Critical Appraisal. J Clin Med 2020; 10:jcm10010036. [PMID: 33374435 PMCID: PMC7794936 DOI: 10.3390/jcm10010036] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/22/2020] [Accepted: 12/23/2020] [Indexed: 02/07/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD), including non-alcoholic fatty liver (NAFL) and non-alcoholic steatohepatitis (NASH), represents the hepatic manifestation of obesity and metabolic syndrome. Due to the spread of the obesity epidemic, NAFLD is becoming the most common chronic liver disease and one of the principal indications for liver transplantation. However, no pharmacological treatment is currently approved to prevent the outbreak of NASH, which leads to fibrosis and cirrhosis. Preclinical research is required to improve our knowledge of NAFLD physiopathology and to identify new therapeutic targets. In the present review, we summarize advances in NAFLD preclinical models from cellular models, including new bioengineered platforms, to in vivo models, with a particular focus on genetic and dietary mouse models. We aim to discuss the advantages and limits of these different models.
Collapse
Affiliation(s)
- Pierre-Antoine Soret
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, Inserm, 75012 Paris, France; (P.-A.S.); (J.M.); (C.H.)
- Assistance Publique-Hôpitaux de Paris (AP-HP), Hepatology Department, Reference Center for Inflammatory Biliary Diseases and Autoimmune Hepatitis, Saint-Antoine Hospital, 75012 Paris, France
| | - Julie Magusto
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, Inserm, 75012 Paris, France; (P.-A.S.); (J.M.); (C.H.)
- Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne Université, Inserm, AP-HP, 75013 Paris, France
| | - Chantal Housset
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, Inserm, 75012 Paris, France; (P.-A.S.); (J.M.); (C.H.)
- Assistance Publique-Hôpitaux de Paris (AP-HP), Hepatology Department, Reference Center for Inflammatory Biliary Diseases and Autoimmune Hepatitis, Saint-Antoine Hospital, 75012 Paris, France
- Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne Université, Inserm, AP-HP, 75013 Paris, France
| | - Jérémie Gautheron
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, Inserm, 75012 Paris, France; (P.-A.S.); (J.M.); (C.H.)
- Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne Université, Inserm, AP-HP, 75013 Paris, France
- Correspondence:
| |
Collapse
|
38
|
Lim HJ, Kim M. EZH2 as a Potential Target for NAFLD Therapy. Int J Mol Sci 2020; 21:ijms21228617. [PMID: 33207561 PMCID: PMC7697020 DOI: 10.3390/ijms21228617] [Citation(s) in RCA: 15] [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: 10/18/2020] [Revised: 11/11/2020] [Accepted: 11/14/2020] [Indexed: 12/14/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a complex disease that is affected by genetic predisposition and epigenetic modification. Deregulation of epigenetic pathways is now recognized as a frequent event in NAFLD, and understanding the mechanistic roles of these epigenetic factors may lead to new strategies for NAFLD treatment. Enhancer of zeste homolog 2 (EZH2) catalyzes methylation on Lys 27 of histone H3, which leads to chromatin compaction and gene silencing. EZH2 regulates embryonic development and cell lineage determination and is related to many human diseases. Recent studies show that EZH2 has critical roles in liver development, homeostasis, and regeneration. Moreover, aberrant activation of EZH2 promotes NAFLD progression. Several EZH2 inhibitors have been developed and studied both in vitro and in clinical trials. In this review, we summarize our current understanding of the role of EZH2 in NAFLD and highlight its potential as a novel therapeutic target for NAFLD treatment.
Collapse
Affiliation(s)
- Hyun Jung Lim
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea;
- Department of Functional Genomics, University of Science and Technology (UST), Daejeon 34113, Korea
| | - Mirang Kim
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea;
- Department of Functional Genomics, University of Science and Technology (UST), Daejeon 34113, Korea
- Correspondence: ; Tel.: +82-42-879-8113
| |
Collapse
|
39
|
Saito K, Kagawa T, Tsuji K, Kumagai Y, Sato K, Sakisaka S, Sakamoto N, Aiso M, Hirose S, Mori N, Tanaka R, Uraoka T, Takata K, Ogawa K, Mori K, Sato M, Nishiya T, Takamatsu K, Arakawa N, Izumi T, Ohno Y, Saito Y, Takikawa H. Plasma Lipid Profiling of Three Types of Drug-Induced Liver Injury in Japanese Patients: A Preliminary Study. Metabolites 2020; 10:355. [PMID: 32878279 PMCID: PMC7569965 DOI: 10.3390/metabo10090355] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/26/2020] [Accepted: 08/28/2020] [Indexed: 12/13/2022] Open
Abstract
Drug-induced liver injury (DILI) is a major adverse event caused by drug treatment, which can be categorized into three types: hepatocellular, mixed, and cholestatic. Although nearly every class of drugs can cause DILI, an overall understanding of lipid profiles in DILI patients is lacking. We used lipidomics to analyze the plasma lipid profiles of patients to understand their hepatic pathophysiology and identify DILI biomarkers. We identified 463 lipids and compared their levels between the acute and recovery phases of the three types of DILI patients. Mixed and cholestatic types demonstrated specific plasma lipid alterations between the phases, but the hepatocellular type did not. Moreover, as specific indicators of mixed-type DILI, levels of several ceramides increased in the acute phase, while those of arachidonic acid-containing ether-linked phosphoglycerolipids decreased. In contrast, as specific indicators of cholestatic-type DILI, levels of palmitic acid-containing saturated or monounsaturated phosphatidylcholines increased in the acute phase, while those of arachidonic acid- or docosahexaenoic acid-containing ether-linked phosphoglycerolipids and phosphatidylinositols decreased. We also identified lipids with a relatively high capacity to discriminate the acute phase from the recovery phase and healthy subjects. These findings may help with understanding the pathophysiology of different DILI types and identify candidate biomarkers.
Collapse
Grants
- 19mk0101045h0005, 19mk0101045s0105, 19mk0101045s0205, 19mk0101045s0305, 19mk0101045s1205, 19mk0101045s1405, 19mk0101045s1505, 19mk0101045s1605, 19mk0101045j0105, 19mk0101045j0305, 19mk0101045j0405 Japan Agency for Medical Research and Development
Collapse
Affiliation(s)
- Kosuke Saito
- Division of Medical Safety Science, National Institute of Health Sciences, Kanagawa 210-9501, Japan; (K.S.); (N.A.)
| | - Tatehiro Kagawa
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Tokai University School of Medicine, Isehara 259-1193, Japan; (T.K.); (S.H.)
| | - Keiji Tsuji
- Department of Gastroenterology, Hiroshima Red Cross Hospital and Atomic-Bomb Survivors Hospital, Hiroshima 730-8619, Japan; (K.T.); (N.M.)
| | - Yuji Kumagai
- Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa 252-0374, Japan; (Y.K.); (R.T.)
| | - Ken Sato
- Department of Gastroenterology and Hepatology, Gunma University Graduate School of Medicine, Maebashi 371-8511, Japan; (K.S.); (T.U.)
| | - Shotaro Sakisaka
- Department of Gastroenterology, Fukuoka University Faculty of Medicine, Fukuoka 814-0180, Japan; (S.S.); (K.T.)
| | - Naoya Sakamoto
- Department of Gastroenterology and Hepatology, Hokkaido University Faculty of Medicine and Graduate School of Medicine, Sapporo 060-8648, Japan; (N.S.); (K.O.)
| | - Mitsuhiko Aiso
- Department of Medicine, Teikyo University School of Medicine, Tokyo 173-8606, Japan; (M.A.); (H.T.)
| | - Shunji Hirose
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Tokai University School of Medicine, Isehara 259-1193, Japan; (T.K.); (S.H.)
| | - Nami Mori
- Department of Gastroenterology, Hiroshima Red Cross Hospital and Atomic-Bomb Survivors Hospital, Hiroshima 730-8619, Japan; (K.T.); (N.M.)
| | - Rieko Tanaka
- Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa 252-0374, Japan; (Y.K.); (R.T.)
| | - Toshio Uraoka
- Department of Gastroenterology and Hepatology, Gunma University Graduate School of Medicine, Maebashi 371-8511, Japan; (K.S.); (T.U.)
| | - Kazuhide Takata
- Department of Gastroenterology, Fukuoka University Faculty of Medicine, Fukuoka 814-0180, Japan; (S.S.); (K.T.)
| | - Koji Ogawa
- Department of Gastroenterology and Hepatology, Hokkaido University Faculty of Medicine and Graduate School of Medicine, Sapporo 060-8648, Japan; (N.S.); (K.O.)
| | | | - Motonobu Sato
- Astellas Pharma Inc., Tsukuba 305-8585, Japan; (M.S.); (K.T.)
| | | | | | - Noriaki Arakawa
- Division of Medical Safety Science, National Institute of Health Sciences, Kanagawa 210-9501, Japan; (K.S.); (N.A.)
| | - Takashi Izumi
- Kihara Memorial Foundation, Yokohama 230-0045, Japan; (T.I.); (Y.O.)
| | - Yasuo Ohno
- Kihara Memorial Foundation, Yokohama 230-0045, Japan; (T.I.); (Y.O.)
| | - Yoshiro Saito
- Division of Medical Safety Science, National Institute of Health Sciences, Kanagawa 210-9501, Japan; (K.S.); (N.A.)
| | - Hajime Takikawa
- Department of Medicine, Teikyo University School of Medicine, Tokyo 173-8606, Japan; (M.A.); (H.T.)
- Faculty of Medical Technology, Teikyo University, Tokyo 173-8606, Japan
| |
Collapse
|
40
|
Chenopodium Quinoa and Salvia Hispanica Provide Immunonutritional Agonists to Ameliorate Hepatocarcinoma Severity under a High-Fat Diet. Nutrients 2020; 12:nu12071946. [PMID: 32629893 PMCID: PMC7400258 DOI: 10.3390/nu12071946] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 06/24/2020] [Accepted: 06/26/2020] [Indexed: 02/06/2023] Open
Abstract
Complex interactions between immunonutritional agonist and high fat intake (HFD), the immune system and finally gut microbiota are important determinants of hepatocarcinoma (HCC) severity. The ability of immunonutritional agonists to modulate major aspects such as liver innate immunity and inflammation and alterations in major lipids profile as well as gut microbiota during HCC development is poorly understood. 1H NMR has been employed to assess imbalances in saturated fatty acids, MUFA and PUFA, which were associated to variations in iron homeostasis. These effects were dependent on the botanical nature (Chenopodium quinoa vs. Salvia hispanica L.) of the compounds. The results showed that immunonutritional agonists' promoted resistance to hepatocarcinogenesis under pro-tumorigenic inflammation reflected, at a different extent, in increased proportions of F4/80+ cells in injured livers as well as positive trends of accumulated immune mediators (CD68/CD206 ratio) in intestinal tissue. Administration of all immunonutritional agonists caused similar variations of fecal microbiota, towards a lower obesity-inducing potential than animals only fed a HFD. Modulation of Firmicutes to Bacteroidetes contents restored the induction of microbial metabolites to improve epithelial barrier function, showing an association with liver saturated fatty acids and the MUFA and PUFA fractions. Collectively, these data provide novel findings supporting beneficial immunometabolic effects targeting hepatocarcinogenesis, influencing innate immunity within the gut-liver axis, and providing novel insights into their immunomodulatory activity.
Collapse
|
41
|
Lee S, Woo DC, Kang J, Ra M, Kim KH, Lee SR, Choi DK, Lee H, Hong KB, Min SH, Lee Y, Yu JH. The Role of the Histone Methyltransferase EZH2 in Liver Inflammation and Fibrosis in STAM NASH Mice. BIOLOGY 2020; 9:biology9050093. [PMID: 32370249 PMCID: PMC7285133 DOI: 10.3390/biology9050093] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 04/24/2020] [Accepted: 04/28/2020] [Indexed: 02/06/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a leading form of chronic liver disease, with few biomarkers and treatment options currently available. Non-alcoholic steatohepatitis (NASH), a progressive disease of NAFLD, may lead to fibrosis, cirrhosis, and hepatocellular carcinoma. Epigenetic modification can contribute to the progression of NAFLD causing non-alcoholic steatohepatitis (NASH), in which the exact role of epigenetics remains poorly understood. To identify potential therapeutics for NASH, we tested small-molecule inhibitors of the epigenetic target histone methyltransferase EZH2, Tazemetostat (EPZ-6438), and UNC1999 in STAM NASH mice. The results demonstrate that treatment with EZH2 inhibitors decreased serum TNF-alpha in NASH. In this study, we investigated that inhibition of EZH2 reduced mRNA expression of inflammatory cytokines and fibrosis markers in NASH mice. In conclusion, these results suggest that EZH2 may present a promising therapeutic target in the treatment of NASH.
Collapse
Affiliation(s)
- Seul Lee
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Korea; (S.L.); (D.K.C.); (H.L.); (K.B.H.); (S.-H.M.)
| | - Dong-Cheol Woo
- Convergence medicine research center, Asan Institute for Life Sciences, Asan Medical Center, and Department of Convergence Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea;
| | - Jeeheon Kang
- Center for Bio-Imaging of New Drug Development, Asan Life Science Institution, Asan Medical Centre, Seoul 05505, Korea;
| | - Moonjin Ra
- Hongcheon Institute of Medicinal Herb, 101 Yeonbongri, Hongcheon 25142, Korea;
| | - Ki Hyun Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 440-746, Korea; (K.H.K.); (S.R.L.)
| | - Seoung Rak Lee
- School of Pharmacy, Sungkyunkwan University, Suwon 440-746, Korea; (K.H.K.); (S.R.L.)
| | - Dong Kyu Choi
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Korea; (S.L.); (D.K.C.); (H.L.); (K.B.H.); (S.-H.M.)
| | - Heejin Lee
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Korea; (S.L.); (D.K.C.); (H.L.); (K.B.H.); (S.-H.M.)
- School of Life Sciences and Biotechnology, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Korea
| | - Ki Bum Hong
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Korea; (S.L.); (D.K.C.); (H.L.); (K.B.H.); (S.-H.M.)
| | - Sang-Hyun Min
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Korea; (S.L.); (D.K.C.); (H.L.); (K.B.H.); (S.-H.M.)
| | - Yongjun Lee
- Hongcheon Institute of Medicinal Herb, 101 Yeonbongri, Hongcheon 25142, Korea;
- Correspondence: (Y.L.); (J.H.Y.); Tel.: +82-33-439-3248 (Y.L.); +82-53-790-5232 (J.H.Y.); Fax: 82-33-439-3239 (Y.L.); 82-53-790-5219 (J.H.Y.)
| | - Ji Hoon Yu
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Korea; (S.L.); (D.K.C.); (H.L.); (K.B.H.); (S.-H.M.)
- Correspondence: (Y.L.); (J.H.Y.); Tel.: +82-33-439-3248 (Y.L.); +82-53-790-5232 (J.H.Y.); Fax: 82-33-439-3239 (Y.L.); 82-53-790-5219 (J.H.Y.)
| |
Collapse
|
42
|
A Pilot Study of Serum Sphingomyelin Dynamics in Subjects with Severe Obesity and Non-alcoholic Steatohepatitis after Sleeve Gastrectomy. Obes Surg 2020; 29:983-989. [PMID: 30488259 DOI: 10.1007/s11695-018-3612-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Non-alcoholic steatohepatitis (NASH) is present in a high percentage of obese patients undergoing bariatric surgery. A significant proportion of patients still present NASH even after considerable weight loss and metabolic improvements after surgery. OBJECTIVE To determine whether the changes in the serum lipidome after sleeve gastrectomy could be used to discriminate obese patients with NASH patients to those with non-alcoholic fatty liver (NAFL). METHODS This study involved 24 patients with grade 3 obesity diagnosed with either NAFL (n = 8) or NASH (n = 16) using the non-invasive OWLiver assay. All patients suffering from NASH were re-evaluated 6 months after bariatric surgery using the OWLiver test to confirm NASH resolution. Serum lipid extracts were assessed at baseline and 6 months post surgery and were analyzed in an ultra-performance liquid chromatography/time-of-flight mass spectrometry (UPLC-TOF-MS)-based platform. RESULTS Lipidomic analysis revealed a differential sphingomyelin profile in patients with NASH resolution after sleeve gastrectomy. Certain serum sphingomyelin species were significantly higher at baseline in NASH patients in comparison to those with NAFL. Sphingomyelin profile of subjects with NASH resolution was similar to that for obese subjects with NAFL before bariatric surgery. CONCLUSION Our study indicates that the serum sphingomyelin levels could be related to the status of non-alcoholic fatty liver disease and that certain sphingomyelin species may be used for the follow-up of obese patients with NASH after sleeve gastrectomy.
Collapse
|
43
|
Funai K, Summers SA, Rutter J. Reign in the membrane: How common lipids govern mitochondrial function. Curr Opin Cell Biol 2020; 63:162-173. [PMID: 32106003 DOI: 10.1016/j.ceb.2020.01.006] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 12/09/2019] [Accepted: 01/07/2020] [Indexed: 12/12/2022]
Abstract
The lipids that make up biological membranes tend to be the forgotten molecules of cell biology. The paucity of data on these important entities likely reflects the difficulties of studying and understanding their biological roles, rather than revealing a lack of importance. Indeed, the lipid composition of biological membranes has a profound impact on a diverse array of cellular processes. The focus of this review is on the effects of different lipid classes on the function of mitochondria, particularly bioenergetics, in health and disease.
Collapse
Affiliation(s)
- Katsuhiko Funai
- Diabetes & Metabolism Research Center, University of Utah, Salt Lake City, UT, USA; Department of Physical Therapy & Athletic Training, University of Utah, Salt Lake City, UT, USA; Department of Nutrition & Integrative Physiology, University of Utah, Salt Lake City, UT, USA.
| | - Scott A Summers
- Diabetes & Metabolism Research Center, University of Utah, Salt Lake City, UT, USA; Department of Nutrition & Integrative Physiology, University of Utah, Salt Lake City, UT, USA; Department of Biochemistry, University of Utah, Salt Lake City, UT, USA.
| | - Jared Rutter
- Diabetes & Metabolism Research Center, University of Utah, Salt Lake City, UT, USA; Department of Nutrition & Integrative Physiology, University of Utah, Salt Lake City, UT, USA; Department of Biochemistry, University of Utah, Salt Lake City, UT, USA; Howard Hughes Medical Institute, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
44
|
Han L, Bittner S, Dong D, Cortez Y, Dulay H, Arshad S, Shen WJ, Kraemer FB, Azhar S. Creosote bush-derived NDGA attenuates molecular and pathological changes in a novel mouse model of non-alcoholic steatohepatitis (NASH). Mol Cell Endocrinol 2019; 498:110538. [PMID: 31415794 PMCID: PMC7273809 DOI: 10.1016/j.mce.2019.110538] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 07/26/2019] [Accepted: 08/11/2019] [Indexed: 02/06/2023]
Abstract
Creosote bush (Larrea tridentata)-derived nordihydroguaiaretic acid (NDGA) was shown to have profound effects on the core components of metabolic syndrome. This study investigated the in vivo potential of NDGA for prevention or attenuation of the pathophysiologic abnormalities of NASH. A novel dietary NASH model with feeding C57BL/6J mice with a high trans-fat, high cholesterol and high fructose (HTF) diet, was used. The HTF diet fed mice exhibited obesity, insulin resistance, hepatic steatosis, fibrosis, inflammation, ER stress, oxidative stress, and liver injury. NDGA attenuated these metabolic abnormalities as well as hepatic steatosis and fibrosis together with attenuated expression of genes encoding fibrosis, progenitor and macrophage markers with no effect on the levels of mRNAs for lipogenic enzymes. NDGA increased expression of fatty acid oxidation genes. In conclusion, NDGA exerts anti-NASH/anti-fibrotic actions and raises the therapeutic potential of NDGA for treatment of NASH patients with fibrosis and other associated complications.
Collapse
Affiliation(s)
- Lu Han
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, CA, USA; Division of Endocrinology, Gerontology and Metabolism, Stanford University, Stanford, CA, USA
| | - Stefanie Bittner
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, CA, USA
| | - Dachuan Dong
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, CA, USA; Division of Endocrinology, Gerontology and Metabolism, Stanford University, Stanford, CA, USA
| | - Yuan Cortez
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, CA, USA
| | - Hunter Dulay
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, CA, USA
| | - Sara Arshad
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, CA, USA; Division of Endocrinology, Gerontology and Metabolism, Stanford University, Stanford, CA, USA
| | - Wen-Jun Shen
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, CA, USA; Division of Endocrinology, Gerontology and Metabolism, Stanford University, Stanford, CA, USA.
| | - Fredric B Kraemer
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, CA, USA; Division of Endocrinology, Gerontology and Metabolism, Stanford University, Stanford, CA, USA; Stanford Diabetes Research Center, USA
| | - Salman Azhar
- Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, CA, USA; Division of Endocrinology, Gerontology and Metabolism, Stanford University, Stanford, CA, USA; Stanford Diabetes Research Center, USA.
| |
Collapse
|
45
|
Liebig M, Dannenberger D, Vollmar B, Abshagen K. n-3 PUFAs reduce tumor load and improve survival in a NASH-tumor mouse model. Ther Adv Chronic Dis 2019; 10:2040622319872118. [PMID: 31523414 PMCID: PMC6728677 DOI: 10.1177/2040622319872118] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 07/16/2019] [Indexed: 12/22/2022] Open
Abstract
Background With 9.1% of all cancer deaths, hepatocellular carcinoma is the second leading cause of cancer deaths worldwide. Due to the increasing prevalence of metabolic syndrome, nonalcoholic fatty liver disease (NAFLD) has evolved into a major risk factor for hepatocellular carcinoma development. Herein, we investigated whether a dietary n-3 polyunsaturated fatty acid (PUFA) supplementation improves the outcome of progressive NAFLD. Methods Feeding three high-fat diets, differing in n-3 and n-6 PUFA contents and ratios (n-3/n-6: 1:8, 1:1, 5:1), the impact of n-3 PUFAs and n-3/n-6 PUFA ratios on NAFLD-related liver fibrosis and tumorigenesis was analyzed in 12- and 20-week-old streptozotocin/high-fat diet (STZ/HFD)-treated mice. Results Feeding of n-3 PUFA-rich diets (1:1 and 5:1) resulted in increased hepatic n-3 PUFA content and n-3/n-6 PUFA ratio with decreased hepatic lipid accumulation. In 20-week-old mice, n-3 PUFA-rich diets alleviated tumor load significantly, with reduced liver/body weight index, tumor size, and tumor number. Finally, these effects were accompanied by a significant improvement of survival of these mice. Conclusions Herein, we showed that increased n-3 PUFA content and n-3/n-6 PUFA ratios lead to improved survival and attenuated tumor progression in STZ/HFD-treated mice. Thus, n-3 PUFAs could be the basis for new therapeutic options against NAFLD-related tumorigenesis.
Collapse
Affiliation(s)
- Marie Liebig
- Institute for Experimental Surgery, University Medicine Rostock, Germany
| | - Dirk Dannenberger
- Institute of Muscle Biology and Growth, Leibniz Institute for Farm Animal Biology, Dummerstorf, Germany
| | - Brigitte Vollmar
- Institute for Experimental Surgery, University Medicine Rostock, Germany
| | - Kerstin Abshagen
- Institute for Experimental Surgery, University Medicine Rostock, Germany
| |
Collapse
|
46
|
Iida A, Kuranuki S, Yamamoto R, Uchida M, Ohta M, Ichimura M, Tsuneyama K, Masaki T, Seike M, Nakamura T. Analysis of amino acid profiles of blood over time and biomarkers associated with non-alcoholic steatohepatitis in STAM mice. Exp Anim 2019; 68:417-428. [PMID: 31155606 PMCID: PMC6842803 DOI: 10.1538/expanim.18-0152] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The changes in free amino acid (AA) levels in blood during the progression from
non-alcoholic steatohepatitis (NASH) to hepatocellular carcinoma (HCC) are unclear. We
investigated serum AA levels, along with biochemical and histological events, in a mouse
model of NASH. We induced NASH in male C57BL/6J mice with a streptozotocin injection and
high-fat diet after 4 weeks of age (STAM group). We chronologically (6, 8, 10, 12, and 16
weeks, n=4–12 mice/group) evaluated the progression from steatohepatitis to HCC by
biochemical and histological analyses. The serum AA levels were determined using an AA
analyzer. Serum aspartate aminotransferase and alanine aminotransferase levels were higher
in the STAM group than in the normal group (non-NASH-induced mice). Histological analysis
revealed that STAM mice had fatty liver, NASH, and fibrosis at 6, 8, and 10 weeks,
respectively. Moreover, the mice exhibited fibrosis and HCC at 16 weeks. The serum
branched-chain AA levels were higher in the STAM group than in the normal group,
especially at 8 and 10 weeks. The Fischer ratio decreased at 16 weeks in the STAM group,
with increasing aromatic AA levels. These results suggested that this model sequentially
depicts the development of fatty liver, NASH, cirrhosis, HCC, and AA metabolism disorders
within a short experimental period. Additionally, serum amyloid A was suggested to be a
useful inflammation biomarker associated with NASH. We believe that the STAM model will be
useful for studying AA metabolism and/or pharmacological effects in NASH.
Collapse
Affiliation(s)
- Ayaka Iida
- School of Nutrition and Dietetics, Faculty of Health and Social Services, Kanagawa University of Human Services, 1-10-1 Heisei-cho, Yokosuka, Kanagawa 238-8522, Japan.,Graduate School of Health and Environmental Sciences, Fukuoka Women's University, 1-1-1 Kasumigaoka, Higashi-ku, Fukuoka 813-8529, Japan
| | - Sachi Kuranuki
- School of Nutrition and Dietetics, Faculty of Health and Social Services, Kanagawa University of Human Services, 1-10-1 Heisei-cho, Yokosuka, Kanagawa 238-8522, Japan
| | - Ryoko Yamamoto
- Department of Applied Biology and Food Sciences, Faculty of Agriculture and Life Science, Hirosaki University, 3 Bunkyo-cho, Hirosaki, Aomori 036-8560, Japan
| | - Masaya Uchida
- Department of Creative Engineering, National Institute of Technology, Ariake College, 150 Higashi hagio-machi, Omuta, Fukuoka 836-8585, Japan
| | - Masanori Ohta
- Graduate School of Health and Environmental Sciences, Fukuoka Women's University, 1-1-1 Kasumigaoka, Higashi-ku, Fukuoka 813-8529, Japan
| | - Mayuko Ichimura
- Department of Pathology and Laboratory Medicine, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Koichi Tsuneyama
- Department of Pathology and Laboratory Medicine, Institute of Biomedical Sciences, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Takayuki Masaki
- Department of Endocrinology, Metabolism, Rheumatology and Nephrology, Faculty of Medicine, Oita University, 1-1 Idaigaoka, Hasama-machi, Yufu, Oita 879-5593, Japan
| | - Masataka Seike
- Department of Gastroenterology, Faculty of Medicine, Oita University, 1-1 Idaigaoka, Hasama-machi, Yufu, Oita 879-5593, Japan
| | - Tsuyoshi Nakamura
- Graduate School of Health and Environmental Sciences, Fukuoka Women's University, 1-1-1 Kasumigaoka, Higashi-ku, Fukuoka 813-8529, Japan
| |
Collapse
|
47
|
Sun G, Jackson CV, Zimmerman K, Zhang LK, Finnearty CM, Sandusky GE, Zhang G, Peterson RG, Wang YXJ. The FATZO mouse, a next generation model of type 2 diabetes, develops NAFLD and NASH when fed a Western diet supplemented with fructose. BMC Gastroenterol 2019; 19:41. [PMID: 30885145 PMCID: PMC6421686 DOI: 10.1186/s12876-019-0958-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 02/27/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Metabolic disorders such as insulin resistance, obesity, and hyperglycemia are prominent risk factors for the development of non-alcoholic fatty liver disease (NAFLD)/steatohepatitis (NASH). Dietary rodent models employ high fat, high cholesterol, high fructose, methionine/choline deficient diets or combinations of these to induce NAFLD/NASH. The FATZO mice spontaneously develop the above metabolic disorders and type 2 diabetes (T2D) when fed with a normal chow diet. The aim of the present study was to determine if FATZO mice fed a high fat and fructose diet would exacerbate the progression of NAFLD/NASH. METHODS Male FATZO mice at the age of 8 weeks were fed with high fat Western diet (D12079B) supplemented with 5% fructose in the drinking water (WDF) for the duration of 20 weeks. The body weight, whole body fat content, serum lipid profiles and liver function markers were examined monthly along with the assessment of liver histology for the development of NASH. In addition, the effects of obeticholic acid (OCA, 30 mg/kg, QD) on improvement of NASH progression in the model were evaluated. RESULTS Compared to normal control diet (CD), FATZO mice fed with WDF were heavier with higher body fat measured by qNMR, hypercholesterolemia and had progressive elevations in AST (~ 6 fold), ALT (~ 6 fold), liver over body weight (~ 2 fold) and liver triglyceride (TG) content (1.4-2.9 fold). Histological examination displayed evidence of NAFLD/NASH, including hepatic steatosis, lobular inflammation, ballooning and fibrosis in FATZO mice fed WDF. Treatment with OCA for 15 weeks in FATZO mice on WDF significantly alleviated hypercholesterolemia and elevation of AST/ALT, reduced liver weight and liver TG contents, attenuated hepatic ballooning, but did not affect body weight and blood TG levels. CONCLUSION WDF fed FATZO mice represent a new model for the study of progressive NAFLD/NASH with concurrent metabolic dysregulation.
Collapse
Affiliation(s)
- Gao Sun
- Crown Bioscience Taicang Inc, Taicang, China
| | | | | | | | - Courtney M Finnearty
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - George E Sandusky
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | | | | | |
Collapse
|
48
|
Otto AC, Gan-Schreier H, Zhu X, Tuma-Kellner S, Staffer S, Ganzha A, Liebisch G, Chamulitrat W. Group VIA phospholipase A2 deficiency in mice chronically fed with high-fat-diet attenuates hepatic steatosis by correcting a defect of phospholipid remodeling. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1864:662-676. [PMID: 30735855 DOI: 10.1016/j.bbalip.2019.01.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 01/22/2019] [Accepted: 01/25/2019] [Indexed: 12/15/2022]
Abstract
A defect of hepatic remodeling of phospholipids (PL) is seen in non-alcoholic fatty liver disease and steatohepatitis (NASH) indicating pivotal role of PL metabolism in this disease. The deletion of group VIA calcium-independent phospholipase A2 (iPla2β) protects ob/ob mice from hepatic steatosis (BBAlip 1861, 2016, 440-461), however its role in high-fat diet (HFD)-induced NASH is still elusive. Here, wild-type and iPla2β-null mice were subjected to chronic feeding with HFD for 6 months. We showed that protection was observed in iPla2β-null mice with an attenuation of diet-induced body and liver-weight gains, liver enzymes, serum free fatty acids as well as hepatic TG and steatosis scores. iPla2β deficiency under HFD attenuated the levels of 1-stearoyl lysophosphatidylcholine (LPC), lysophosphatidylethanolamine (LPE), and lysophosphatidylinositol (LPI) as well as elevation of hepatic arachidonate, arachidonate-containing cholesterol esters and prostaglandin E2. More importantly, this deficiency rescued a defect in PL remodeling and attenuated the ratio of saturated and unsaturated PL. The protection by iPla2β deficiency was not observed during short-term HFD feeding of 3 or 5 weeks which showed no PL remodeling defect. In addition to PC/PE, this deficiency reversed the suppression of PC/PI and PE/PI among monounsaturated PL. However, this deficiency did not modulate hepatic PL contents and PL ratios in ER fractions, ER stress, fibrosis, and inflammation markers. Hence, iPla2β inactivation protected mice against hepatic steatosis and obesity during chronic dietary NASH by correcting PL remodeling defect and PI composition relative to PC and PE.
Collapse
Affiliation(s)
- Ann-Christin Otto
- Department of Internal Medicine IV, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Hongying Gan-Schreier
- Department of Internal Medicine IV, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Xingya Zhu
- Department of Internal Medicine IV, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Sabine Tuma-Kellner
- Department of Internal Medicine IV, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Simone Staffer
- Department of Internal Medicine IV, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Alexandra Ganzha
- Institute of Clinical Chemistry and Laboratory Medicine, University Regensburg, Franz-Josef-Strauss-Allee 11, 93053 Regensburg, Germany
| | - Gerhard Liebisch
- Institute of Clinical Chemistry and Laboratory Medicine, University Regensburg, Franz-Josef-Strauss-Allee 11, 93053 Regensburg, Germany
| | - Walee Chamulitrat
- Department of Internal Medicine IV, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany.
| |
Collapse
|
49
|
Abstract
Hepatocellular carcinoma (HCC) is one of the most fatal and fastest-growing cancers. Recently, non-alcoholic steatohepatitis (NASH) has been recognized as a major HCC catalyst. However, it is difficult to decipher the molecular mechanisms underlying the pathogenesis of NASH and understand how it progresses to HCC by studying humans. Progress in this field depends on the availability of reliable preclinical models amenable to genetic and functional analyses and exhibiting robust NASH-to-HCC progression. Although numerous mouse models of NASH have been described, many do not faithfully mimic the human disease and few reliably progress to HCC. Here, we review current literature on the molecular etiology of NASH-related HCC and critically evaluate existing mouse models and their suitability for studying this malignancy. We also compare human transcriptomic and histopathological profiles with data from MUP-uPA mice, a reliable model of NASH-driven HCC that has been useful for evaluation of HCC-targeting immunotherapies.
Collapse
|
50
|
Nakashima A, Sugimoto R, Suzuki K, Shirakata Y, Hashiguchi T, Yoshida C, Nakano Y. Anti-fibrotic activity of Euglena gracilis and paramylon in a mouse model of non-alcoholic steatohepatitis. Food Sci Nutr 2019; 7:139-147. [PMID: 30680167 PMCID: PMC6341149 DOI: 10.1002/fsn3.828] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 09/10/2018] [Accepted: 09/13/2018] [Indexed: 12/18/2022] Open
Abstract
Progression to non-alcoholic steatohepatitis (NASH) manifests as hepatitis, fibrosis, and sometimes carcinoma, resulting in liver failure. Various clinical trials have indicated that several pharmacological agents, including angiotensin II receptor blockers (ARBs) or farnesoid X receptor (FXR) agonists, are effective in NASH treatment. In addition, functional foods are expected to be important alternatives for treating or preventing NASH. Recently, focus has been directed toward microalgae as dietary supplements, mainly for lifestyle-related diseases, because they contain various nutrients and functional ingredients. Specifically, a unicellular microalga Euglena gracilis stores a unique β-1,3-glucan particle called paramylon that stimulates the immune system. In this study, we evaluated the effects of Euglena and paramylon on NASH in Stelic Animal Model (STAM) mice using Sirius red staining and confirmed that oral administration of Euglena or paramylon inhibits the process of liver fibrosis. Moreover, compared with controls, paramylon decreased non-alcoholic fatty liver disease (NAFLD) activity scores related to inflammation. These results indicate that the oral administration of Euglena and paramylon inhibits fibrosis and ameliorates NASH.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yoshihisa Nakano
- Center for Research and Development of BioresourcesOsaka Prefecture UniversityOsakaJapan
| |
Collapse
|