1
|
Yang Z, Wang J, Zhao T, Wang L, Liang T, Zheng Y. Mitochondrial structure and function: A new direction for the targeted treatment of chronic liver disease with Chinese herbal medicine. JOURNAL OF ETHNOPHARMACOLOGY 2024; 334:118461. [PMID: 38908494 DOI: 10.1016/j.jep.2024.118461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 06/10/2024] [Accepted: 06/13/2024] [Indexed: 06/24/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Excessive fat accumulation, biological clock dysregulation, viral infections, and sustained inflammatory responses can lead to liver inflammation, fibrosis, and cancer, thus promoting the development of chronic liver disease. A comprehensive understanding of the etiological factors leading to chronic liver disease and the intrinsic mechanisms influencing its onset and progression can aid in identifying potential targets for targeted therapy. Mitochondria, as key organelles that maintain the metabolic homeostasis of the liver, provide an important foundation for exploring therapeutic targets for chronic liver disease. Recent studies have shown that active ingredients in herbal medicines and their natural products can modulate chronic liver disease by influencing the structure and function of mitochondria. Therefore, studying how Chinese herbs target mitochondrial structure and function to treat chronic liver diseases is of great significance. AIM OF THE STUDY Investigating the prospects of herbal medicine the Lens of chronic liver disease based on mitochondrial structure and function. MATERIALS AND METHODS A computerized search of PubMed was conducted using the keywords "mitochondrial structure", "mitochondrial function", "mitochondria and chronic liver disease", "botanicals, mitochondria and chronic liver disease".Data from the Web of Science and Science Direct databases were also included. The research findings regarding herbal medicines targeting mitochondrial structure and function for the treatment of chronic liver disease are summarized. RESULTS A computerized search of PubMed using the keywords "mitochondrial structure", "mitochondrial function", "mitochondria and chronic liver disease", "phytopharmaceuticals, mitochondria, and chronic liver disease", as well as the Web of Science and Science Direct databases was conducted to summarize information on studies of mitochondrial structure- and function-based Chinese herbal medicines for the treatment of chronic liver disease and to suggest that the effects of herbal medicines on mitochondrial division and fusion.The study suggested that there is much room for research on the influence of Chinese herbs on mitochondrial division and fusion. CONCLUSIONS Targeting mitochondrial structure and function is crucial for herbal medicine to combat chronic liver disease.
Collapse
Affiliation(s)
- Zhihui Yang
- Department of Medicine, Faculty of Chinese Medicine Science Guangxi University of Chinese Medicine, Nanning, Guangxi, 530222, China
| | - Jiahui Wang
- Department of Medicine, Faculty of Chinese Medicine Science Guangxi University of Chinese Medicine, Nanning, Guangxi, 530222, China
| | - Tiejian Zhao
- Department of Medicine, Faculty of Chinese Medicine Science Guangxi University of Chinese Medicine, Nanning, Guangxi, 530222, China
| | - Lei Wang
- Department of Medicine, Faculty of Chinese Medicine Science Guangxi University of Chinese Medicine, Nanning, Guangxi, 530222, China
| | - Tianjian Liang
- Department of Medicine, Faculty of Chinese Medicine Science Guangxi University of Chinese Medicine, Nanning, Guangxi, 530222, China.
| | - Yang Zheng
- Department of Medicine, Faculty of Chinese Medicine Science Guangxi University of Chinese Medicine, Nanning, Guangxi, 530222, China.
| |
Collapse
|
2
|
Kotulkar M, Barbee J, Paine-Cabrera D, Robarts D, O'Neil M, Apte U. Role of HNF4α-cMyc Interaction in CDE Diet-Induced Liver Injury and Regeneration. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:1218-1229. [PMID: 38588852 PMCID: PMC11317903 DOI: 10.1016/j.ajpath.2024.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 02/27/2024] [Accepted: 03/15/2024] [Indexed: 04/10/2024]
Abstract
Hepatocyte nuclear factor 4 alpha (HNF4α) is a nuclear factor essential for liver function that regulates the expression of cMyc and plays an important role during liver regeneration. This study investigated the role of the HNF4α-cMyc interaction in regulating liver injury and regeneration using the choline-deficient and ethionine-supplemented (CDE) diet model. Wild-type (WT), hepatocyte-specific HNF4α-knockout (KO), cMyc-KO, and HNF4α-cMyc double KO (DKO) mice were fed a CDE diet for 1 week to induce subacute liver injury. To study regeneration, normal chow diet was fed for 1 week after CDE diet. WT mice exhibited significant liver injury and decreased HNF4α mRNA and protein expression after CDE diet. HNF4α deletion resulted in significantly higher injury with increased inflammation, fibrosis, proliferation, and hepatic progenitor cell activation compared with WT mice after CDE diet but indicated similar recovery. Deletion of cMyc lowered liver injury with activation of inflammatory genes compared with WT and HNF4α-KO mice after CDE diet. DKO mice had a phenotype comparable to that of the HNF4α-KO mice after CDE diet and a complete recovery. DKO mice exhibited a significant increase in hepatic progenitor cell markers both after injury and recovery phase. Taken together, these data show that HNF4α protects against inflammatory and fibrotic changes after CDE diet-induced injury, which is driven by cMyc.
Collapse
Affiliation(s)
- Manasi Kotulkar
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Julia Barbee
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Diego Paine-Cabrera
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Dakota Robarts
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Maura O'Neil
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Udayan Apte
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas.
| |
Collapse
|
3
|
Yang M, Wang D, Wang X, Mei J, Gong Q. Role of Folate in Liver Diseases. Nutrients 2024; 16:1872. [PMID: 38931227 PMCID: PMC11206401 DOI: 10.3390/nu16121872] [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: 05/06/2024] [Revised: 06/06/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
Folate is a water-soluble B vitamin involved in the synthesis of purines and pyrimidines and is one of the essential vitamins for human growth and reproduction. Folate deficiency due to low dietary intake, poor absorption of folate, and alterations in folate metabolism due to genetic defects or drug interactions significantly increases the risk of diseases such as neural tube defects, cardiovascular disease, cancer, and cognitive dysfunction. Recent studies have shown that folate deficiency can cause hyperhomocysteinemia, which increases the risk of hypertension and cardiovascular disease, and that high homocysteine levels are an independent risk factor for liver fibrosis and cirrhosis. In addition, folate deficiency results in increased secretion of pro-inflammatory factors and impaired lipid metabolism in the liver, leading to lipid accumulation in hepatocytes and fibrosis. There is substantial evidence that folate deficiency contributes to the development and progression of a variety of liver diseases, including non-alcoholic fatty liver disease (NAFLD), non-alcoholic steatohepatitis (NASH), alcoholic liver disease (ALD), viral hepatitis, hepatic fibrosis, and liver cancer. Here we review key studies on the role of folate in the pathophysiology of liver diseases, summarize the current status of studies on folate in the treatment of liver diseases, and speculate that folate may be a potential therapeutic target for liver diseases.
Collapse
Affiliation(s)
- Minlan Yang
- School of Medicine, Yangtze University, Jingzhou 434020, China
| | | | | | | | - Quan Gong
- School of Medicine, Yangtze University, Jingzhou 434020, China
| |
Collapse
|
4
|
Zacarías-Fluck MF, Soucek L, Whitfield JR. MYC: there is more to it than cancer. Front Cell Dev Biol 2024; 12:1342872. [PMID: 38510176 PMCID: PMC10952043 DOI: 10.3389/fcell.2024.1342872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 02/20/2024] [Indexed: 03/22/2024] Open
Abstract
MYC is a pleiotropic transcription factor involved in multiple cellular processes. While its mechanism of action and targets are not completely elucidated, it has a fundamental role in cellular proliferation, differentiation, metabolism, ribogenesis, and bone and vascular development. Over 4 decades of research and some 10,000 publications linking it to tumorigenesis (by searching PubMed for "MYC oncogene") have led to MYC becoming a most-wanted target for the treatment of cancer, where many of MYC's physiological functions become co-opted for tumour initiation and maintenance. In this context, an abundance of reviews describes strategies for potentially targeting MYC in the oncology field. However, its multiple roles in different aspects of cellular biology suggest that it may also play a role in many additional diseases, and other publications are indeed linking MYC to pathologies beyond cancer. Here, we review these physiological functions and the current literature linking MYC to non-oncological diseases. The intense efforts towards developing MYC inhibitors as a cancer therapy will potentially have huge implications for the treatment of other diseases. In addition, with a complementary approach, we discuss some diseases and conditions where MYC appears to play a protective role and hence its increased expression or activation could be therapeutic.
Collapse
Affiliation(s)
- Mariano F. Zacarías-Fluck
- Models of Cancer Therapies Laboratory, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Laura Soucek
- Models of Cancer Therapies Laboratory, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
- Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- Peptomyc S.L., Barcelona, Spain
| | - Jonathan R. Whitfield
- Models of Cancer Therapies Laboratory, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| |
Collapse
|
5
|
Nevzorova YA, Cubero FJ. Obesity under the moonlight of c-MYC. Front Cell Dev Biol 2023; 11:1293218. [PMID: 38116204 PMCID: PMC10728299 DOI: 10.3389/fcell.2023.1293218] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 11/07/2023] [Indexed: 12/21/2023] Open
Abstract
The moonlighting protein c-Myc is a master regulator of multiple biological processes including cell proliferation, differentiation, angiogenesis, apoptosis and metabolism. It is constitutively and aberrantly expressed in more than 70% of human cancers. Overwhelming evidence suggests that c-Myc dysregulation is involved in several inflammatory, autoimmune, metabolic and other non-cancerous diseases. In this review, we addressed the role of c-Myc in obesity. Obesity is a systemic disease, accompanied by multi-organ dysfunction apart from white adipose tissue (WAT), such as the liver, the pancreas, and the intestine. c-Myc plays a big diversity of functions regulating cellular proliferation, the maturation of progenitor cells, fatty acids (FAs) metabolism, and extracellular matrix (ECM) remodeling. Moreover, c-Myc drives the expression of a wide range of metabolic genes, modulates the inflammatory response, induces insulin resistance (IR), and contributes to the regulation of intestinal dysbiosis. Altogether, c-Myc is an interesting diagnostic tool and/or therapeutic target in order to mitigate obesity and its consequences.
Collapse
Affiliation(s)
- Yulia A. Nevzorova
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Francisco Javier Cubero
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| |
Collapse
|
6
|
Liu F, Liao Z, Zhang Z. MYC in liver cancer: mechanisms and targeted therapy opportunities. Oncogene 2023; 42:3303-3318. [PMID: 37833558 DOI: 10.1038/s41388-023-02861-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 09/28/2023] [Accepted: 10/03/2023] [Indexed: 10/15/2023]
Abstract
MYC, a major oncogenic transcription factor, regulates target genes involved in various pathways such as cell proliferation, metabolism and immune evasion, playing a critical role in the tumor initiation and development in multiple types of cancer. In liver cancer, MYC and its signaling pathways undergo significant changes, exerting a profound impact on liver cancer progression, including tumor proliferation, metastasis, dedifferentiation, metabolism, immune microenvironment, and resistance to comprehensive therapies. This makes MYC an appealing target, despite it being previously considered an undruggable protein. In this review, we discuss the role and mechanisms of MYC in liver physiology, chronic liver diseases, hepatocarcinogenesis, and liver cancer progression, providing a theoretical basis for targeting MYC as an ideal therapeutic target for liver cancer. We also summarize and prospect the strategies for targeting MYC, including direct and indirect approaches to abolish the oncogenic function of MYC in liver cancer.
Collapse
Affiliation(s)
- Furong Liu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, 430030, China
- Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Zhibin Liao
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, 430030, China
- Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Zhanguo Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, 430030, China.
- Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China.
| |
Collapse
|
7
|
Qu W, Yan Y, Gerrish K, Scappini E, Tucker CJ, Dixon D, Merrick BA. Chronic PFOA exposure in vitro causes acquisition of multiple tumor cell characteristics in rat liver cells. Toxicol In Vitro 2023; 89:105577. [PMID: 36849026 PMCID: PMC10427995 DOI: 10.1016/j.tiv.2023.105577] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 02/09/2023] [Accepted: 02/18/2023] [Indexed: 02/27/2023]
Abstract
Perfluorooctanoic acid (PFOA) is tumorigenic in rats and mice and potentially tumorigenic in humans. Here, we studied long-term PFOA exposure with an in vitro transformation model using the rat liver epithelial cell, TRL 1215. Cells were cultured in 10 μM (T10), 50 μM (T50) and 100 μM (T100) PFOA for 38 weeks and compared to passage-matched control cells. T100 cells showed morphological changes, loss of cell contact inhibition, formation of multinucleated giant and spindle-shaped cells. T10, T50, and T100 cells showed increased LC50 values 20%, 29% to 35% above control with acute PFOA treatment, indicating a resistance to PFOA toxicity. PFOA-treated cells showed increases in Matrix metalloproteinase-9 secretion, cell migration, and developed more and larger colonies in soft agar. Microarray data showed Myc pathway activation at T50 and T100, associating Myc upregulation with PFOA-induced morphological transformation. Western blot confirmed that PFOA produced significant increases in c-MYC protein expression in a time- and concentration-related manner. Tumor invasion indicators MMP-2 and MMP-9, cell cycle regulator cyclin D1, and oxidative stress protein GST were all significantly overexpressed in T100 cells. Taken together, chronic in vitro PFOA exposure produced multiple cell characteristics of malignant progression and differential gene expression changes suggestive of rat liver cell transformation.
Collapse
Affiliation(s)
- Wei Qu
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences (NIEHS), 111 TW Alexander Drive, Research Triangle Park, NC 27709, USA.
| | - Yitang Yan
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences (NIEHS), 111 TW Alexander Drive, Research Triangle Park, NC 27709, USA
| | - Kevin Gerrish
- Molecular Genomics Core Laboratory, NIEHS, 111 TW Alexander Drive, Research Triangle Park, NC 27709, USA
| | - Erica Scappini
- Fluorescence Microscopy and Imaging Center, Signal Transduction Laboratory, NIEHS, 111 TW Alexander Drive, Research Triangle Park, NC 27709, USA
| | - Charles J Tucker
- Fluorescence Microscopy and Imaging Center, Signal Transduction Laboratory, NIEHS, 111 TW Alexander Drive, Research Triangle Park, NC 27709, USA
| | - Darlene Dixon
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences (NIEHS), 111 TW Alexander Drive, Research Triangle Park, NC 27709, USA
| | - B Alex Merrick
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences (NIEHS), 111 TW Alexander Drive, Research Triangle Park, NC 27709, USA
| |
Collapse
|
8
|
Ramadori P, Woitok MM, Estévez-Vázquez O, Benedé-Ubieto R, Leal-Lassalle H, Lamas-Paz A, Guo F, Fabre J, Otto J, Verwaayen A, Reissing J, Bruns T, Erschfeld S, Haas U, Paffen D, Nelson LJ, Vaquero J, Bañares R, Trautwein C, Cubero FJ, Liedtke C, Nevzorova YA. Lack of Cyclin E1 in hepatocytes aggravates ethanol-induced liver injury and hepatic steatosis in experimental murine model of acute and chronic alcohol-associated liver disease. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166646. [PMID: 36736843 DOI: 10.1016/j.bbadis.2023.166646] [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: 09/13/2022] [Revised: 01/13/2023] [Accepted: 01/17/2023] [Indexed: 02/04/2023]
Abstract
BACKGROUND Cyclin E1 is the regulatory subunit of cyclin-dependent kinase 2 (Cdk2) and one of the central players in cell cycle progression. We recently showed its crucial role for initiation of liver fibrosis and hepatocarcinogenesis. In the present study, we investigated the role of Cyclin E1 in the development of alcohol-associated liver disease (ALD). METHODS Mice with constitutive (E1-/-), hepatocyte-specific (Cyclin E1Δhepa), or intestinal-epithelial-cell-specific (Cyclin E1ΔIEC) inactivation of Cyclin E1 and corresponding wild type littermate controls (WT) were administered either a Lieber-DeCarli ethanol diet (LDE) for 3 weeks or acute ethanol binges (6 g/kg) through oral gavage. Serum parameters of liver functionality were measured; hepatic tissues were collected for biochemical and histological analyses. RESULTS The administration of acute EtOH binge and chronic LDE diet to E1-/- mice enhanced hepatic steatosis, worsened liver damage and triggered body weight loss. Similarly, in the acute EtOH binge model, Cyclin E1Δhepa mice revealed a significantly worsened liver phenotype. In contrast, inactivation of Cyclin E1 only in intestinal epithelial cell (IECs)did not lead to any significant changes in comparison to WT mice after acute EtOH challenge. Remarkably, both acute and chronic EtOH administration in E1-/- animals resulted in increased levels of ADH and decreased expression of ALDH1/2. The additional application of a pan-Cdk inhibitor (S-CR8) further promoted liver damage in EtOH-treated WT mice. CONCLUSION Our data point to a novel unexpected role of Cyclin E1 in hepatocytes for alcohol metabolism, which seems to be independent of the canonical Cyclin E1/Cdk2 function as a cell cycle regulator.
Collapse
Affiliation(s)
- Pierluigi Ramadori
- Division of Chronic Inflammation and Cancer, German Cancer Research Center Heidelberg (DKFZ), Heidelberg, Germany
| | | | - Olga Estévez-Vázquez
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, Madrid, Spain
| | - Raquel Benedé-Ubieto
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, Madrid, Spain; Department of Genetics, Physiology and Microbiology, Faculty of Biology, Complutense University, Madrid, Spain
| | - Hector Leal-Lassalle
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, Madrid, Spain
| | - Arantza Lamas-Paz
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, Madrid, Spain; 12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Feifei Guo
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, Madrid, Spain; Department of Obstetrics and Gynaecology, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Jeanne Fabre
- Polytech Angers, Département Génie Biologique et Santé, Angers, France
| | - Julia Otto
- Department of Internal Medicine III, University Hospital RWTH, Aachen, Germany
| | - Anna Verwaayen
- Department of Internal Medicine III, University Hospital RWTH, Aachen, Germany
| | - Johanna Reissing
- Department of Internal Medicine III, University Hospital RWTH, Aachen, Germany
| | - Tony Bruns
- Department of Internal Medicine III, University Hospital RWTH, Aachen, Germany
| | - Stephanie Erschfeld
- Department of Internal Medicine III, University Hospital RWTH, Aachen, Germany
| | - Ute Haas
- Department of Internal Medicine III, University Hospital RWTH, Aachen, Germany
| | - Daniela Paffen
- Department of Internal Medicine III, University Hospital RWTH, Aachen, Germany
| | - Leonard J Nelson
- Institute for Bioengineering (IBioE), School of Engineering, The University of Edinburgh, Faraday Building, Edinburgh EH9 3 JL, UK
| | - Javier Vaquero
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain,; Servicio de Aparato Digestivo, Hospital General Universitario Gregorio Marañón, Madrid, Spain,; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Rafael Bañares
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain,; Servicio de Aparato Digestivo, Hospital General Universitario Gregorio Marañón, Madrid, Spain,; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Christian Trautwein
- Department of Internal Medicine III, University Hospital RWTH, Aachen, Germany
| | - Francisco Javier Cubero
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain,; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Christian Liedtke
- Department of Internal Medicine III, University Hospital RWTH, Aachen, Germany
| | - Yulia A Nevzorova
- Department of Internal Medicine III, University Hospital RWTH, Aachen, Germany; Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain,; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.
| |
Collapse
|
9
|
Cheng W, Li M, Zhang L, Zhou C, Yu S, Peng X, Zhang W, Zhang W. New roles of N6-methyladenosine methylation system regulating the occurrence of non-alcoholic fatty liver disease with N6-methyladenosine-modified MYC. Front Pharmacol 2022; 13:973116. [PMID: 36120320 PMCID: PMC9471244 DOI: 10.3389/fphar.2022.973116] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 07/28/2022] [Indexed: 12/04/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) has become a major chronic disease in contemporary society, affected by N6-methyladenosine (m6A) RNA methylation, one of the most common RNA modifications. Compared with healthy control, m6A RNA methyltransferase 3 (METTL3) and METTL14 increased, while Wilms tumor 1-associated protein (WTAP) and RNA-binding motif protein 15 (RBM15) decreased significantly in NAFLD, and the m6A demethylases fat mass and obesity-associated protein (FTO) elevated. Meanwhile, the m6A binding proteins, YT521-B homology (YTH) domain-containing 1 (YTHDC1), YTHDC2, insulin-like growth factor 2 mRNA binding protein 1 (IGF2BP1), heterogeneous nuclear ribonucleoprotein C (HNRNPC), and HNRNPA2B1 were decreased, while eukaryotic translation initiation factor 3 subunit H (EIF3H) was increased significantly. All these changes of m6A regulators had significant differences between healthy control and NAFLD, but no differences between the NAFL and NASH group. The expression level of RBM15, HNRNPC, and HNRNPA2B1 were related to body fat index. RBM15, YTHDC2, HNRNPC, HNRNPA2B1, and EIF3H were related to steatosis. Also, KIAA1429 and YTH domain family 1 (YTHDF1) were related to lobular inflammation. Taken together, m6A regulators were involved in the occurrence of NAFLD. More importantly, abnormal MYC was determined as a key link to m6A regulation of NAFLD. The higher MYC mRNA level was accompanied by higher HDL cholesterol and unsaturated fatty acid proportions, as well as lower fat mass, glucose, and transaminase. Taken together, dysregulation of m6A methylation caused steatosis and fibrosis, affecting the occurrence of NAFLD, and MYC might be its potential target.
Collapse
Affiliation(s)
- Wenli Cheng
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Min Li
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Luyun Zhang
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Cheng Zhou
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Susu Yu
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Xinyue Peng
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Wenji Zhang
- Guangdong Provincial Engineering and Technology Research Center for Tobacco Breeding and Comprehensive Utilization, Crops Research Institute, Guangdong Academy of Agricultural Science, Guangzhou, China
- *Correspondence: Wenji Zhang, ; Wenjuan Zhang,
| | - Wenjuan Zhang
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
- *Correspondence: Wenji Zhang, ; Wenjuan Zhang,
| |
Collapse
|
10
|
Sun R, Lei C, Chen L, He L, Guo H, Zhang X, Feng W, Yan J, McClain CJ, Deng Z. Alcohol-driven metabolic reprogramming promotes development of RORγt-deficient thymic lymphoma. Oncogene 2022; 41:2287-2302. [PMID: 35246617 PMCID: PMC9018612 DOI: 10.1038/s41388-022-02257-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 02/10/2022] [Accepted: 02/17/2022] [Indexed: 11/23/2022]
Abstract
RORγt is a master regulator of Th17 cells. Despite evidence linking RORγt deficiency/inhibition with metastatic thymic T cell lymphomas, the role of RORγt in lymphoma metabolism is unknown. Chronic alcohol consumption plays a causal role in many human cancers. The risk of T cell lymphoma remains unclear in humans with alcohol use disorders (AUD) after chronic RORγt inhibition. Here we demonstrated that alcohol consumption accelerates RORγt deficiency-induced lymphomagenesis. Loss of RORγt signaling in the thymus promotes aerobic glycolysis and glutaminolysis and increases allocation of glutamine carbon into lipids. Importantly, alcohol consumption results in a shift from aerobic glycolysis to glutaminolysis. Both RORγt deficiency- and alcohol-induced metabolic alterations are mediated by c-Myc, as silencing of c-Myc decreases the effects of alcohol consumption and RORγt deficiency on glutaminolysis, biosynthesis, and tumor growth in vivo. The ethanol-mediated c-Myc activation coupled with increased glutaminolysis underscore the critical role of RORγt-Myc signaling and translation in lymphoma.
Collapse
Affiliation(s)
- Rui Sun
- Department of Surgery, Division of Immunotherapy, University of Louisville, Louisville, KY, USA
- Department of Oncology, Wuhan Fourth Hospital, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430033, China
- Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Chao Lei
- Department of Surgery, Division of Immunotherapy, University of Louisville, Louisville, KY, USA
- Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Liang Chen
- Department of Surgery, Division of Immunotherapy, University of Louisville, Louisville, KY, USA
- Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Liqing He
- Department of Chemistry, University of Louisville, Louisville, KY, USA
| | - Haixun Guo
- Department of Radiology, University of Louisville, Louisville, KY, USA
| | - Xiang Zhang
- Department of Chemistry, University of Louisville, Louisville, KY, USA
- Alcohol Research Center, University of Louisville, Louisville, KY, USA
- Hepatobiology & Toxicology Center, University of Louisville, Louisville, KY, USA
| | - Wenke Feng
- Alcohol Research Center, University of Louisville, Louisville, KY, USA
- Hepatobiology & Toxicology Center, University of Louisville, Louisville, KY, USA
- Department of Medicine, University of Louisville, Louisville, KY, USA
| | - Jun Yan
- Department of Surgery, Division of Immunotherapy, University of Louisville, Louisville, KY, USA
- Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Craig J McClain
- Alcohol Research Center, University of Louisville, Louisville, KY, USA
- Hepatobiology & Toxicology Center, University of Louisville, Louisville, KY, USA
- Department of Medicine, University of Louisville, Louisville, KY, USA
- Robley Rex VA Medical Center, Louisville, KY, USA
| | - Zhongbin Deng
- Department of Surgery, Division of Immunotherapy, University of Louisville, Louisville, KY, USA.
- Brown Cancer Center, University of Louisville, Louisville, KY, USA.
- Alcohol Research Center, University of Louisville, Louisville, KY, USA.
- Hepatobiology & Toxicology Center, University of Louisville, Louisville, KY, USA.
| |
Collapse
|
11
|
Zhu Y, Wang X, Zhu L, Tu Y, Chen W, Gong L, Pan T, Lin H, Lin J, Sun H, Ge Y, Wei L, Guo Y, Lu C, Chen Y, Xu L. Lactobacillus rhamnosus GG combined with inosine ameliorates alcohol-induced liver injury through regulation of intestinal barrier and Treg/Th1 cells. Toxicol Appl Pharmacol 2022; 439:115923. [PMID: 35176292 DOI: 10.1016/j.taap.2022.115923] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 02/08/2022] [Accepted: 02/11/2022] [Indexed: 02/06/2023]
Abstract
BACKGROUND Intestinal epithelial barrier disruption and bacterial translocation exacerbates the progression of alcoholic liver disease. Lactobacillus rhamnosus GG (LGG), a probiotic, has been shown benefits in chronic liver disease and in regulating gut dysbiosis. Previous studies showed the protective roles of LGG in ethanol-disrupted gut barrier functions and liver injury. Inosine, a metabolite produced by intestinal bacteria, has the anti-inflammatory and immunregulatory functions. In this study, the synergistic effect of LGG and inosine was investigated in a mouse model of alcohol-induced liver disease (ALD). METHODS Male C57BL/6 mice were fed with a Lieber-DeCarli diet containing 5% alcohol for four weeks to establish a model of alcohol-induced liver injury. LGG or a combination of LGG and inosine were administrated orally to explore a new therapeutic method for alcohol-induced liver disease and to investigate the underlying mechanisms. Liver damage was evaluated by transaminases and pathological changes. Tight junction proteins, composition of the gut microbiome, cytokines, lipopolysaccharides (LPS), glutathione (GSH), superoxide dismutase (SOD), malondialdehyde (MDA), F4/80+ macrophages, as well as p38, Jun N-terminal kinase (JNK), were determined by qRT-PCR, RNAseq, ELISA, IHC and western blot. Regulatory T (Treg) cells were characterized by positive staining of CD4, CD25 and Foxp3 using flow cytometry. IFN-γ-producing CD4+ T (Th1) cells were examined by intracellular cytokine staining. RESULTS Alcohol consumption induced elevated liver enzymes, steatosis and inflammation, while LGG combined with inosine treatment was more significant to ameliorate these symptoms compared with LGG alone. When LGG combined with inosine were administered to ALD mice, intestinal microecology significantly improved reflected by intestinal villi and tight junction proteins recovery and the restoration of intestinal flora. Combined therapy inhibited phosphorylation of p38 and JNK to alleviate hepatic inflammation. Moreover, flow cytometry analysis showed that long-term excessive alcohol consumption reduced Tregs population while increased Th1 population, which was restored by a combination of LGG and inosine treatment. CONCLUSIONS The findings from the study indicate that the combined LGG and inosine treatment ameliorates ALD by improving the gut ecosystem, intestinal barrier function, immune homeostasis and liver injury.
Collapse
Affiliation(s)
- Yin Zhu
- Department of Infectious Diseases and liver Diseases, Ningbo Medical Centre Lihuili Hospital, Affiliated Lihuili Hospital of Ningbo University, Ningbo Institute of Innovation for Combined Medicine and Engineering, Ningbo 315040, China; Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Hepatology Institute of Wenzhou Medical University, Wenzhou 325025, China
| | - Xiaozhi Wang
- Department of General and Gastrointestinal Surgery, The Affiliated Mindong Hospital of Fujian Medical University, Fujian 355000, China
| | - Lujian Zhu
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Hepatology Institute of Wenzhou Medical University, Wenzhou 325025, China
| | - Yulu Tu
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Hepatology Institute of Wenzhou Medical University, Wenzhou 325025, China
| | - Wanting Chen
- Department of Infectious Diseases and liver Diseases, Ningbo Medical Centre Lihuili Hospital, Affiliated Lihuili Hospital of Ningbo University, Ningbo Institute of Innovation for Combined Medicine and Engineering, Ningbo 315040, China
| | - Lingwen Gong
- Department of Infectious Diseases and liver Diseases, Ningbo Medical Centre Lihuili Hospital, Affiliated Lihuili Hospital of Ningbo University, Ningbo Institute of Innovation for Combined Medicine and Engineering, Ningbo 315040, China
| | - Tongtong Pan
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Hepatology Institute of Wenzhou Medical University, Wenzhou 325025, China
| | - Hongwei Lin
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Hepatology Institute of Wenzhou Medical University, Wenzhou 325025, China
| | - Jing Lin
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Hepatology Institute of Wenzhou Medical University, Wenzhou 325025, China
| | - Huiling Sun
- Department of Infectious Diseases and Liver Diseases, Lishui City People's Hospital, Lishui 323020, China
| | - Yuli Ge
- Department of Infectious Diseases and Liver Diseases, Lishui City People's Hospital, Lishui 323020, China
| | - Li Wei
- Department of Infectious Diseases and liver Diseases, Ningbo Medical Centre Lihuili Hospital, Affiliated Lihuili Hospital of Ningbo University, Ningbo Institute of Innovation for Combined Medicine and Engineering, Ningbo 315040, China
| | - Yu Guo
- Department of Infectious Diseases and liver Diseases, Ningbo Medical Centre Lihuili Hospital, Affiliated Lihuili Hospital of Ningbo University, Ningbo Institute of Innovation for Combined Medicine and Engineering, Ningbo 315040, China
| | - Caide Lu
- Department of Hepatobiliary and Pancreatic Surgery, Ningbo Medical Centre Lihuili Hospital, Affiliated Lihuili Hospital of Ningbo University, Ningbo 315040, China.
| | - Yongping Chen
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Hepatology Institute of Wenzhou Medical University, Wenzhou 325025, China.
| | - Lanman Xu
- Department of Infectious Diseases and liver Diseases, Ningbo Medical Centre Lihuili Hospital, Affiliated Lihuili Hospital of Ningbo University, Ningbo Institute of Innovation for Combined Medicine and Engineering, Ningbo 315040, China; Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Hepatology Institute of Wenzhou Medical University, Wenzhou 325025, China.
| |
Collapse
|
12
|
Liedtke C, Nevzorova YA, Luedde T, Zimmermann H, Kroy D, Strnad P, Berres ML, Bernhagen J, Tacke F, Nattermann J, Spengler U, Sauerbruch T, Wree A, Abdullah Z, Tolba RH, Trebicka J, Lammers T, Trautwein C, Weiskirchen R. Liver Fibrosis-From Mechanisms of Injury to Modulation of Disease. Front Med (Lausanne) 2022; 8:814496. [PMID: 35087852 PMCID: PMC8787129 DOI: 10.3389/fmed.2021.814496] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 12/15/2021] [Indexed: 12/12/2022] Open
Abstract
The Transregional Collaborative Research Center "Organ Fibrosis: From Mechanisms of Injury to Modulation of Disease" (referred to as SFB/TRR57) was funded for 13 years (2009-2021) by the German Research Council (DFG). This consortium was hosted by the Medical Schools of the RWTH Aachen University and Bonn University in Germany. The SFB/TRR57 implemented combined basic and clinical research to achieve detailed knowledge in three selected key questions: (i) What are the relevant mechanisms and signal pathways required for initiating organ fibrosis? (ii) Which immunological mechanisms and molecules contribute to organ fibrosis? and (iii) How can organ fibrosis be modulated, e.g., by interventional strategies including imaging and pharmacological approaches? In this review we will summarize the liver-related key findings of this consortium gained within the last 12 years on these three aspects of liver fibrogenesis. We will highlight the role of cell death and cell cycle pathways as well as nutritional and iron-related mechanisms for liver fibrosis initiation. Moreover, we will define and characterize the major immune cell compartments relevant for liver fibrogenesis, and finally point to potential signaling pathways and pharmacological targets that turned out to be suitable to develop novel approaches for improved therapy and diagnosis of liver fibrosis. In summary, this review will provide a comprehensive overview about the knowledge on liver fibrogenesis and its potential therapy gained by the SFB/TRR57 consortium within the last decade. The kidney-related research results obtained by the same consortium are highlighted in an article published back-to-back in Frontiers in Medicine.
Collapse
Affiliation(s)
- Christian Liedtke
- Department of Internal Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Yulia A. Nevzorova
- Department of Internal Medicine III, University Hospital RWTH Aachen, Aachen, Germany
- Department of Immunology, Ophthalmology and Otolaryngology, School of Medicine, Complutense University Madrid, Madrid, Spain
| | - Tom Luedde
- Medical Faculty, Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Duesseldorf, Heinrich Heine University, Duesseldorf, Germany
| | - Henning Zimmermann
- Department of Internal Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Daniela Kroy
- Department of Internal Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Pavel Strnad
- Department of Internal Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Marie-Luise Berres
- Department of Internal Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Jürgen Bernhagen
- Chair of Vascular Biology, Institute for Stroke and Dementia Research (ISD), Klinikum der Universität München (KUM), Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany
| | - Jacob Nattermann
- Department of Internal Medicine I, University Hospital Bonn, Bonn, Germany
| | - Ulrich Spengler
- Department of Internal Medicine I, University Hospital Bonn, Bonn, Germany
| | - Tilman Sauerbruch
- Department of Internal Medicine I, University Hospital Bonn, Bonn, Germany
| | - Alexander Wree
- Department of Hepatology and Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany
| | - Zeinab Abdullah
- Institute for Molecular Medicine and Experimental Immunology, University Hospital of Bonn, Bonn, Germany
| | - René H. Tolba
- Institute for Laboratory Animal Science and Experimental Surgery, RWTH Aachen University Hospital, Aachen, Germany
| | - Jonel Trebicka
- Department of Internal Medicine I, University Hospital Frankfurt, Frankfurt, Germany
| | - Twan Lammers
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen, Germany
| | - Christian Trautwein
- Department of Internal Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), University Hospital RWTH Aachen, Aachen, Germany
| |
Collapse
|
13
|
Guo F, Estévez-Vázquez O, Benedé-Ubieto R, Maya-Miles D, Zheng K, Gallego-Durán R, Rojas Á, Ampuero J, Romero-Gómez M, Philip K, Egbuniwe IU, Chen C, Simon J, Delgado TC, Martínez-Chantar ML, Sun J, Reissing J, Bruns T, Lamas-Paz A, del Moral MG, Woitok MM, Vaquero J, Regueiro JR, Liedtke C, Trautwein C, Bañares R, Cubero FJ, Nevzorova YA. A Shortcut from Metabolic-Associated Fatty Liver Disease (MAFLD) to Hepatocellular Carcinoma (HCC): c-MYC a Promising Target for Preventative Strategies and Individualized Therapy. Cancers (Basel) 2021; 14:cancers14010192. [PMID: 35008356 PMCID: PMC8750626 DOI: 10.3390/cancers14010192] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/16/2021] [Accepted: 12/20/2021] [Indexed: 12/02/2022] Open
Abstract
Simple Summary Metabolic-associated fatty liver disease (MAFLD) is a chronic liver disease associated with obesity, diabetes mellitus type 2 (DM2), and hyperlipidemia. It can also progress to end-stage hepatocellular carcinoma (HCC); the underlying mechanisms are still unknown, but endogenous (i.e., genetic) factors such as oncogenes have been suggested to play a role. We found that c-MYC transgenic mice with ageing are prone to develop obesity, metabolic syndrome (MS), and abnormal accumulation of lipids in the liver compared to control mice. A short-term application of the Western diet (WD) significantly worsened the phenotype and accelerate HCC development. Importantly, we found that metformin as therapeutic approach significantly attenuated MAFLD phenotype in transgenic mice. We also observed that c-MYC is up-regulated in human patients with MAFLD and MAFLD-related HCC. Altogether the current study suggests an important role of the oncogene c-MYC during the progression from MAFLD to HCC and makes c-MYC a possible target for preventative strategies and individualized therapy. Abstract Background: Metabolic-associated fatty liver disease (MAFLD) has risen as one of the leading etiologies for hepatocellular carcinoma (HCC). Oncogenes have been suggested to be responsible for the high risk of MAFLD-related HCC. We analyzed the impact of the proto-oncogene c-MYC in the development of human and murine MAFLD and MAFLD-associated HCC. Methods: alb-myctg mice were studied at baseline conditions and after administration of Western diet (WD) in comparison to WT littermates. c-MYC expression was analyzed in biopsies of patients with MAFLD and MAFLD-associated HCC by immunohistochemistry. Results: Mild obesity, spontaneous hyperlipidaemia, glucose intolerance and insulin resistance were characteristic of 36-week-old alb-myctg mice. Middle-aged alb-myctg exhibited liver steatosis and increased triglyceride content. Liver injury and inflammation were associated with elevated ALT, an upregulation of ER-stress response and increased ROS production, collagen deposition and compensatory proliferation. At 52 weeks, 20% of transgenic mice developed HCC. WD feeding exacerbated metabolic abnormalities, steatohepatitis, fibrogenesis and tumor prevalence. Therapeutic use of metformin partly attenuated the spontaneous MAFLD phenotype of alb-myctg mice. Importantly, upregulation and nuclear localization of c-MYC were characteristic of patients with MAFLD and MAFLD-related HCC. Conclusions: A novel function of c-MYC in MAFLD progression was identified opening new avenues for preventative strategies.
Collapse
Affiliation(s)
- Feifei Guo
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University of Madrid, 12 de Octubre (imas12) Health Research Institute, 28040 Madrid, Spain; (F.G.); (O.E.-V.); (R.B.-U.); (K.Z.); (C.C.); (A.L.-P.); (J.R.R.); (R.B.); (F.J.C.)
- Department of Obstetrics and Gynaecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210023, China
| | - Olga Estévez-Vázquez
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University of Madrid, 12 de Octubre (imas12) Health Research Institute, 28040 Madrid, Spain; (F.G.); (O.E.-V.); (R.B.-U.); (K.Z.); (C.C.); (A.L.-P.); (J.R.R.); (R.B.); (F.J.C.)
| | - Raquel Benedé-Ubieto
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University of Madrid, 12 de Octubre (imas12) Health Research Institute, 28040 Madrid, Spain; (F.G.); (O.E.-V.); (R.B.-U.); (K.Z.); (C.C.); (A.L.-P.); (J.R.R.); (R.B.); (F.J.C.)
- Department of Physiology, Genetics and Microbiology, Faculty of Biology, Complutense University Madrid, 28040 Madrid, Spain
| | - Douglas Maya-Miles
- Institute of Biomedicine of Seville (IBiS), SeLiver Group, Virgen del Rocío University Hospital/CSIC/University of Seville, 41013 Seville, Spain; (D.M.-M.); (R.G.-D.); (Á.R.); (J.A.); (M.R.-G.)
- UCM Digestive Diseases, Virgen del Rocío University Hospital, 41013 Seville, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), 28220 Madrid, Spain; (J.S.); (M.L.M.-C.); (J.V.)
| | - Kang Zheng
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University of Madrid, 12 de Octubre (imas12) Health Research Institute, 28040 Madrid, Spain; (F.G.); (O.E.-V.); (R.B.-U.); (K.Z.); (C.C.); (A.L.-P.); (J.R.R.); (R.B.); (F.J.C.)
- Department of Anesthesiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China;
| | - Rocío Gallego-Durán
- Institute of Biomedicine of Seville (IBiS), SeLiver Group, Virgen del Rocío University Hospital/CSIC/University of Seville, 41013 Seville, Spain; (D.M.-M.); (R.G.-D.); (Á.R.); (J.A.); (M.R.-G.)
- UCM Digestive Diseases, Virgen del Rocío University Hospital, 41013 Seville, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), 28220 Madrid, Spain; (J.S.); (M.L.M.-C.); (J.V.)
| | - Ángela Rojas
- Institute of Biomedicine of Seville (IBiS), SeLiver Group, Virgen del Rocío University Hospital/CSIC/University of Seville, 41013 Seville, Spain; (D.M.-M.); (R.G.-D.); (Á.R.); (J.A.); (M.R.-G.)
- UCM Digestive Diseases, Virgen del Rocío University Hospital, 41013 Seville, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), 28220 Madrid, Spain; (J.S.); (M.L.M.-C.); (J.V.)
| | - Javier Ampuero
- Institute of Biomedicine of Seville (IBiS), SeLiver Group, Virgen del Rocío University Hospital/CSIC/University of Seville, 41013 Seville, Spain; (D.M.-M.); (R.G.-D.); (Á.R.); (J.A.); (M.R.-G.)
- UCM Digestive Diseases, Virgen del Rocío University Hospital, 41013 Seville, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), 28220 Madrid, Spain; (J.S.); (M.L.M.-C.); (J.V.)
| | - Manuel Romero-Gómez
- Institute of Biomedicine of Seville (IBiS), SeLiver Group, Virgen del Rocío University Hospital/CSIC/University of Seville, 41013 Seville, Spain; (D.M.-M.); (R.G.-D.); (Á.R.); (J.A.); (M.R.-G.)
- UCM Digestive Diseases, Virgen del Rocío University Hospital, 41013 Seville, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), 28220 Madrid, Spain; (J.S.); (M.L.M.-C.); (J.V.)
- Department of Medicine, University of Seville, 41009 Seville, Spain
| | - Kaye Philip
- Department of Pathology, Nottingham University Hospitals NHS Trust, Queen’s Medical Centre Campus, Nottingham NG7 2UH, UK; (K.P.); (I.U.E.)
| | - Isioma U. Egbuniwe
- Department of Pathology, Nottingham University Hospitals NHS Trust, Queen’s Medical Centre Campus, Nottingham NG7 2UH, UK; (K.P.); (I.U.E.)
| | - Chaobo Chen
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University of Madrid, 12 de Octubre (imas12) Health Research Institute, 28040 Madrid, Spain; (F.G.); (O.E.-V.); (R.B.-U.); (K.Z.); (C.C.); (A.L.-P.); (J.R.R.); (R.B.); (F.J.C.)
- Department of General Surgery, Wuxi Xishan People’s Hospital, Wuxi 214000, China
- Department of Hepatic-Biliary-Pancreatic Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210023, China
| | - Jorge Simon
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), 28220 Madrid, Spain; (J.S.); (M.L.M.-C.); (J.V.)
- Liver Disease Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), 48160 Derio, Spain;
| | - Teresa C. Delgado
- Liver Disease Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), 48160 Derio, Spain;
| | - María Luz Martínez-Chantar
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), 28220 Madrid, Spain; (J.S.); (M.L.M.-C.); (J.V.)
- Liver Disease Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), 48160 Derio, Spain;
| | - Jie Sun
- Department of Anesthesiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China;
| | - Johanna Reissing
- Department of Internal Medicine III, University Hospital RWTH Aachen, 52074 Aachen, Germany; (J.R.); (T.B.); (M.M.W.); (C.L.); (C.T.)
| | - Tony Bruns
- Department of Internal Medicine III, University Hospital RWTH Aachen, 52074 Aachen, Germany; (J.R.); (T.B.); (M.M.W.); (C.L.); (C.T.)
| | - Arantza Lamas-Paz
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University of Madrid, 12 de Octubre (imas12) Health Research Institute, 28040 Madrid, Spain; (F.G.); (O.E.-V.); (R.B.-U.); (K.Z.); (C.C.); (A.L.-P.); (J.R.R.); (R.B.); (F.J.C.)
| | - Manuel Gómez del Moral
- Department of Cell Biology, Complutense University School of Medicine, 28040 Madrid, Spain;
| | - Marius Maximilian Woitok
- Department of Internal Medicine III, University Hospital RWTH Aachen, 52074 Aachen, Germany; (J.R.); (T.B.); (M.M.W.); (C.L.); (C.T.)
| | - Javier Vaquero
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), 28220 Madrid, Spain; (J.S.); (M.L.M.-C.); (J.V.)
- Servicio de Aparato Digestivo, Hospital General Universitario Gregorio Marañón, 28009 Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), 28007 Madrid, Spain
| | - José R. Regueiro
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University of Madrid, 12 de Octubre (imas12) Health Research Institute, 28040 Madrid, Spain; (F.G.); (O.E.-V.); (R.B.-U.); (K.Z.); (C.C.); (A.L.-P.); (J.R.R.); (R.B.); (F.J.C.)
| | - Christian Liedtke
- Department of Internal Medicine III, University Hospital RWTH Aachen, 52074 Aachen, Germany; (J.R.); (T.B.); (M.M.W.); (C.L.); (C.T.)
| | - Christian Trautwein
- Department of Internal Medicine III, University Hospital RWTH Aachen, 52074 Aachen, Germany; (J.R.); (T.B.); (M.M.W.); (C.L.); (C.T.)
| | - Rafael Bañares
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University of Madrid, 12 de Octubre (imas12) Health Research Institute, 28040 Madrid, Spain; (F.G.); (O.E.-V.); (R.B.-U.); (K.Z.); (C.C.); (A.L.-P.); (J.R.R.); (R.B.); (F.J.C.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), 28220 Madrid, Spain; (J.S.); (M.L.M.-C.); (J.V.)
- Servicio de Aparato Digestivo, Hospital General Universitario Gregorio Marañón, 28009 Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), 28007 Madrid, Spain
| | - Francisco Javier Cubero
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University of Madrid, 12 de Octubre (imas12) Health Research Institute, 28040 Madrid, Spain; (F.G.); (O.E.-V.); (R.B.-U.); (K.Z.); (C.C.); (A.L.-P.); (J.R.R.); (R.B.); (F.J.C.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), 28220 Madrid, Spain; (J.S.); (M.L.M.-C.); (J.V.)
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), 28007 Madrid, Spain
| | - Yulia A. Nevzorova
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Complutense University of Madrid, 12 de Octubre (imas12) Health Research Institute, 28040 Madrid, Spain; (F.G.); (O.E.-V.); (R.B.-U.); (K.Z.); (C.C.); (A.L.-P.); (J.R.R.); (R.B.); (F.J.C.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), 28220 Madrid, Spain; (J.S.); (M.L.M.-C.); (J.V.)
- Department of Internal Medicine III, University Hospital RWTH Aachen, 52074 Aachen, Germany; (J.R.); (T.B.); (M.M.W.); (C.L.); (C.T.)
- Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), 28007 Madrid, Spain
- Correspondence: ; Tel.: +49-(0)241-80-80662; Fax: +49-(0)241-80-82455
| |
Collapse
|
14
|
Luo Y, Yang S, Wu X, Takahashi S, Sun L, Cai J, Krausz KW, Guo X, Dias HB, Gavrilova O, Xie C, Jiang C, Liu W, Gonzalez FJ. Intestinal MYC modulates obesity-related metabolic dysfunction. Nat Metab 2021; 3:923-939. [PMID: 34211180 PMCID: PMC9944847 DOI: 10.1038/s42255-021-00421-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 05/26/2021] [Indexed: 12/14/2022]
Abstract
MYC is a transcription factor with broad biological functions, notably in the control of cell proliferation. Here, we show that intestinal MYC regulates systemic metabolism. We find that MYC expression is increased in ileum biopsies from individuals with obesity and positively correlates with body mass index. Intestine-specific reduction of MYC in mice improves high-fat-diet-induced obesity, insulin resistance, hepatic steatosis and steatohepatitis. Mechanistically, reduced expression of MYC in the intestine promotes glucagon-like peptide-1 (GLP-1) production and secretion. Moreover, we identify Cers4, encoding ceramide synthase 4, catalysing de novo ceramide synthesis, as a MYC target gene. Finally, we show that administration of the MYC inhibitor 10058-F4 has beneficial effects on high-fat-diet-induced metabolic disorders, and is accompanied by increased GLP-1 and reduced ceramide levels in serum. This study positions intestinal MYC as a putative drug target against metabolic diseases, including non-alcoholic fatty liver disease and non-alcoholic steatohepatitis.
Collapse
Affiliation(s)
- Yuhong Luo
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Shoumei Yang
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Xuan Wu
- Department of Laboratory Medicine and Central Laboratory, Shanghai Tenth People's Hospital, Tongji University, Shanghai, P.R. China
- Department of Laboratory Medicine, Shanghai Skin Disease Hospital, Tongji University, Shanghai, P.R. China
| | - Shogo Takahashi
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC, USA
| | - Lulu Sun
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jie Cai
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kristopher W Krausz
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Xiaozhen Guo
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, P.R. China
| | - Henrique B Dias
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Oksana Gavrilova
- Mouse Metabolism Core Laboratory, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Cen Xie
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, P.R. China
| | - Changtao Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P.R. China
| | - Weiwei Liu
- Department of Laboratory Medicine and Central Laboratory, Shanghai Tenth People's Hospital, Tongji University, Shanghai, P.R. China.
- Department of Laboratory Medicine, Shanghai Skin Disease Hospital, Tongji University, Shanghai, P.R. China.
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
15
|
Shibasaki H, Kinoh H, Cabral H, Quader S, Mochida Y, Liu X, Toh K, Miyano K, Matsumoto Y, Yamasoba T, Kataoka K. Efficacy of pH-Sensitive Nanomedicines in Tumors with Different c-MYC Expression Depends on the Intratumoral Activation Profile. ACS NANO 2021; 15:5545-5559. [PMID: 33625824 DOI: 10.1021/acsnano.1c00364] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Effective inhibition of the protein derived from cellular myelocytomatosis oncogene (c-Myc) is one of the most sought-after goals in cancer therapy. While several c-Myc inhibitors have demonstrated therapeutic potential, inhibiting c-Myc has proven challenging, since c-Myc is essential for normal tissues and tumors may present heterogeneous c-Myc levels demanding contrasting therapeutic strategies. Herein, we developed tumor-targeted nanomedicines capable of treating both tumors with high and low c-Myc levels by adjusting their ability to spatiotemporally control drug action. These nanomedicines loaded homologues of the bromodomain and extraterminal (BET) motif inhibitor JQ1 as epigenetic c-Myc inhibitors through pH-cleavable bonds engineered for fast or slow drug release at intratumoral pH. In tumors with high c-Myc expression, the fast-releasing (FR) nanomedicines suppressed tumor growth more effectively than the slow-releasing (SR) ones, whereas, in the low c-Myc tumors, the efficacy of the nanomedicines was the opposite. By studying the tumor distribution and intratumoral activation of the nanomedicines, we found that, despite SR nanomedicines achieved higher accumulation than the FR counterparts in both c-Myc high and low tumors, the antitumor activity profiles corresponded with the availability of activated drugs inside the tumors. These results indicate the potential of engineered nanomedicines for c-Myc inhibition and spur the idea of precision pH-sensitive nanomedicine based on cancer biomarker levels.
Collapse
Affiliation(s)
- Hitoshi Shibasaki
- Department of Otorhinolaryngology and Head and Neck Surgery, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Hiroaki Kinoh
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Horacio Cabral
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Sabina Quader
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Yuki Mochida
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Xueying Liu
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Kazuko Toh
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Kazuki Miyano
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
- Department of Otorhinolaryngology, Tokyo Yamate Medical Center, 3-22-1, Hyakunin-cho, Shinjuku-ku, Tokyo 169-0073, Japan
| | - Yu Matsumoto
- Department of Otorhinolaryngology and Head and Neck Surgery, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Tatsuya Yamasoba
- Department of Otorhinolaryngology and Head and Neck Surgery, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Kazunori Kataoka
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
- Policy Alternative Research Institute, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| |
Collapse
|
16
|
Nevzorova YA, Boyer-Diaz Z, Cubero FJ, Gracia-Sancho J. Animal models for liver disease - A practical approach for translational research. J Hepatol 2020; 73:423-440. [PMID: 32330604 DOI: 10.1016/j.jhep.2020.04.011] [Citation(s) in RCA: 145] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 04/06/2020] [Accepted: 04/06/2020] [Indexed: 12/11/2022]
Abstract
Animal models are crucial for improving our understanding of human pathogenesis, enabling researchers to identify therapeutic targets and test novel drugs. In the current review, we provide a comprehensive summary of the most widely used experimental models of chronic liver disease, starting from early stages of fatty liver disease (non-alcoholic and alcoholic) to steatohepatitis, advanced cirrhosis and end-stage primary liver cancer. We focus on aspects such as reproducibility and practicality, discussing the advantages and weaknesses of available models for researchers who are planning to perform animal studies in the near future. Additionally, we summarise current and prospective models based on human tissue bioengineering.
Collapse
Affiliation(s)
- Yulia A Nevzorova
- Department of Genetics, Physiology and Microbiology, Faculty of Biology, Complutense University, Madrid, Spain; 12 de Octubre Health Research Institute (imas12), Madrid, Spain; Department of Internal Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Zoe Boyer-Diaz
- Liver Vascular Biology Research Group, Barcelona Hepatic Hemodynamic Unit, IDIBAPS Biomedical Research Institute, Barcelona, Spain; Barcelona Liver Bioservices, Barcelona, Spain
| | - Francisco Javier Cubero
- 12 de Octubre Health Research Institute (imas12), Madrid, Spain; Department of Immunology, Ophthalmology & ENT, Complutense University School of Medicine, Madrid, Spain.
| | - Jordi Gracia-Sancho
- Liver Vascular Biology Research Group, Barcelona Hepatic Hemodynamic Unit, IDIBAPS Biomedical Research Institute, Barcelona, Spain; Barcelona Liver Bioservices, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain; Hepatology, Department of Biomedical Research, University of Bern, Bern, Switzerland.
| |
Collapse
|
17
|
Benedé-Ubieto R, Estévez-Vázquez O, Ramadori P, Cubero FJ, Nevzorova YA. Guidelines and Considerations for Metabolic Tolerance Tests in Mice. Diabetes Metab Syndr Obes 2020; 13:439-450. [PMID: 32110077 PMCID: PMC7038777 DOI: 10.2147/dmso.s234665] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 12/24/2019] [Indexed: 01/13/2023] Open
Abstract
The epidemic of the century, Diabetes Mellitus (DM) is continuously rising. Intensive research is urgently needed whereby experimental models represent an essential tool to optimise the diagnostic strategy and to improve therapy. In this review, we describe the central principles of the metabolic tests available in order to study glucose and insulin homeostasis in mice, focusing on the most widely used - the glucose and insulin tolerance tests. We provide detailed experimental procedures as well as the practical implementation of these methods and discuss the main factors that should be taken into account when using this methodology.
Collapse
Affiliation(s)
- Raquel Benedé-Ubieto
- Department of Physiology, Genetics and Microbiology, Faculty of Biology, Complutense University, Madrid, Spain
| | - Olga Estévez-Vázquez
- Department of Physiology, Genetics and Microbiology, Faculty of Biology, Complutense University, Madrid, Spain
| | - Pierluigi Ramadori
- Division of Chronic Inflammation and Cancer, German Cancer Research Center Heidelberg (DKFZ), Heidelberg, Germany
| | - Francisco Javier Cubero
- Department of Immunology, Ophthalmology & ENT, Complutense University School of Medicine, Madrid, Spain
- 12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Yulia A Nevzorova
- Department of Physiology, Genetics and Microbiology, Faculty of Biology, Complutense University, Madrid, Spain
- 12 de Octubre Health Research Institute (imas12), Madrid, Spain
- Department of Internal Medicine III, University Hospital RWTH Aachen, Aachen, Germany
- Correspondence: Yulia A Nevzorova Department of Physiology, Genetics and Microbiology, Faculty of Biology, Complutense University, c/José A. Novais, 2, Madrid28040, SpainTel +49-(0)241-80-80662Fax +49-(0)241-80-82455 Email
| |
Collapse
|
18
|
Wu H, Yang TY, Li Y, Ye WL, Liu F, He XS, Wang JR, Gan WJ, Li XM, Zhang S, Zhao YY, Li JM. Tumor Necrosis Factor Receptor-Associated Factor 6 Promotes Hepatocarcinogenesis by Interacting With Histone Deacetylase 3 to Enhance c-Myc Gene Expression and Protein Stability. Hepatology 2020; 71:148-163. [PMID: 31155734 DOI: 10.1002/hep.30801] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 05/29/2019] [Indexed: 12/12/2022]
Abstract
The oncogene c-Myc is aberrantly expressed and plays a key role in malignant transformation and progression of hepatocellular carcinoma (HCC). Here, we report that c-Myc is significantly up-regulated by tumor necrosis factor receptor-associated factor 6 (TRAF6), an E3 ubiquitin ligase, in hepatocarcinogenesis. High TRAF6 expression in clinical HCC samples correlates with poor prognosis, and the loss of one copy of the Traf6 gene in Traf6+/- mice significantly impairs liver tumorigenesis. Mechanistically, TRAF6 first interacts with and ubiquitinates histone deacetylase 3 (HDAC3) with K63-linked ubiquitin chains, which leads to the dissociation of HDAC3 from the c-Myc promoter and subsequent acetylation of histone H3 at K9, thereby epigenetically enhancing the mRNA expression of c-Myc. Second, the K63-linked ubiquitination of HDAC3 impairs the HDAC3 interaction with c-Myc and promotes c-Myc protein acetylation, which thereby enhances c-Myc protein stability by inhibiting carboxyl terminus of heat shock cognate 70-kDa-interacting protein-mediated c-Myc ubiquitination and degradation. Importantly, TRAF6/HDAC3/c-Myc signaling is also primed in hepatitis B virus-transgenic mice, unveiling a critical role for a mechanism in inflammation-cancer transition. In clinical specimens, TRAF6 positively correlates with c-Myc at both the mRNA and protein levels, and high TRAF6 and c-Myc expression is associated with an unfavorable prognosis, suggesting that TRAF6 collaborates with c-Myc to promote human hepatocarcinogenesis. Consistently, curbing c-Myc expression by inhibition of TRAF6 activity with a TRAF6 inhibitor peptide or the silencing of c-Myc by small interfering RNA significantly suppressed tumor growth in mice. Conclusion: These findings demonstrate the oncogenic potential of TRAF6 during hepatocarcinogenesis by modulating TRAF6/HDAC3/c-Myc signaling, with potential implications for HCC therapy.
Collapse
Affiliation(s)
- Hua Wu
- Department of Pathology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Department of Pathology, Soochow University, Suzhou, China
| | - Tian-Yu Yang
- Department of Pathology, Soochow University, Suzhou, China
| | - Yi Li
- Department of Pathology, Soochow University, Suzhou, China
| | - Wen-Long Ye
- Department of Pathology, Soochow University, Suzhou, China
| | - Feng Liu
- Department of General Surgery, Canglang Hospital of Suzhou, Suzhou, China
| | - Xiao-Shun He
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jing-Ru Wang
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Wen-Juan Gan
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiu-Ming Li
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Shen Zhang
- Department of Pathology, Soochow University, Suzhou, China
| | - Yuan-Yuan Zhao
- Department of Pathology, Soochow University, Suzhou, China
| | - Jian-Ming Li
- Department of Pathology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Department of Pathology, Soochow University, Suzhou, China
| |
Collapse
|
19
|
Chen H, Liu S, Li M, Huang P, Li X. circ_0003418 Inhibits Tumorigenesis And Cisplatin Chemoresistance Through Wnt/β-Catenin Pathway In Hepatocellular Carcinoma. Onco Targets Ther 2019; 12:9539-9549. [PMID: 31807029 PMCID: PMC6857737 DOI: 10.2147/ott.s229507] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 10/25/2019] [Indexed: 12/13/2022] Open
Abstract
Background Accumulating evidences indicate that circRNAs play important roles in the progression and chemoresistance of human cancers. The present study is designated for researching the roles of circ_0003418 in hepatocellular carcinoma (HCC). Methods We detected the expression profile of circ_0003418 in human HCC tissues and cell lines by quantitative real-time-PCR assays. CCK-8 assay, transwell migration assay, transwell invasion assay and drug-sensitivity analysis were carried out to estimate the effects of circ_0003418 on HCC cells' proliferation, migration, invasion and resistance to cisplatin, respectively. Mouse xenograft model was conducted to monitor the role of circ_0003418 in cisplatin resistance in vivo. Western blotting was performed to explore the changes of the Wnt/β-catenin pathway after knockdown of circ_0003418. The rescue experiment was carried out to explore circ_0003418-activated biological functions through Wnt/β-catenin pathway. Results The expression level of circ_0003418 was downregulated in HCC tissues and cell lines, and the level correlated with tumor size, TNM stage and HBsAg level in HCC patients. circ_0003418 knockdown promoted HCC cells' proliferation, migration, and invasion. Additionally, suppression of circ_0003418 enhanced cisplatin resistance of HCC cells in vivo and vitro. Knockdown of circ_0003418 activated the Wnt/β-catenin signalling pathway in HCC cells. The effect of circ-0003418 on sensitivity of HCC cells to cisplatin was reversed after inhibition of Wnt/β-catenin pathway. Conclusion circ-0003418 exerts an antitumorigenic role in HCC and advances the sensitivity of HCC cells to cisplatin by restraining the Wnt/β-catenin pathway. Thus, circ-0003418 may represent a novel biomarker and provide us a new strategy for the treatment of HCC.
Collapse
Affiliation(s)
- Hang Chen
- Department of Oncology and Hematology, The People's Hospital of Tongliang District, Chongqing 402560, People's Republic of China
| | - Shan Liu
- Department of Oncology and Hematology, The People's Hospital of Tongliang District, Chongqing 402560, People's Republic of China
| | - Molin Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing 400000, People's Republic of China
| | - Ping Huang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing 400000, People's Republic of China
| | - Xiaoping Li
- Department of Oncology and Hematology, The People's Hospital of Tongliang District, Chongqing 402560, People's Republic of China
| |
Collapse
|
20
|
Isoflurane preconditioning protects hepatocytes from oxygen glucose deprivation injury by regulating FoxO6. J Biosci 2019. [DOI: 10.1007/s12038-019-9967-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
21
|
Lamas-Paz A, Hao F, Nelson LJ, Vázquez MT, Canals S, Gómez del Moral M, Martínez-Naves E, Nevzorova YA, Cubero FJ. Alcoholic liver disease: Utility of animal models. World J Gastroenterol 2018; 24:5063-5075. [PMID: 30568384 PMCID: PMC6288648 DOI: 10.3748/wjg.v24.i45.5063] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 11/08/2018] [Accepted: 11/09/2018] [Indexed: 02/06/2023] Open
Abstract
Alcoholic liver disease (ALD) is a major cause of acute and chronic liver injury. Extensive evidence has been accumulated on the pathological process of ALD during the past decades. However, effective treatment options for ALD are very limited due to the lack of suitable in vivo models that recapitulate the full spectrum of ALD. Experimental animal models of ALD, particularly rodents, have been used extensively to mimic human ALD. An ideal animal model should recapitulate all aspects of the ALD process, including significant steatosis, hepatic neutrophil infiltration, and liver injury. A better strategy against ALD depends on clear diagnostic biomarkers, accurate predictor(s) of its progression and new therapeutic approaches to modulate stop or even reverse the disease. Numerous models employing rodent animals have been established in the last decades to investigate the effects of acute and chronic alcohol exposure on the initiation and progression of ALD. Although significant progress has been made in gaining better knowledge on the mechanisms and pathology of ALD, many features of ALD are unknown, and require further investigation, ideally with improved animal models that more effectively mimic human ALD. Although differences in the degree and stages of alcoholic liver injury inevitably exist between animal models and human ALD, the acquisition and translational relevance will be greatly enhanced with the development of new and improved animal models of ALD.
Collapse
Affiliation(s)
- Arantza Lamas-Paz
- Department of Immunology, Ophthalmology and ORL, Complutense University School of Medicine, Madrid 28040, Spain
- Yulia A Nevzovova, Francisco Javier Cubero, 12 de Octubre Health Research Institute (imas12), Madrid 28041, Spain
| | - Fengjie Hao
- Department of Immunology, Ophthalmology and ORL, Complutense University School of Medicine, Madrid 28040, Spain
- Yulia A Nevzovova, Francisco Javier Cubero, 12 de Octubre Health Research Institute (imas12), Madrid 28041, Spain
| | - Leonard J Nelson
- Institute for Bioengineering (IBioE), School of Engineering, Faraday Building, The University of Edinburgh, Edinburgh EH9 3 JL, Scotland, United Kingdom
| | - Maria Teresa Vázquez
- Department of Human Anatomy and Embryology, Complutense University School of Medicine, Madrid 28040, Spain
| | - Santiago Canals
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas, Universidad Miguel Hernández, San Juan de Alicante 03550, Spain
| | - Manuel Gómez del Moral
- Department of Cell Biology, Complutense University School of Medicine, Madrid 28040, Spain
| | - Eduardo Martínez-Naves
- Department of Immunology, Ophthalmology and ORL, Complutense University School of Medicine, Madrid 28040, Spain
- Yulia A Nevzovova, Francisco Javier Cubero, 12 de Octubre Health Research Institute (imas12), Madrid 28041, Spain
| | - Yulia A Nevzorova
- Department of Genetics, Physiology and Microbiology, Faculty of Biology, Universidad Complutense, Madrid 28040, Spain
- Department of Internal Medicine III, University Hospital RWTH Aachen, Aachen 52062, Germany
| | - Francisco Javier Cubero
- Department of Immunology, Ophthalmology and ORL, Complutense University School of Medicine, Madrid 28040, Spain
| |
Collapse
|
22
|
Guo F, Zheng K, Benedé-Ubieto R, Cubero FJ, Nevzorova YA. The Lieber-DeCarli Diet-A Flagship Model for Experimental Alcoholic Liver Disease. Alcohol Clin Exp Res 2018; 42:1828-1840. [DOI: 10.1111/acer.13840] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 07/09/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Feifei Guo
- Department of Genetics, Physiology and Microbiology; Faculty of Biology; Complutense University of Madrid; Madrid Spain
| | - Kang Zheng
- Department of Immunology, Ophthalmology & ORL; School of Medicine; Complutense University of Madrid; Madrid Spain
- 12 de Octubre Health Research Institute (imas12); Madrid Spain
| | - Raquel Benedé-Ubieto
- Department of Genetics, Physiology and Microbiology; Faculty of Biology; Complutense University of Madrid; Madrid Spain
| | - Francisco Javier Cubero
- Department of Immunology, Ophthalmology & ORL; School of Medicine; Complutense University of Madrid; Madrid Spain
- 12 de Octubre Health Research Institute (imas12); Madrid Spain
| | - Yulia A. Nevzorova
- Department of Genetics, Physiology and Microbiology; Faculty of Biology; Complutense University of Madrid; Madrid Spain
- Department of Internal Medicine III; University Hospital RWTH Aachen; Aachen Germany
| |
Collapse
|
23
|
Inhibition of Caspase-8 does not protect from alcohol-induced liver apoptosis but alleviates alcoholic hepatic steatosis in mice. Cell Death Dis 2017; 8:e3152. [PMID: 29072704 PMCID: PMC5680911 DOI: 10.1038/cddis.2017.532] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 08/11/2017] [Accepted: 09/13/2017] [Indexed: 12/15/2022]
Abstract
Hepatic apoptosis is involved in the progression of alcoholic liver disease (ALD). Caspase-8, the apical initiator in death receptor-mediated apoptosis, has been implicated in acute liver injury and in non-alcoholic steatohepatitis. However, the relevance of Caspase-8 in the pathogenesis of ALD remains unclear. In the present study, we investigated the impact of Caspase-8 in human and murine alcohol-induced apoptosis and in ALD. We investigated human samples from ALD patients, primary mouse hepatocytes, and hepatocyte-specific Caspase-8 knockout (Casp8Δhepa) mice in acute and chronic models of ethanol (EtOH) administration. Caspase-8 activation was detected in liver biopsies from ALD patients, as well as in livers of wild-type (WT) mice after chronic ethanol feeding for 8 weeks using the Lieber-DeCarli model. Lack of Caspase-8 expression in Casp8Δhepa animals failed to prevent alcohol-induced liver damage and apoptosis. Instead, inhibition of Caspase-8 shifted the ethanol-induced death signals towards pronounced activation of the intrinsic, mitochondria-dependent apoptosis pathway in Casp8Δhepa livers involving enhanced release of cytochrome c, stronger Caspase-9 activation and specific morphological changes of mitochondria. In vitro and in vivo intervention using a pan-caspase inhibitor markedly attenuated alcohol-induced hepatocyte damage in a Caspase-8-independent manner. Surprisingly, EtOH-fed Casp8Δhepa mice displayed significantly attenuated steatosis and reduced hepatic triglyceride and free fatty acids content. Caspase-8 is dispensable for alcohol-induced apoptosis, but plays an unexpected role for alcohol-dependent fat metabolism. We provide evidence that simultaneous inhibition of extrinsic and intrinsic apoptosis signaling using pan-caspase inhibitors in vivo might be an optimal approach to treat alcohol-induced liver injury.
Collapse
|
24
|
Ramadori P, Cubero FJ, Liedtke C, Trautwein C, Nevzorova YA. Alcohol and Hepatocellular Carcinoma: Adding Fuel to the Flame. Cancers (Basel) 2017; 9:cancers9100130. [PMID: 28946672 PMCID: PMC5664069 DOI: 10.3390/cancers9100130] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 09/15/2017] [Accepted: 09/19/2017] [Indexed: 02/06/2023] Open
Abstract
Primary tumors of the liver represent the fifth most common type of cancer in the world and the third leading cause of cancer-related death. Case-control studies from different countries report that chronic ethanol consumption is associated with an approximately 2-fold increased odds ratio for hepatocellular carcinoma (HCC). Despite the substantial epidemiologic data in humans demonstrating that chronic alcohol consumption is a major risk factor for HCC development, the pathways causing alcohol-induced liver cancer are poorly understood. In this overview, we summarize the epidemiological evidence for the association between alcohol and liver cancer, review the genetic, oncogenic, and epigenetic factors that drive HCC development synergistically with ethanol intake and discuss the essential molecular and metabolic pathways involved in alcohol-induced liver tumorigenesis.
Collapse
Affiliation(s)
- Pierluigi Ramadori
- Department of Internal Medicine III, University Hospital RWTH Aachen, Pauwelsstrasse 30, D-52074 Aachen, Germany.
| | - Francisco Javier Cubero
- Department of Immunology, Complutense University School of Medicine, Madrid 28040, Spain.
- 13 de Octubre Health Research Institute (imas12), Madrid 28041, Spain.
| | - Christian Liedtke
- Department of Internal Medicine III, University Hospital RWTH Aachen, Pauwelsstrasse 30, D-52074 Aachen, Germany.
| | - Christian Trautwein
- Department of Internal Medicine III, University Hospital RWTH Aachen, Pauwelsstrasse 30, D-52074 Aachen, Germany.
| | - Yulia A Nevzorova
- Department of Internal Medicine III, University Hospital RWTH Aachen, Pauwelsstrasse 30, D-52074 Aachen, Germany.
- Department of Animal Physiology II, Faculty of Biology, Complutense University, Madrid 28040, Spain.
| |
Collapse
|
25
|
Barbhuiya MA, Mirando AC, Simons BW, Lemtiri-Chlieh G, Green JJ, Popel AS, Pandey NB, Tran PT. Therapeutic potential of an anti-angiogenic multimodal biomimetic peptide in hepatocellular carcinoma. Oncotarget 2017; 8:101520-101534. [PMID: 29254183 PMCID: PMC5731893 DOI: 10.18632/oncotarget.21148] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 08/26/2017] [Indexed: 12/13/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a major cause of cancer-related death worldwide. Due to inadequate screening methods and the common coexistence of limited functional liver reserves, curative treatment options are limited. Liver transplantation is the only curative treatment modality for early HCC. There are multidisciplinary treatment options like ablative treatments, radiation and systemic therapy available for more advanced patients or those that are inoperable. Treatment resistance and progression is inevitable for these HCC patients. Newer therapeutics need to be explored for better management of HCC. HCC is a hypervascular tumor and many pro-angiogenic proteins are found significantly overexpressed in HCC. Here we explored the therapeutic potential of the anti-angiogenic, anti-lymphangiogenic, and directly anti-tumorigenic biomimetic collagen IV-derived peptide developed by our group. Human HCC cell lines HuH7, Hep3b and HepG2 showed significant disruption of cell adhesion and migration upon treatment with the peptide. Consistent with previously described multimodal inhibitory properties, the peptide was found to inhibit both c-Met and IGF1R signaling in HepG2 cells and blocked HepG2 conditioned media stimulation of microvascular endothelial cell (MEC) tube formation. Furthermore, the peptide treatment of mouse HepG2 tumor xenografts significantly inhibited growth relative to untreated controls. The peptide was also found to improve the survival of autochthonous Myc-induced HCC in a transgenic mouse model. Mechanistically, we found that the peptide treatment reduced microvascular density in the autochthonous liver tumors with increased apoptosis. This study shows the promising therapeutic potential of our biomimetic peptide in the treatment of HCC.
Collapse
Affiliation(s)
- Mustafa A Barbhuiya
- Department of Radiation Oncology and Molecular and Radiation Sciences, Sidney Kimmel Comprehensive Cancer Centre, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Adam C Mirando
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Brian W Simons
- Department of Medical Oncology, Sidney Kimmel Comprehensive Cancer Centre and Department of Urology, The Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ghali Lemtiri-Chlieh
- Department of Radiation Oncology and Molecular and Radiation Sciences, Sidney Kimmel Comprehensive Cancer Centre, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Jordan J Green
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Aleksander S Popel
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Niranjan B Pandey
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Phuoc T Tran
- Department of Radiation Oncology and Molecular and Radiation Sciences, Sidney Kimmel Comprehensive Cancer Centre, Johns Hopkins School of Medicine, Baltimore, MD, USA.,Department of Medical Oncology, Sidney Kimmel Comprehensive Cancer Centre and Department of Urology, The Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
26
|
Li XJ, Mu YM, Qin QF, Zeng ZX, Li YS, Zhang WK, Tang HB, Tian GH, Shang HC. Chronic high-dosage fish oil exacerbates gut-liver axis injury in alcoholic steatohepatitis in mice: the roles of endotoxin and IL-4 in Kupffer cell polarization imbalance. Toxicol Res (Camb) 2017; 6:611-620. [PMID: 30090529 DOI: 10.1039/c7tx00037e] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 07/06/2017] [Indexed: 01/21/2023] Open
Abstract
In the present study, intestinal tight junctions (TJs) and Kupffer cell polarization were investigated in an alcoholic steatohepatitis (ASH) mouse model to uncover the potential side effects of overexposure to fish oil or omega-3 fatty acids. The mice were fed ad libitum with a liquid diet containing ethanol and fish oil. In the meantime, ethanol was given every 5-7 days by gavage to simulate binge drinking. After the 7th binge, steatosis, necrosis, inflammatory infiltration, and bridging fibrosis were observed in the liver by histological staining. After the 13th binge, the inducers, markers and other downstream genes/proteins of the Kupffer cell M1/M2 phenotype in the liver, serum, and small intestine were analysed. The results suggested that a chronic high dosage of fish oil alone reduced the mRNA levels of most genes tested and showed a tendency to damage the intestinal zonula occludens-1 localization and reduce the number of M2 Kupffer cells. Meanwhile, the combination of fish oil and ethanol damaged the intestinal TJs, resulting in an increased endotoxin level in the liver. Gut-derived endotoxin polarized Kupffer cells to the M1 phenotype, whereas the number of cells with the M2 phenotype (markers: CD163 and CD206) was decreased. Interleukin-4 (IL-4), an M2 Kupffer cell inducer, was also decreased. Moreover, in vitro experiments showed that IL-4 reversed eicosapentaenoic acid-induced CD163 and CD206 mRNA suppression in RAW 264.7 cells. Overall, our results showed that a chronic high dosage of fish oil exacerbated gut-liver axis injury in alcoholic liver disease in mice, and endotoxin/IL-4-induced Kupffer cell polarization imbalance might play an important role in that process.
Collapse
Affiliation(s)
- Xiao-Jun Li
- Department of Pharmacology , School of Pharmaceutical Sciences , South-Central University for Nationalities , No. 182 Minyuan Road , 430074 , Wuhan , China . ; ; Tel: +86 27 6784 2332
| | - Yun-Mei Mu
- Department of Pharmacology , School of Pharmaceutical Sciences , South-Central University for Nationalities , No. 182 Minyuan Road , 430074 , Wuhan , China . ; ; Tel: +86 27 6784 2332
| | - Qiu-Fang Qin
- Department of Pharmacology , School of Pharmaceutical Sciences , South-Central University for Nationalities , No. 182 Minyuan Road , 430074 , Wuhan , China . ; ; Tel: +86 27 6784 2332
| | - Zi-Xuan Zeng
- Department of Pharmacology , School of Pharmaceutical Sciences , South-Central University for Nationalities , No. 182 Minyuan Road , 430074 , Wuhan , China . ; ; Tel: +86 27 6784 2332
| | - Yu-Sang Li
- Department of Pharmacology , School of Pharmaceutical Sciences , South-Central University for Nationalities , No. 182 Minyuan Road , 430074 , Wuhan , China . ; ; Tel: +86 27 6784 2332
| | - Wei Kevin Zhang
- Department of Pharmacology , School of Pharmaceutical Sciences , South-Central University for Nationalities , No. 182 Minyuan Road , 430074 , Wuhan , China . ; ; Tel: +86 27 6784 2332
| | - He-Bin Tang
- Department of Pharmacology , School of Pharmaceutical Sciences , South-Central University for Nationalities , No. 182 Minyuan Road , 430074 , Wuhan , China . ; ; Tel: +86 27 6784 2332.,Key Laboratory of Chinese Internal Medicine of MOE and Beijing , Dongzhimen Hospital , Beijing University of Chinese Medicine , 100029 , Beijing , China . ; ; Tel: +86 10 8401 2510.,Research Institute of Huazhong University of Science and Technology in Shenzhen , 518057 , Shenzhen , China
| | - Gui-Hua Tian
- Key Laboratory of Chinese Internal Medicine of MOE and Beijing , Dongzhimen Hospital , Beijing University of Chinese Medicine , 100029 , Beijing , China . ; ; Tel: +86 10 8401 2510
| | - Hong-Cai Shang
- Key Laboratory of Chinese Internal Medicine of MOE and Beijing , Dongzhimen Hospital , Beijing University of Chinese Medicine , 100029 , Beijing , China . ; ; Tel: +86 10 8401 2510
| |
Collapse
|
27
|
c-MYC-Making Liver Sick: Role of c-MYC in Hepatic Cell Function, Homeostasis and Disease. Genes (Basel) 2017; 8:genes8040123. [PMID: 28422055 PMCID: PMC5406870 DOI: 10.3390/genes8040123] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 03/30/2017] [Accepted: 04/12/2017] [Indexed: 12/20/2022] Open
Abstract
Over 35 years ago, c-MYC, a highly pleiotropic transcription factor that regulates hepatic cell function, was identified. In recent years, a considerable increment in the number of publications has significantly shifted the way that the c-MYC function is perceived. Overexpression of c-MYC alters a wide range of roles including cell proliferation, growth, metabolism, DNA replication, cell cycle progression, cell adhesion and differentiation. The purpose of this review is to broaden the understanding of the general functions of c-MYC, to focus on c-MYC-driven pathogenesis in the liver, explain its mode of action under basal conditions and during disease, and discuss efforts to target c-MYC as a plausible therapy for liver disease.
Collapse
|
28
|
Akinyeke T, Weber SJ, Davenport AT, Baker EJ, Daunais JB, Raber J. Effects of alcohol on c-Myc protein in the brain. Behav Brain Res 2016; 320:356-364. [PMID: 27832980 DOI: 10.1016/j.bbr.2016.11.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 10/28/2016] [Accepted: 11/04/2016] [Indexed: 12/18/2022]
Abstract
Alcoholism is a disorder categorized by significant impairment that is directly related to persistent and extreme use of alcohol. The effects of alcoholism on c-Myc protein expression in the brain have been scarcely studied. This is the first study to investigate the role different characteristics of alcoholism have on c-Myc protein in the brain. We analyzed c-Myc protein in the hypothalamus and amygdala from five different animal models of alcohol abuse. c-Myc protein was increased following acute ethanol exposure in a mouse knockout model and following chronic ethanol consumption in vervet monkeys. We also observed increases in c-Myc protein exposure in animals that are genetically predisposed to alcohol and methamphetamine abuse. Lastly, c-Myc protein was increased in animals that were acutely exposed to methamphetamine when compared to control treated animals. These results suggest that in substance abuse c-Myc plays an important role in the brain's response.
Collapse
Affiliation(s)
- Tunde Akinyeke
- Department of Behavioral Neuroscience, ONPRC, Oregon Health and Science University, Portland, OR 97239, United States
| | - Sydney J Weber
- Department of Behavioral Neuroscience, ONPRC, Oregon Health and Science University, Portland, OR 97239, United States
| | - April T Davenport
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston Salem, NC 27106, United States
| | - Erich J Baker
- School of Engineering and Department of Computer Science, Baylor University Waco, TX 76978, United States
| | - James B Daunais
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston Salem, NC 27106, United States
| | - Jacob Raber
- Department of Behavioral Neuroscience, ONPRC, Oregon Health and Science University, Portland, OR 97239, United States; Departments of Neurology and Radiation Medicine, Division of Neuroscience, ONPRC, Oregon Health and Science University, Portland, OR 97239, United States.
| |
Collapse
|