1
|
Long J, Xu Y, Zhang X, Wu B, Wang C. Role of FXR in the development of NAFLD and intervention strategies of small molecules. Arch Biochem Biophys 2024; 757:110024. [PMID: 38703803 DOI: 10.1016/j.abb.2024.110024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 04/18/2024] [Accepted: 04/30/2024] [Indexed: 05/06/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD) remains a prevailing etiological agent behind hepatocyte diseases like chronic liver disease. The spectrum of processes involved in NAFLD stages includes hepatic steatosis, non-alcoholic fatty liver, and non-alcoholic steatohepatitis (NASH). Without intervention, the progression of NASH can further deteriorate into cirrhosis and ultimately, hepatocellular carcinoma. The cardinal features that characterize NAFLD are insulin resistance, lipogenesis, oxidative stress and inflammation, extracellular matrix deposition and fibrosis. Due to its complex pathogenesis, existing pharmaceutical agents fail to take a curative or ameliorative effect on NAFLD. Consequently, it is imperative to identify novel therapeutic targets and strategies for NAFLD, ideally to improve the aforementioned key features in patients. As an enterohepatic regulator of bile acid homeostasis, lipid metabolism, and inflammation, FarnesoidX receptor (FXR) is an important pharmacological target for the treatment of NAFLD. Manipulating FXR to regulate lipid metabolic signaling pathways is a potential mechanism to mitigate NAFLD. Therefore, elucidating the modulatory character of FXR in regulating lipid metabolism in NAFLD has the potential to yield groundbreaking perspectives for drug design. This review details recent advances in the regulation of lipid depletion in hepatocytes and investigates the pivotal function of FXR in the progress of NAFLD.
Collapse
Affiliation(s)
- Jiachan Long
- State Key Laboratory of Traditional Chinese Medicine Syndrome, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Yuanhang Xu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Xuerong Zhang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Bingxing Wu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Caiyan Wang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| |
Collapse
|
2
|
Engin A. Protein Kinases in Obesity, and the Kinase-Targeted Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:199-229. [PMID: 39287853 DOI: 10.1007/978-3-031-63657-8_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
The action of protein kinases and protein phosphatases is essential for multiple physiological responses. Each protein kinase displays its own unique substrate specificity and a regulatory mechanism that may be modulated by association with other proteins. Protein kinases are classified as dual-specificity kinases and dual-specificity phosphatases. Dual-specificity phosphatases are important signal transduction enzymes that regulate various cellular processes in coordination with protein kinases and play an important role in obesity. Impairment of insulin signaling in obesity is largely mediated by the activation of the inhibitor of kappa B-kinase beta and the c-Jun N-terminal kinase (JNK). Oxidative stress and endoplasmic reticulum (ER) stress activate the JNK pathway which suppresses insulin biosynthesis. Adenosine monophosphate (AMP)-activated protein kinase (AMPK) and mammalian target of rapamycin (mTOR) are important for proper regulation of glucose metabolism in mammals at both the hormonal and cellular levels. Additionally, obesity-activated calcium/calmodulin dependent-protein kinase II/p38 suppresses insulin-induced protein kinase B phosphorylation by activating the ER stress effector, activating transcription factor-4. To alleviate lipotoxicity and insulin resistance, promising targets are pharmacologically inhibited. Nifedipine, calcium channel blocker, stimulates lipogenesis and adipogenesis by downregulating AMPK and upregulating mTOR, which thereby enhances lipid storage. Contrary to the nifedipine, metformin activates AMPK, increases fatty acid oxidation, suppresses fatty acid synthesis and deposition, and thus alleviates lipotoxicity. Obese adults with vascular endothelial dysfunction have greater endothelial cells activation of unfolded protein response stress sensors, RNA-dependent protein kinase-like ER eukaryotic initiation factor-2 alpha kinase (PERK), and activating transcription factor-6. The transcriptional regulation of adipogenesis in obesity is influenced by AGC (protein kinase A (PKA), PKG, PKC) family signaling kinases. Obesity may induce systemic oxidative stress and increase reactive oxygen species in adipocytes. An increase in intracellular oxidative stress can promote PKC-β activation. Activated PKC-β induces growth factor adapter Shc phosphorylation. Shc-generated peroxides reduce mitochondrial oxygen consumption and enhance triglyceride accumulation and lipotoxicity. Liraglutide attenuates mitochondrial dysfunction and reactive oxygen species generation. Co-treatment of antiobesity and antidiabetic herbal compound, berberine with antipsychotic drug olanzapine decreases the accumulation of triglyceride. While low-dose rapamycin, metformin, amlexanox, thiazolidinediones, and saroglitazar protect against insulin resistance, glucagon-like peptide-1 analog liraglutide inhibits palmitate-induced inflammation by suppressing mTOR complex 1 (mTORC1) activity and protects against lipotoxicity.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
3
|
Fang X, Song J, Zhou K, Zi X, Sun B, Bao H, Li L. Molecular Mechanism Pathways of Natural Compounds for the Treatment of Non-Alcoholic Fatty Liver Disease. Molecules 2023; 28:5645. [PMID: 37570615 PMCID: PMC10419790 DOI: 10.3390/molecules28155645] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 07/21/2023] [Accepted: 07/21/2023] [Indexed: 08/13/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease in the world, and its incidence continues to increase each year. Yet, there is still no definitive drug that can stop its development. This review focuses mainly on lipotoxicity, oxidative stress, inflammation, and intestinal flora dysbiosis to understand NAFLD's pathogenesis. In this review, we used NCBI's PubMed database for retrieval, integrating in vivo and in vitro experiments to reveal the therapeutic effects of natural compounds on NAFLD. We also reviewed the mechanisms by which the results of these experiments suggest that these compounds can protect the liver from damage by modulating inflammation, reducing oxidative stress, decreasing insulin resistance and lipid accumulation in the liver, and interacting with the intestinal microflora. The natural compounds discussed in these papers target a variety of pathways, such as the AMPK pathway and the TGF-β pathway, and have significant therapeutic effects. This review aims to provide new possible therapeutic lead compounds and references for the development of novel medications and the clinical treatment of NAFLD. It offers fresh perspectives on the development of natural compounds in preventing and treating NAFLD.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Lijing Li
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China; (X.F.)
| |
Collapse
|
4
|
Xie J, Chen Q, Zhao Y, Luo M, Zeng X, Qin L, Tan D, He Y. Transcriptome Sequencing Reveals Autophagy Networks in Rat Livers during the Development of NAFLD and Identifies Autophagy Hub Genes. Int J Mol Sci 2023; 24:ijms24076437. [PMID: 37047411 PMCID: PMC10094595 DOI: 10.3390/ijms24076437] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/21/2023] [Accepted: 03/28/2023] [Indexed: 04/14/2023] Open
Abstract
(1) Autophagy is an important biological process in cells and is closely associated with the development and progression of non-alcoholic fatty liver disease (NAFLD). Therefore, this study aims to investigate the biological function of the autophagy hub genes, which could be used as a potential therapeutic target and diagnostic markers for NAFLD. (2) Male C57BL/6J mice were sacrificed after 16 and 38 weeks of a high-fat diet, serum biochemical indexes were detected, and liver lobules were collected for pathological observation and transcriptome sequencing. The R software was used to identify differentially expressed autophagy genes (DEGs) from the transcriptome sequencing data of mice fed with a normal diet for 38 weeks (ND38) and a high-fat diet for 38 weeks (HFD38). Gene ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis were performed on the DEGs, a protein-protein interaction (PPI) network of the DEGs was established using the STRING data website, and the results were visualized through Cytoscape. (3) After 16 weeks and 38 weeks of a high-fat diet, there was a significant increase in body weight, serum total cholesterol (TC), low-density lipoprotein-cholesterol (LDL-C) and triglycerides (TG) in mice, along with lipid accumulation in the liver, which was more severe at 38 weeks than at 16 weeks. The transcriptome data showed significant changes in the expression profile of autophagy genes in the livers of NAFLD mice following a long-term high-fat diet. Among the 31 differentially expressed autophagy-related genes, 13 were upregulated and 18 were downregulated. GO and KEGG pathway analysis revealed that these DEGs were primarily involved in autophagy, cholesterol transport, triglyceride metabolism, apoptosis, the FoxO signaling pathway, the p53 signaling pathway and the IL-17 signaling pathway. Four hub genes were identified by the PPI network analysis, of which Irs2, Pnpla2 and Plin2 were significantly downregulated, while Srebf2 was significantly upregulated by the 38-week high-fat diet. (4) The hub genes Irs2, Pnpla2, Srebf2 and Plin2 may serve as key therapeutic targets and early diagnostic markers in the progression of NAFLD.
Collapse
Affiliation(s)
- Jian Xie
- Guizhou Engineering Research Center of Industrial Key-Technology for Dendrobium Nobile, Zunyi Medical University, Zunyi 563000, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563000, China
- 2011 Cooperative Inovational Center for Guizhou Traditional Chinese Medicine and Ethnic Medicine, Zunyi Medical University, Zunyi 563000, China
- Department of Medical Genetics, Zunyi Medical University, Zunyi 563000, China
| | - Qiuyi Chen
- Guizhou Engineering Research Center of Industrial Key-Technology for Dendrobium Nobile, Zunyi Medical University, Zunyi 563000, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563000, China
- 2011 Cooperative Inovational Center for Guizhou Traditional Chinese Medicine and Ethnic Medicine, Zunyi Medical University, Zunyi 563000, China
| | - Yongxia Zhao
- Guizhou Engineering Research Center of Industrial Key-Technology for Dendrobium Nobile, Zunyi Medical University, Zunyi 563000, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563000, China
- 2011 Cooperative Inovational Center for Guizhou Traditional Chinese Medicine and Ethnic Medicine, Zunyi Medical University, Zunyi 563000, China
| | - Mingxia Luo
- Guizhou Engineering Research Center of Industrial Key-Technology for Dendrobium Nobile, Zunyi Medical University, Zunyi 563000, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563000, China
- 2011 Cooperative Inovational Center for Guizhou Traditional Chinese Medicine and Ethnic Medicine, Zunyi Medical University, Zunyi 563000, China
| | - Xin Zeng
- Department of Medical Genetics, Zunyi Medical University, Zunyi 563000, China
| | - Lin Qin
- Guizhou Engineering Research Center of Industrial Key-Technology for Dendrobium Nobile, Zunyi Medical University, Zunyi 563000, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563000, China
- 2011 Cooperative Inovational Center for Guizhou Traditional Chinese Medicine and Ethnic Medicine, Zunyi Medical University, Zunyi 563000, China
| | - Daopeng Tan
- Guizhou Engineering Research Center of Industrial Key-Technology for Dendrobium Nobile, Zunyi Medical University, Zunyi 563000, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563000, China
- 2011 Cooperative Inovational Center for Guizhou Traditional Chinese Medicine and Ethnic Medicine, Zunyi Medical University, Zunyi 563000, China
| | - Yuqi He
- Guizhou Engineering Research Center of Industrial Key-Technology for Dendrobium Nobile, Zunyi Medical University, Zunyi 563000, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563000, China
- 2011 Cooperative Inovational Center for Guizhou Traditional Chinese Medicine and Ethnic Medicine, Zunyi Medical University, Zunyi 563000, China
| |
Collapse
|
5
|
Omar IS, Abd Jamil AH, Mat Adenan NA, Chung I. MPA alters metabolic phenotype of endometrial cancer-associated fibroblasts from obese women via IRS2 signaling. PLoS One 2022; 17:e0270830. [PMID: 35816477 PMCID: PMC9273069 DOI: 10.1371/journal.pone.0270830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 06/20/2022] [Indexed: 11/18/2022] Open
Abstract
Obese women have a higher risk of developing endometrial cancer (EC) than lean women. Besides affecting EC progression, obesity also affects sensitivity of patients to treatment including medroxprogesterone acetate (MPA). Obese women have a lower response to MPA with an increased risk for tumor recurrence. While MPA inhibits the growth of normal fibroblasts, human endometrial cancer-associated fibroblasts (CAFs) were reported to be less responsive to MPA. However, it is still unknown how CAFs from obese women respond to progesterone. CAFs from the EC tissues of obese (CO) and non-obese (CN) women were established as primary cell models. MPA increased cell proliferation and downregulated stromal differentiation genes, including BMP2 in CO than in CN. Induction of IRS2 (a BMP2 regulator) mRNA expression by MPA led to activation of glucose metabolism in CO, with evidence of greater mRNA levels of GLUT6, GAPDH, PKM2, LDHA, and increased in GAPDH enzymatic activity. Concomitantly, MPA increased the mRNA expression of a fatty acid transporter, CD36 and lipid droplet formation in CO. MPA-mediated increase in glucose metabolism genes in CO was reversed with a progesterone receptor inhibitor, mifepristone (RU486), leading to a decreased proliferation. Our data suggests that PR signaling is aberrantly activated by MPA in CAFs isolated from endometrial tissues of obese women, leading to activation of IRS2 and glucose metabolism, which may lead to lower response and sensitivity to progesterone in obese women.
Collapse
Affiliation(s)
- Intan Sofia Omar
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
- Universiti Malaya Cancer Research Institute, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Amira Hajirah Abd Jamil
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Noor Azmi Mat Adenan
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
- Department of Obstetrics and Gynaecology, Ara Damansara and Subang Jaya Medical Center, Ramsay Sime Darby Health Care, Subang Jaya, Selangor, Malaysia
| | - Ivy Chung
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, Universiti Malaya, Kuala Lumpur, Malaysia
- * E-mail:
| |
Collapse
|
6
|
Sabir U, Irfan HM, Alamgeer, Umer I, Niazi ZR, Asjad HMM. Phytochemicals targeting NAFLD through modulating the dual function of forkhead box O1 (FOXO1) transcription factor signaling pathways. Naunyn Schmiedebergs Arch Pharmacol 2022; 395:741-755. [PMID: 35357518 DOI: 10.1007/s00210-022-02234-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 03/18/2022] [Indexed: 02/06/2023]
Abstract
Literature evidence reveals that natural compounds are potential candidates for ameliorating obesity-associated non-alcoholic fatty liver disease (NAFLD) by targeting forkhead box O1 (FOXO1) transcription factor. FOXO1 has a dual and complex role in regulating both increase and decrease in lipid accumulation in hepatocytes and adipose tissues (AT) at different stages of NAFLD. In insulin resistance (IR), it is constitutively expressed, resulting in increased hepatic glucose output and lipid metabolism irregularity. The studies on different phytochemicals indicate that dysregulation of FOXO1 causes disturbance in cellular nutrients homeostasis, and the natural entities have an enduring impact on the mitigation of these abnormalities. The current review communicates and evaluates certain phytochemicals through different search engines, targeting FOXO1 and its downstream cellular pathways to find lead compounds as potential therapeutic agents for treating NAFLD and related metabolic disorders. The findings of this review confirm that polyphenols, flavonoids, alkaloids, terpenoids, and anthocyanins are capable of modulating FOXO1 and associated signaling pathways, and they are potential therapeutic agents for NAFLD and related complications. HIGHLIGHTS: • FOXO1 has the potential to be targeted by novel drugs from natural sources for the treatment of NAFLD and obesity. • FOXO1 regulates cellular autophagy, inflammation, oxidative stress, and lipogenesis through alternative mechanisms. • Phytochemicals treat NAFLD by acting on FOXO1 or SREBP1c and PPARγ transcription factor signaling pathways.
Collapse
Affiliation(s)
- Usman Sabir
- Department of Pharmacology, College of Pharmacy, University of Sargodha, Sargodha, Pakistan
| | - Hafiz Muhammad Irfan
- Department of Pharmacology, College of Pharmacy, University of Sargodha, Sargodha, Pakistan.
| | - Alamgeer
- Punjab University College of Pharmacy, University of the Punjab Lahore, Lahore, Pakistan
| | - Ihtisham Umer
- Pharmacy Department, Comsat International University Lahore Campus, Lahore, Pakistan
| | | | | |
Collapse
|
7
|
Di Vincenzo O, Marra M, Sacco AM, Pasanisi F, Scalfi L. Bioelectrical impedance (BIA)-derived phase angle in adults with obesity: A systematic review. Clin Nutr 2021; 40:5238-5248. [PMID: 34474193 DOI: 10.1016/j.clnu.2021.07.035] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 07/05/2021] [Accepted: 07/28/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Bioimpedance analysis-derived phase angle (PhA), as marker of body cell mass and cell integrity, might be altered in obesity, a condition which is characterized by alterations in muscle structure and function. The aim of this systematic review was to evaluate whether and to which extent PhA varies in individuals/patients with excess body weight focusing on: a) changes in PhA due to obesity; b) changes in PhA after bariatric interventions or training programs. METHODS According to PRISMA criteria, a systematic literature search until February 2021 using PubMed, Embase, Scopus, and Web of Science was performed. Selection criteria included studies on patients with obesity without comorbidities other than metabolic diseases. RESULTS A total of 278 articles were first identified. After removing duplicates and excluding studies that did not fulfil the inclusion criteria, the full text of the remaining 80 potentially relevant studies was examined to finally retrieve 11 cross-sectional and 10 longitudinal studies. Few studies have shown that PhA is lower in individuals/patients with obesity than in controls. The only study on the matter showed a decrease with age after the fourth decade of life. Four out of five studies reported consistently greater mean values in men than in women. In two studies PhA was lower in patients with severe obesity; in addition, Patients with low PhA had higher BMI and PhA was inversely correlated with fat mass. Longitudinal studies showed that PhA markedly decreased after bariatric surgery and slightly increased after training programs. CONCLUSIONS Thus, a relatively low number of studies have evaluated PhA in individuals/patients with obesity with sometimes contradictory and preliminary results. PhA might be useful to evaluate muscle quality in individuals/patients with obesity but further studies are needed to more accurately associate this variable with changes in muscle structure and strength, as well as in metabolic functions.
Collapse
Affiliation(s)
- Olivia Di Vincenzo
- Department of Clinical Medicine and Surgery, Federico II University of Naples, via S. Pansini 5, 80131, Naples, Italy; Casa di Cura Santa Maria del Pozzo, via Pomigliano 40, 80049, Somma Vesuviana (NA), Italy.
| | - Maurizio Marra
- Department of Clinical Medicine and Surgery, Federico II University of Naples, via S. Pansini 5, 80131, Naples, Italy
| | - Anna Maria Sacco
- Department of Public Health, Federico II University of Naples, via S. Pansini 5, 80131, Naples, Italy
| | - Fabrizio Pasanisi
- Department of Clinical Medicine and Surgery, Federico II University of Naples, via S. Pansini 5, 80131, Naples, Italy
| | - Luca Scalfi
- Department of Public Health, Federico II University of Naples, via S. Pansini 5, 80131, Naples, Italy; Casa di Cura Santa Maria del Pozzo, via Pomigliano 40, 80049, Somma Vesuviana (NA), Italy
| |
Collapse
|
8
|
Chhimwal J, Patial V, Padwad Y. Beverages and Non-alcoholic fatty liver disease (NAFLD): Think before you drink. Clin Nutr 2021; 40:2508-2519. [PMID: 33932796 DOI: 10.1016/j.clnu.2021.04.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 03/26/2021] [Accepted: 04/03/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND & AIMS Beverages and Non-alcoholic fatty liver disease (NAFLD) both the terms are associated with westernized diet and sedentary lifestyle. Throughout recent decades, dietary changes have boosted demand of beverages to meet the liquid consumption needs, among which rising consumption of several calorie-rich beverages have increased the risk of fatty liver disease. Meanwhile, certain beverages have capacity to deliver many unanticipated health benefits thereby reducing the burden of NAFLD and metabolic diseases. The present review therefore addresses the increasing interconnections between beverages intake among population, dietary patterns and the overall effect of these beverage on the development and prevention of NAFLD. Methods In the present review, some frequently consumed beverage groups have been analyzed in light of their role in the advancement and prevention of NAFLD, including sugar sweetened, hot and alcoholic beverages. The nutritional composition of different beverages makes the progression of NAFLD distinctive. RESULTS The ingestion of sugar-rich beverages has demonstrated the metabolic burden and in all cases, raises the risk of NAFLD, while intake of coffee and tea has decreased this risk without any significant adverse effects. In some cases, low to moderate alcohol intake has been shown to minimize the risk of advanced fibrosis and NAFLD-mortality. CONCLUSION Together, this review discusses and supports work on new dietary approaches and clinical studies to accomplish nutrition-oriented NAFLD care by improving the drinking habits.
Collapse
Affiliation(s)
- Jyoti Chhimwal
- Pharmacology and Toxicology Laboratory, Dietetics & Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, 176061, H.P., India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, U.P., India
| | - Vikram Patial
- Pharmacology and Toxicology Laboratory, Dietetics & Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, 176061, H.P., India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, U.P., India
| | - Yogendra Padwad
- Pharmacology and Toxicology Laboratory, Dietetics & Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, 176061, H.P., India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, U.P., India.
| |
Collapse
|
9
|
Zhang J, Choudhury M. Benzyl Butyl Phthalate Induced Early lncRNA H19 Regulation in C3H10T1/2 Stem Cell Line. Chem Res Toxicol 2021; 34:54-62. [PMID: 33395283 DOI: 10.1021/acs.chemrestox.0c00129] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Exposure to endocrine-disrupting chemicals used in plastic manufacturing may contribute to the current obesity and diabetes epidemic. Our previous study demonstrated that benzyl butyl phthalate (BBP) induced adipogenesis in the C3H10T1/2 stem cell line. Here we investigated if BBP deregulated long noncoding RNA H19 and its downstream pathway and whether BBP plays a role in the insulin signaling pathway during adipocyte diiferentiation. Cells treated with an 8 day BBP regimen showed that H19 expression was decreased at day 2 with 50 μM BBP exposure (p < 0.05). However, no significant changes were observed from day 4 to day 8. Expression of miRNA-103/107, H19 regulated miRNAs, was upregulated at day 2 (p < 0.05) but not from day 4 to day 8. Similarly, expression of the let-7 family members (a, b, c, d, f, and g) was also significantly increased at day 2 (p < 0.05 or p < 0.01), except for let-7e. Both let-7 and miRNA-103/107 are targets of H19 and play roles in insulin signaling. Insulin receptor substrate (IRS)-1, one of the key insulin signal transduction regulators, was significantly downregulated from day 2 to day 8 (p < 0.05). Gene expression of insulin receptor (IR) and IRS-2 were not altered by BBP exposure. The ratio of IRS1/IRS2 was significantly decreased from day 2 to day 8. On day 4, phospho-Akt protein expression was significantly decreased (p < 0.05). In conclusion, BBP exposure may lead to metabolic dysregulation by altering vital epigenetic regulators such as lncRNA H19 and its target microRNAs at an earlier stage, which further regulates insulin signaling.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Pharmaceutical Sciences, Rangel College of Pharmacy, Texas A&M University, College Station, 77843-1114 TX, United States of America
| | - Mahua Choudhury
- Department of Pharmaceutical Sciences, Rangel College of Pharmacy, Texas A&M University, College Station, 77843-1114 TX, United States of America
| |
Collapse
|
10
|
Krause C, Geißler C, Tackenberg H, El Gammal AT, Wolter S, Spranger J, Mann O, Lehnert H, Kirchner H. Multi-layered epigenetic regulation of IRS2 expression in the liver of obese individuals with type 2 diabetes. Diabetologia 2020; 63:2182-2193. [PMID: 32710190 PMCID: PMC7476982 DOI: 10.1007/s00125-020-05212-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 04/30/2020] [Indexed: 12/15/2022]
Abstract
AIMS/HYPOTHESIS IRS2 is an important molecular switch that mediates insulin signalling in the liver. IRS2 dysregulation is responsible for the phenomenon of selective insulin resistance that is observed in type 2 diabetes. We hypothesise that epigenetic mechanisms are involved in the regulation of IRS2 in the liver of obese and type 2 diabetic individuals. METHODS DNA methylation of seven CpG sites was studied by bisulphite pyrosequencing and mRNA and microRNA (miRNA) expression was assessed by quantitative real-time PCR in liver biopsies of 50 obese non-diabetic and 31 obese type 2 diabetic participants, in a cross-sectional setting. Methylation-sensitive luciferase assays and electrophoretic mobility shift assays were performed. Furthermore, HepG2 cells were treated with insulin and high glucose concentrations to induce miRNA expression and IRS2 downregulation. RESULTS We found a significant downregulation of IRS2 expression in the liver of obese individuals with type 2 diabetes (0.84 ± 0.08-fold change; p = 0.0833; adjusted p value [pa] = 0.0417; n = 31) in comparison with non-diabetic obese participants (n = 50). This downregulation correlated with hepatic IRS2 DNA methylation at CpG5. Additionally, CpG6, which is located in intron 1 of IRS2, was hypomethylated in type 2 diabetes; this site spans the sterol regulatory element binding transcription factor 1 (SREBF1) recognition motif, which likely acts as transcriptional repressor. The adjacent polymorphism rs4547213 (G>A) was significantly associated with DNA methylation at a specificity-protein-1 (SP1) binding site (CpG3). Moreover, DNA methylation of cg25924746, a CpG site located in the shore region of the IRS2 promoter-associated CpG island, was increased in the liver of individuals with type 2 diabetes, as compared with those without diabetes. A second epigenetic mechanism, upregulation of hepatic miRNA hsa-let-7e-5p (let-7e-5p) in obese individuals with type 2 diabetes (n = 29) vs non-diabetic obese individuals (n = 49) (1.2 ± 0.08-fold change; p = 0.0332; pa = 0.0450), is likely to act synergistically with altered IRS2 DNA methylation to decrease IRS2 expression. Mechanistic in vitro experiments demonstrated an acute upregulation of let-7e-5p expression and simultaneous IRS2 downregulation in a liver (HepG2) cell line upon hyperinsulinaemic and hyperglycaemic conditions. CONCLUSIONS/INTERPRETATION Our study highlights a new multi-layered epigenetic network that could be involved in subtle dysregulation of IRS2 in the liver of individuals with type 2 diabetes. This might lead to fine-tuning of IRS2 expression and is likely to be supplementary to the already known factors regulating IRS2 expression. Thereby, our findings could support the discovery of new diagnostic and therapeutic strategies for type 2 diabetes. Graphical abstract.
Collapse
Affiliation(s)
- Christin Krause
- First Department of Medicine, Division of Epigenetics and Metabolism, University of Lübeck, Ratzeburger Allee 160, 23562, Lübeck, Germany
| | - Cathleen Geißler
- First Department of Medicine, Division of Epigenetics and Metabolism, University of Lübeck, Ratzeburger Allee 160, 23562, Lübeck, Germany
| | - Heidi Tackenberg
- First Department of Medicine, Division of Epigenetics and Metabolism, University of Lübeck, Ratzeburger Allee 160, 23562, Lübeck, Germany
| | - Alexander T El Gammal
- Department of General, Visceral and Thoracic Surgery, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Wolter
- Department of General, Visceral and Thoracic Surgery, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Joachim Spranger
- Department of Endocrinology and Metabolism, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Oliver Mann
- Department of General, Visceral and Thoracic Surgery, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Hendrik Lehnert
- First Department of Medicine, Division of Epigenetics and Metabolism, University of Lübeck, Ratzeburger Allee 160, 23562, Lübeck, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Henriette Kirchner
- First Department of Medicine, Division of Epigenetics and Metabolism, University of Lübeck, Ratzeburger Allee 160, 23562, Lübeck, Germany.
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany.
| |
Collapse
|
11
|
Wang Z, Li S, Wang R, Guo L, Xu D, Zhang T, Xu Y, Wang W, Wang M, Gan Z, Wang X. The protective effects of the β3 adrenergic receptor agonist BRL37344 against liver steatosis and inflammation in a rat model of high-fat diet-induced nonalcoholic fatty liver disease (NAFLD). Mol Med 2020; 26:54. [PMID: 32503411 PMCID: PMC7275314 DOI: 10.1186/s10020-020-00164-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 03/30/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Our objective was to investigate the efficacy of the beta-3 adrenergic receptor (β3-AR) agonist BRL37344 for the prevention of liver steatosis and inflammation associated with nonalcoholic fatty liver disease (NAFLD). METHODS Four groups were established: a control group (given a standard diet), a high-fat diet (HFD) group, an HFD + β3-AR agonist (β3-AGO) group, and an HFD + β3-AR antagonist (β3-ANT) group. All rats were fed for 12 weeks. The β3-AR agonist BRL37344 and the antagonist L748337 were administered for the last 4 weeks with Alzet micro-osmotic pumps. The rat body weights (g) were measured at the end of the 4th, 8th, and 12th weeks. At the end of the 12th week, the liver weights were measured. Serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) were analyzed with a Hitachi automatic analyzer. The lipid levels of the triglycerides (TGs), total cholesterol (TC), and low-density lipoprotein cholesterol (LDL-C) and the concentrations of free fatty acids (FFAs) were also measured. An oil red O kit was used to detect lipid droplet accumulation in hepatocytes. Steatosis, ballooning degeneration and inflammation were histopathologically determined. The protein and mRNA expression levels of β3-AR, peroxisome proliferator-activated receptor-alpha (PPAR-α), peroxisome proliferator-activated receptor-gamma (PPAR-γ), mitochondrial carnitine palmitoyltransferase-1 (mCPT-1), and fatty acid translocase (FAT)/CD36 were measured by western blot analysis and RT-qPCR, respectively. RESULTS After treatment with the β3-AR agonist BRL37344 for 4 weeks, the levels of ALT, AST, TGs, TC, LDL-C and FFAs were decreased in the NAFLD model group compared with the HFD group. Body and liver weights, liver index values and lipid droplet accumulation were lower in the HFD + β3-AGO group than in the HFD group. Decreased NAFLD activity scores (NASs) also showed that liver steatosis and inflammation were ameliorated after treatment with BRL37344. Moreover, the β3-AR antagonist L748337 reversed these effects. Additionally, the protein and gene expression levels of β3-AR, PPAR-α, and mCPT-1 were increased in the HFD + β3-AGO group, whereas those of PPAR-γ and FAT/CD36 were decreased. CONCLUSION The β3-AR agonist BRL37344 is beneficial for reducing liver fat accumulation and for ameliorating liver steatosis and inflammation in NAFLD. These effects may be associated with PPARs/mCPT-1 and FAT/CD36.
Collapse
Affiliation(s)
- Ziwen Wang
- Gastroenterology Department, the First Affiliated Hospital of Harbin Medical University, #23 Postal Street, Harbin, 150001 Heilongjiang China
| | - Shanshan Li
- Gastroenterology Department, the First Affiliated Hospital of Harbin Medical University, #23 Postal Street, Harbin, 150001 Heilongjiang China
| | - Ruifeng Wang
- Gastroenterology Department, the Fourth Affiliated Hospital of Harbin Medical University, #37 Yiyuan Street, Harbin, 150001 Heilongjiang China
| | - Liansheng Guo
- Gastroenterology Department, the First Affiliated Hospital of Harbin Medical University, #23 Postal Street, Harbin, 150001 Heilongjiang China
| | - Dan Xu
- Gastroenterology Department, the First Affiliated Hospital of Harbin Medical University, #23 Postal Street, Harbin, 150001 Heilongjiang China
| | - Tieyuan Zhang
- Harbin Medical University, #157 Baojian Street, Harbin, 150081 Heilongjiang China
| | - Yifan Xu
- Harbin Medical University, #157 Baojian Street, Harbin, 150081 Heilongjiang China
| | - Wenlong Wang
- Harbin Medical University, #157 Baojian Street, Harbin, 150081 Heilongjiang China
| | - Min Wang
- Harbin Medical University, #157 Baojian Street, Harbin, 150081 Heilongjiang China
| | - Zhongwei Gan
- Harbin Medical University, #157 Baojian Street, Harbin, 150081 Heilongjiang China
| | - Xiaobing Wang
- Gastroenterology Department, the First Affiliated Hospital of Harbin Medical University, #23 Postal Street, Harbin, 150001 Heilongjiang China
| |
Collapse
|
12
|
Fujii H, Kawada N. The Role of Insulin Resistance and Diabetes in Nonalcoholic Fatty Liver Disease. Int J Mol Sci 2020; 21:ijms21113863. [PMID: 32485838 PMCID: PMC7312931 DOI: 10.3390/ijms21113863] [Citation(s) in RCA: 137] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 05/22/2020] [Accepted: 05/24/2020] [Indexed: 12/12/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) consists of the entire spectrum of fatty liver disease in patients without significant alcohol consumption, ranging from nonalcoholic fatty liver (NAFL) to nonalcoholic steatohepatitis (NASH) to cirrhosis, with NASH recently shown as an important cause of hepatocellular carcinoma (HCC). There is a close relationship between insulin resistance (IR) and NAFLD, with a five-fold higher prevalence of NAFLD in patients with type 2 diabetes (T2DM) compared to that in patients without T2DM. IR is involved in the progression of disease conditions such as steatosis and NASH, as well as hepatic fibrosis progression. The mechanisms underlying these processes involve genetic factors, hepatic fat accumulation, alterations in energy metabolism, and inflammatory signals derived from various cell types including immune cells. In NASH-associated fibrosis, the principal cell type responsible for extracellular matrix production is the hepatic stellate cell (HSC). HSC activation by IR involves “direct” and “indirect” pathways. This review will describe the molecular mechanisms of inflammation and hepatic fibrosis in IR, the relationship between T2DM and hepatic fibrosis, and the relationship between T2DM and HCC in patients with NAFLD.
Collapse
Affiliation(s)
- Hideki Fujii
- Department of Premier Preventive Medicine, Graduate School of Medicine, Osaka City University, Osaka 545-8585, Japan;
| | - Norifumi Kawada
- Department of Hepatology, Graduate School of Medicine, Osaka City University, Osaka 545-8585, Japan
- Correspondence: ; Tel.: +81-6-6645-3897
| | | |
Collapse
|
13
|
Lequoy M, Gigante E, Couty JP, Desbois-Mouthon C. Hepatocellular carcinoma in the context of non-alcoholic steatohepatitis (NASH): recent advances in the pathogenic mechanisms. Horm Mol Biol Clin Investig 2020; 41:/j/hmbci.ahead-of-print/hmbci-2019-0044/hmbci-2019-0044.xml. [PMID: 32112699 DOI: 10.1515/hmbci-2019-0044] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 01/16/2020] [Indexed: 12/15/2022]
Abstract
Hepatocellular carcinoma (HCC) is the most common type of liver cancer. HCC is particularly aggressive and is one of the leading causes of cancer mortality. In recent decades, the epidemiological landscape of HCC has undergone significant changes. While chronic viral hepatitis and excessive alcohol consumption have long been identified as the main risk factors for HCC, non-alcoholic steatohepatitis (NASH), paralleling the worldwide epidemic of obesity and type 2 diabetes, has become a growing cause of HCC in the US and Europe. Here, we review the recent advances in epidemiological, genetic, epigenetic and pathogenic mechanisms as well as experimental mouse models that have improved the understanding of NASH progression toward HCC. We also discuss the clinical management of patients with NASH-related HCC and possible therapeutic approaches.
Collapse
Affiliation(s)
- Marie Lequoy
- Service d'Hépato-Gastro-Entérologie, AP-HP, F-75012 Paris, France
- Centre de Recherche Saint-Antoine, INSERM, Sorbonne Université, F-75012 Paris, France
| | - Elia Gigante
- Service d'Hépato-Gastro-Entérologie, AP-HP, F-75012 Paris, France
| | - Jean-Pierre Couty
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, USPC, Université Paris Descartes, Université Paris Diderot, F-75006 Paris, France
| | - Christèle Desbois-Mouthon
- Centre de Recherche des Cordeliers, INSERM UMR_S1138, 15 rue de l'école de médecine, Sorbonne Université, USPC, Université Paris Descartes, Université Paris Diderot, F-75006 Paris, France
| |
Collapse
|
14
|
Simvastatin Reduces Hepatic Oxidative Stress and Endoplasmic Reticulum Stress in Nonalcoholic Steatohepatitis Experimental Model. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:3201873. [PMID: 31316716 PMCID: PMC6604429 DOI: 10.1155/2019/3201873] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 03/12/2019] [Indexed: 12/14/2022]
Abstract
Objective In this study, we evaluated the efficacy of simvastatin in the treatment of nonalcoholic steatohepatitis induced by methionine and choline-deficient diet in mice and its possible effect on factors involved in the pathogenesis of the disease including oxidative stress and endoplasmic reticulum stress. Method Male C57BL6 mice were fed either a normal diet (control) or a methionine and choline-deficient diet for four weeks and then treated orally with simvastatin (4 mg/kg once a day) for two final weeks. At the end of the experimental period, liver integrity, biochemical analysis, hepatic lipids, histology, DNA damage, biomarkers of oxidative stress, and endoplasmic reticulum stress were assessed. Results Simvastatin treatment was able to significantly reduce hepatic damage enzymes and hepatic lipids and lower the degree of hepatocellular ballooning, without showing genotoxic effects. Simvastatin caused significant decreases in lipid peroxidation, with some changes in antioxidant enzymes superoxide dismutase and glutathione peroxidase. Simvastatin activates antioxidant enzymes via Nrf2 and inhibits endoplasmic reticulum stress in the liver. Conclusions In summary, the results provide evidence that in mice with experimental nonalcoholic steatohepatitis induced by a methionine and choline-deficient diet, the reduction of liver damage by simvastatin is associated with attenuated oxidative and endoplasmic reticulum stress.
Collapse
|
15
|
Park JG, Mok JS, Han YI, Park TS, Kang KW, Choi CS, Park HD, Park J. Connectivity mapping of angiotensin-PPAR interactions involved in the amelioration of non-alcoholic steatohepatitis by Telmisartan. Sci Rep 2019; 9:4003. [PMID: 30850637 PMCID: PMC6408578 DOI: 10.1038/s41598-019-40322-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 01/30/2019] [Indexed: 12/18/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a global health problem that is associated with various metabolic disorders. Telmisartan is a potential treatment for NAFLD due to its ability to improve insulin sensitivity and decrease hepatic fat accumulation via modulation of PPARγ, and to suppress hepatic fibrosis by blocking angiotensin II receptors. However, the underlying mechanisms of action of telmisartan have yet to be fully elucidated. In the present study, diabetic nonalcoholic steatohepatitis (NASH) mice (STAM mice) received daily administrations of telmisartan for 6 weeks to assess the improvements in NASH. Hepatic transcriptome analyses revealed that the amelioration of NASH likely occurred through the regulation of inflammatory- and fibrosis-related gene responses. An integrated network analysis including transcriptional and non-transcriptional genes regulated by telmisartan showed that the NAFLD pathway is interconnected with the dysregulated RAS-PPAR-NFκB pathways. The downstream targets of PPARα, PPARδ, and RELA in this network significantly overlapped with telmisartan-induced differentially expressed genes (DEGs), which were verified in palmitate-treated Hepa1c1c7 cell line. This transcriptome approach accompanied with cell-based molecular analyses provided the opportunity to understand the fundamental molecular mechanisms underpinning the therapeutic effects of telmisartan, and will contribute to the establishment of a novel pharmacological treatment for NASH patients.
Collapse
Affiliation(s)
| | - Jong Soo Mok
- Graduate School of International Agricultural Technology, Seoul National University, Seoul, Korea
| | - Young In Han
- Institute of Green Bio Science and Technology, Seoul National University, Seoul, Korea
| | - Tae Sub Park
- Graduate School of International Agricultural Technology, Seoul National University, Seoul, Korea.,Institute of Green Bio Science and Technology, Seoul National University, Seoul, Korea
| | - Keon Wook Kang
- College of pharmacy, Seoul National University, Seoul, Korea
| | - Cheol Soo Choi
- Korea mouse metabolic phenotyping center, Lee Gil Ya cancer and diabetes institute, Gachon University School of Medicine, Seongnam-si, Republic of Korea.,Endocrinology, Internal Medicine, Gachon University Gil Medical Center, Seongnam-si, Republic of Korea
| | | | - Joonghoon Park
- Graduate School of International Agricultural Technology, Seoul National University, Seoul, Korea. .,Institute of Green Bio Science and Technology, Seoul National University, Seoul, Korea.
| |
Collapse
|
16
|
Manzano-Núñez F, Arámbul-Anthony MJ, Galán Albiñana A, Leal Tassias A, Acosta Umanzor C, Borreda Gascó I, Herrera A, Forteza Vila J, Burks DJ, Noon LA. Insulin resistance disrupts epithelial repair and niche-progenitor Fgf signaling during chronic liver injury. PLoS Biol 2019; 17:e2006972. [PMID: 30695023 PMCID: PMC6368328 DOI: 10.1371/journal.pbio.2006972] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 02/08/2019] [Accepted: 01/08/2019] [Indexed: 02/07/2023] Open
Abstract
Insulin provides important information to tissues about feeding behavior and energy status. Defective insulin signaling is associated with ageing, tissue dysfunction, and impaired wound healing. In the liver, insulin resistance leads to chronic damage and fibrosis, but it is unclear how tissue-repair mechanisms integrate insulin signals to coordinate an appropriate injury response or how they are affected by insulin resistance. In this study, we demonstrate that insulin resistance impairs local cellular crosstalk between the fibrotic stroma and bipotent adult liver progenitor cells (LPCs), whose paracrine interactions promote epithelial repair and tissue remodeling. Using insulin-resistant mice deficient for insulin receptor substrate 2 (Irs2), we highlight dramatic impairment of proregenerative fibroblast growth factor 7 (Fgf7) signaling between stromal niche cells and LPCs during chronic injury. We provide a detailed account of the role played by IRS2 in promoting Fgf7 ligand and receptor (Fgfr2-IIIb) expression by the two cell compartments, and we describe an insulin/IRS2-dependent feed-forward loop capable of sustaining hepatic re-epithelialization by driving FGFR2-IIIb expression. Finally, we shed light on the regulation of IRS2 and FGF7 within the fibrotic stroma and show—using a human coculture system—that IRS2 silencing shifts the equilibrium away from paracrine epithelial repair in favor of fibrogenesis. Hence, we offer a compelling insight into the contribution of insulin resistance to the pathogenesis of chronic liver disease and propose IRS2 as a positive regulator of communication between cell types and the transition between phases of stromal to epithelial repair. “Insulin resistance” is a chronic state of reduced sensitivity to the effects of circulating insulin. It is one of the hallmarks of metabolic disease and a consequence of ageing, but insulin resistance is also observed in otherwise healthy individuals after severe trauma/hemorrhage/sepsis, suggesting that it plays a physiological role in modulating the response to injury. Defective insulin signals are linked to impaired wound healing, yet it remains unclear how systemic changes affect locally the cells that coordinate tissue repair. In this study, we used the liver to assess how insulin resistance impacts the injury response in mice. We provide proof of concept that insulin signals are locally integrated by the fibrotic microenvironment surrounding the adult liver stem cells during chronic injury, resulting in the increased expression of epithelial repair signals. Insulin also simultaneously primes stem cells to respond to these stromal growth factors, leading to an increased participation in epithelial repair. Insulin resistance disrupts this local paracrine circuit, resulting in a blunted epithelial response to chronic injury that exacerbates tissue damage. Our model highlights a potential role for insulin in switching the hepatic injury response from a stromal repair process to an epithelial repair process. To our knowledge, our data provide a new perspective from which to reassess how insulin resistance influences fibrosis, wound healing, and tissue remodeling during injury.
Collapse
Affiliation(s)
- Fátima Manzano-Núñez
- CIBERDEM (Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas), Madrid, Spain
- Centro de Investigación Príncipe Felipe, Valencia, Spain
| | - María José Arámbul-Anthony
- CIBERDEM (Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas), Madrid, Spain
- Centro de Investigación Príncipe Felipe, Valencia, Spain
| | | | | | - Carlos Acosta Umanzor
- CIBERDEM (Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas), Madrid, Spain
- Centro de Investigación Príncipe Felipe, Valencia, Spain
| | - Irene Borreda Gascó
- Instituto Valenciano de Patología, Universidad Católica de Valencia San Vicente Màrtir, Valencia, Spain
| | | | - Jerónimo Forteza Vila
- Centro de Investigación Príncipe Felipe, Valencia, Spain
- Instituto Valenciano de Patología, Universidad Católica de Valencia San Vicente Màrtir, Valencia, Spain
| | - Deborah J. Burks
- CIBERDEM (Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas), Madrid, Spain
- Centro de Investigación Príncipe Felipe, Valencia, Spain
| | - Luke A. Noon
- CIBERDEM (Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas), Madrid, Spain
- Centro de Investigación Príncipe Felipe, Valencia, Spain
- * E-mail:
| |
Collapse
|
17
|
Kling DN, DeBose-Scarlett EM, Teixeira LD, Gezan SA, Lorca GL, Gonzalez CF. Sex Modulates Lactobacillus johnsonii N6.2 and Phytophenol Effectiveness in Reducing High Fat Diet Induced mTOR Activation in Sprague-Dawley Rats. Front Microbiol 2018; 9:2649. [PMID: 30459740 PMCID: PMC6232610 DOI: 10.3389/fmicb.2018.02649] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 10/17/2018] [Indexed: 12/12/2022] Open
Abstract
Metabolic syndrome (MetS) is the underlying cause of some devastating diseases, including type 2 diabetes and cardiovascular disease. These diseases have been associated with over-activation of the mechanistic Target of Rapamycin (mTOR) pathway. This study utilizes a high fat diet (HFD) to induce MetS and to dissect the effects of a beneficial bacterium, L. johnsonii N6.2, and natural phenolics on mTOR complex 1 (mTORC1) expression compared to a reduced energy density diet (REDD). HFD significantly elevated MetS markers in males, as noted through an increase in weight, glucose levels, and triglyceride levels. Treatments were effective in reducing mTORC1-activating phosphorylation of pAKT-T308 and pAKT-S473 (p = 0.0012 and 0.0049, respectively) in HFD-fed females, with the combined treatments of L. johnsonii and phytophenols reducing phosphorylation below REDD-fed control levels, and significantly below HFD-fed control levels. Meanwhile, diet was the significant factor influencing male mTORC1-activating phosphorylation (p < 0.0001), as treatments were only effective in reducing phosphorylation in REDD-fed animals. Downstream analysis of mTORC1 activated genes phosphogluconate dehydrogenase (pgd) and phosphofructose kinase (pfk) followed this similar trend, enforcing the significant effect sex has on a treatments’ ability to modulate diet induced abnormalities. Analyzing mTORC1 stimulators such as insulin, inflammatory cytokines, and tryptophan, revealed no significant differences among groups. These results indicate that the effects observed on mTORC1 are a direct consequence of the treatments, and not exerted indirectly via the modulation of stimuli. This study highlights the potential use of commensal microorganisms and natural compounds in reducing the onset of metabolic diseases through mTORC1.
Collapse
Affiliation(s)
- Danielle N Kling
- Department of Microbiology and Cell Science, Genetics Institute, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, Unites States
| | - Evon M DeBose-Scarlett
- Department of Microbiology and Cell Science, Genetics Institute, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, Unites States
| | - Leandro D Teixeira
- Department of Microbiology and Cell Science, Genetics Institute, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, Unites States
| | - Salvador A Gezan
- School of Forest Resources and Conservation, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, Unites States
| | - Graciela L Lorca
- Department of Microbiology and Cell Science, Genetics Institute, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, Unites States
| | - Claudio F Gonzalez
- Department of Microbiology and Cell Science, Genetics Institute, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, Unites States
| |
Collapse
|
18
|
Selective insulin resistance with differential expressions of IRS-1 and IRS-2 in human NAFLD livers. Int J Obes (Lond) 2018; 42:1544-1555. [PMID: 29717275 PMCID: PMC6160396 DOI: 10.1038/s41366-018-0062-9] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 01/31/2018] [Accepted: 02/13/2018] [Indexed: 12/30/2022]
Abstract
Background/objective: Insulin signals, via the regulation of key enzyme expression, both suppress gluconeogenesis and enhance lipid synthesis in the liver. Animal studies have revealed insulin signaling favoring gluconeogenesis suppression to be selectively impaired in steatotic livers. However, whether, and if so how, such selective insulin resistance occurs in human steatotic livers remains unknown. Our aim was to investigate selective insulin resistance in human livers with non-alcoholic fatty liver disease (NAFLD). Subjects/methods: We examined mRNA expressions of key molecules for insulin signaling, gluconeogenesis and lipogenesis in human liver biopsy samples obtained from 51 non-diabetic subjects: 9 healthy controls and 42 NAFLD patients, and analyzed associations of these molecules with each other and with detailed pathological and clinical biochemistry data. Results: In NAFLD patients, insulin receptor substrate (IRS)-2 expression was decreased, while those of key enzymes for gluconeogenesis were increased. These alterations of IRS-2 and gluconeogenesis enzymes were induced both in simple steatosis (SS) and non-alcoholic steatohepatitis (NASH), while these expression levels did not differ between SS and NASH. Furthermore, alterations in the expressions of IRS-2 and gluconeogenesis enzymes showed strong negative correlations and were concurrently induced in the early histological stage of NAFLD. In contrast, fatty acid synthase (FAS) expression was not decreased in NAFLD, despite IRS-2 downregulation, but correlated strongly with IRS-1 expression. Furthermore, no histological scores were associated with these molecules. Thus, IRS-1 signaling, which is not impaired in NAFLD, appears to modulate FAS expression. Conclusion: These analyses revealed that selective insulin resistance is present in human NAFLD livers and occurs in its early phases. The effect of insulin, during the IRS step, on gene expressions for lipogenesis and gluconeogenesis are apparently distinct and preferential downregulation of IRS-2 may contribute to selective resistance to the suppressive effects of insulin on gluconeogenesis.
Collapse
|
19
|
Ma J, Matkar S, He X, Hua X. FOXO family in regulating cancer and metabolism. Semin Cancer Biol 2018; 50:32-41. [PMID: 29410116 DOI: 10.1016/j.semcancer.2018.01.018] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 01/28/2018] [Accepted: 01/31/2018] [Indexed: 01/07/2023]
Abstract
FOXO proteins are a sub-group of a superfamily of forkhead box (FOX)-containing transcription factors (TFs). FOXOs play an important role in regulating a plethora of biological activities ranging from development, cell signaling, and tumorigenesis to cell metabolism. Here we mainly focus on reviewing the role of FOXOs in regulating tumor and metabolism. Moreover, how crosstalk among various pathways influences the function of FOXOs will be reviewed. Further, the paradoxical role for FOXOs in controlling the fate of cancer and especially resistance/sensitivity of cancer to the class of drugs that target PI3K/AKT will also be reviewed. Finally, how FOXOs regulate crosstalk between common cancer pathways and cell metabolism pathways, and how these crosstalks affect the fate of the cancer will be discussed.
Collapse
Affiliation(s)
- Jian Ma
- Department of Cancer Biology, Abramson Family Cancer Research Institute, Abramson Cancer Center, Institute of Diabetes, Obesity, and Metabolism (IDOM), University of Pennsylvania Perelman School of Medicine, 421 Curie Blvd., Philadelphia, PA 19104, USA; State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 678 Haping Road, Harbin, Heilongjiang 150069, China; College of Life Science, Northeast Forestry University, 26 Hexing Road, Harbin, Heilongjiang 150040, China.
| | - Smita Matkar
- Department of Cancer Biology, Abramson Family Cancer Research Institute, Abramson Cancer Center, Institute of Diabetes, Obesity, and Metabolism (IDOM), University of Pennsylvania Perelman School of Medicine, 421 Curie Blvd., Philadelphia, PA 19104, USA.
| | - Xin He
- Department of Cancer Biology, Abramson Family Cancer Research Institute, Abramson Cancer Center, Institute of Diabetes, Obesity, and Metabolism (IDOM), University of Pennsylvania Perelman School of Medicine, 421 Curie Blvd., Philadelphia, PA 19104, USA.
| | - Xianxin Hua
- Department of Cancer Biology, Abramson Family Cancer Research Institute, Abramson Cancer Center, Institute of Diabetes, Obesity, and Metabolism (IDOM), University of Pennsylvania Perelman School of Medicine, 421 Curie Blvd., Philadelphia, PA 19104, USA.
| |
Collapse
|
20
|
Zhang SB, Yang S, Zhang Z, Zhang A, Zhang M, Yin L, Casey-Sawicki K, Swarts S, Vidyasagar S, Zhang L, Okunieff P. Thoracic gamma irradiation-induced obesity in C57BL/6 female mice. Int J Radiat Biol 2017; 93:1334-1342. [DOI: 10.1080/09553002.2017.1385871] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Steven B. Zhang
- Department of Radiation Oncology, University of Florida Health Cancer Center, Gainesville, FL, USA
| | - Shanmin Yang
- Department of Radiation Oncology, University of Florida Health Cancer Center, Gainesville, FL, USA
| | - Zhenhuan Zhang
- Department of Radiation Oncology, University of Florida Health Cancer Center, Gainesville, FL, USA
| | - Amy Zhang
- Department of Radiation Oncology, University of Florida Health Cancer Center, Gainesville, FL, USA
| | - Mei Zhang
- Department of Radiation Oncology, University of Florida Health Cancer Center, Gainesville, FL, USA
| | - Liangjie Yin
- Department of Radiation Oncology, University of Florida Health Cancer Center, Gainesville, FL, USA
| | - Katherine Casey-Sawicki
- Department of Radiation Oncology, University of Florida Health Cancer Center, Gainesville, FL, USA
| | - Steven Swarts
- Department of Radiation Oncology, University of Florida Health Cancer Center, Gainesville, FL, USA
| | - Sadasivan Vidyasagar
- Department of Radiation Oncology, University of Florida Health Cancer Center, Gainesville, FL, USA
| | - Lurong Zhang
- Department of Radiation Oncology, University of Florida Health Cancer Center, Gainesville, FL, USA
| | - Paul Okunieff
- Department of Radiation Oncology, University of Florida Health Cancer Center, Gainesville, FL, USA
| |
Collapse
|
21
|
Insulin resistance promotes Lysyl Oxidase Like 2 induction and fibrosis accumulation in non-alcoholic fatty liver disease. Clin Sci (Lond) 2017; 131:1301-1315. [PMID: 28468951 DOI: 10.1042/cs20170175] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Revised: 04/10/2017] [Accepted: 05/02/2017] [Indexed: 12/12/2022]
Abstract
In patients with non-alcoholic fatty liver disease (NAFLD), insulin resistance (IR) associates with fibrosis progression independently of the hepatic inflammation, but the mechanisms are still unclear. We modeled the independent contribution of inflammation (non-alcoholic steatohepatitis: NASH) by exploiting the methionine-choline deficient (MCD) diet, and that of IR by insulin receptor (InsR) haploinsufficiency (InsR+/-) in the pathogenesis of liver fibrosis in C57BL/6 mice. We confirmed the study findings in 96 patients with NAFLD. InsR+/- enhanced hepatic fat content and impaired hepatic insulin signaling leading to Forkhead box protein O1 (FoxO1) accumulation in MCD-fed mice. Remarkably, despite reduced inflammation and hampered transdifferentiation of hepatic stellate cells (HSCs), InsR+/- promoted hepatic fibrosis accumulation, which correlated with the induction of the Lysyl Oxidase Like 2 (Loxl2), involved in matrix stabilization. Loxl2 up-regulation was not a cell autonomous property of insulin resistant HSCs, but was dependent on microparticles (MPs) released specifically by insulin resistant hepatocytes (HEPs) exposed to fatty acids. The mechanism entailed FoxO1 up-regulation, as FoxO1 silencing normalized Loxl2 expression reversing fibrosis in InsR+/- MCD-fed mice. Loxl2 up-regulation was similarly detected during IR induced by obesity, but not by lipogenic stimuli (fructose feeding). Most importantly, LOXL2 up-regulation was observed in NAFLD patients with type 2 diabetes (T2D) and LOXL2 hepatic and circulating levels correlated with histological fibrosis progression. IR favors fibrosis deposition independently of the classic 'inflammation - HSC transdifferentiation' pathway. The mechanism entails a cross-talk between enhanced lipotoxicity in insulin resistant HEPs and Loxl2 production by HSCs, which was confirmed in patients with diabetes, thereby facilitating extracellular matrix (ECM) stabilization.
Collapse
|
22
|
Engin A. Human Protein Kinases and Obesity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 960:111-134. [DOI: 10.1007/978-3-319-48382-5_5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
23
|
Dongiovanni P, Rametta R, Meroni M, Valenti L. The role of insulin resistance in nonalcoholic steatohepatitis and liver disease development--a potential therapeutic target? Expert Rev Gastroenterol Hepatol 2016; 10:229-42. [PMID: 26641143 DOI: 10.1586/17474124.2016.1110018] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Insulin resistance (IR) is defined by the inability of insulin to exert its metabolic actions, due to impaired activation of intracellular insulin signaling. This condition is caused by genetic defects or by environmental conditions, among which the most common is obesity. Systemic IR determines the development of hepatic fat accumulation, which can progress to nonalcoholic steatohepatitis, cirrhosis and hepatocellular carcinoma, and is a major determinant of liver disease independently of coexisting factors. Therefore, insulin-sensitizing drugs are currently under evaluation to improve steatohepatitis. Indeed, manipulation of nuclear hormone receptors is already under scrutiny for liver disease prevention by amelioration of IR, whereas NOTCH signaling inhibition represents a novel approach. Nevertheless, further research is warranted to better understand the mechanism linking IR to progressive fibrogenesis in the absence of inflammation and to identify novel drug targets.
Collapse
Affiliation(s)
- Paola Dongiovanni
- a Department of Pathophysiology and Transplantation , Università degli Studi di Milano, and Internal Medicine and Metabolic Diseases, Fondazione IRCCS Ca' Granda Ospedale Policlinico Milano , Milano , Italy
| | - Raffaela Rametta
- a Department of Pathophysiology and Transplantation , Università degli Studi di Milano, and Internal Medicine and Metabolic Diseases, Fondazione IRCCS Ca' Granda Ospedale Policlinico Milano , Milano , Italy
| | - Marica Meroni
- a Department of Pathophysiology and Transplantation , Università degli Studi di Milano, and Internal Medicine and Metabolic Diseases, Fondazione IRCCS Ca' Granda Ospedale Policlinico Milano , Milano , Italy
| | - Luca Valenti
- a Department of Pathophysiology and Transplantation , Università degli Studi di Milano, and Internal Medicine and Metabolic Diseases, Fondazione IRCCS Ca' Granda Ospedale Policlinico Milano , Milano , Italy
| |
Collapse
|
24
|
Valenti L, Bugianesi E, Pajvani U, Targher G. Nonalcoholic fatty liver disease: cause or consequence of type 2 diabetes? Liver Int 2016; 36:1563-1579. [PMID: 27276701 DOI: 10.1111/liv.13185] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Accepted: 06/06/2016] [Indexed: 02/13/2023]
Abstract
Growing epidemiological evidence suggests that nonalcoholic fatty liver disease (NAFLD) is an early predictor of and determinant for the development of type 2 diabetes and other features of the metabolic syndrome. This finding may have important clinical implications for the diagnosis, prevention and treatment of type 2 diabetes and its chronic complications. However, given the complex and bi-directional relationships between NAFLD, insulin resistance and chronic hyperglycaemia, it is extremely difficult to distinguish whether NAFLD is a cause or a consequence of insulin resistance and type 2 diabetes. Indeed, at the molecular level, hepatic lipogenesis and hepatic glucose production depend on differentially regulated branches of the insulin signalling pathway. Furthermore, genetic studies suggest that excess hepatic fat is associated with progressive liver disease, but does not always increase the risk of incident type 2 diabetes. Here, we will briefly review the epidemiological, pathophysiological and molecular evidence linking NAFLD to the development of type 2 diabetes. We will also discuss some recent genetic and therapeutic advances that seem to challenge a causal role of NAFLD in the pathogenesis type 2 diabetes, and propose a working hypothesis to explain this apparent conundrum. In conclusion, progressive liver disease and type 2 diabetes are divergent though inter-related consequences of insulin resistance and the metabolic syndrome.
Collapse
Affiliation(s)
- Luca Valenti
- Internal Medicine and Metabolic Diseases, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy. .,Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milano, Italy.
| | - Elisabetta Bugianesi
- Division of Gastroenterology, Department of Medical Sciences, A.O.U. Città della Salute e della Scienza, Università di Torino, Torino, Italy
| | - Utpal Pajvani
- Division of Endocrinology, Columbia University, New York, NY, USA
| | - Giovanni Targher
- Section of Endocrinology, Diabetes and Metabolism, Department of Medicine, University and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
| |
Collapse
|
25
|
Jiang S, He H, Tan L, Wang L, Su Z, Liu Y, Zhu H, Zhang M, Hou FF, Li A. Proteomic and phosphoproteomic analysis of renal cortex in a salt-load rat model of advanced kidney damage. Sci Rep 2016; 6:35906. [PMID: 27775022 PMCID: PMC5075906 DOI: 10.1038/srep35906] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 10/07/2016] [Indexed: 12/21/2022] Open
Abstract
Salt plays an essential role in the progression of chronic kidney disease and hypertension. However, the mechanisms underlying pathogenesis of salt-induced kidney damage remain largely unknown. Here, Sprague-Dawley rats, that underwent 5/6 nephrectomy (5/6Nx, a model of advanced kidney damage) or sham operation, were treated for 2 weeks with a normal or high-salt diet. We employed aTiO2 enrichment, iTRAQ labeling and liquid-chromatography tandem mass spectrometry strategy for proteomic and phosphoproteomic profiling of the renal cortex. We found 318 proteins differentially expressed in 5/6Nx group relative to sham group, and 310 proteins significantly changed in response to salt load in 5/6Nx animals. Totally, 1810 unique phosphopeptides corresponding to 550 phosphoproteins were identified. We identified 113 upregulated and 84 downregulated phosphopeptides in 5/6Nx animals relative to sham animals. Salt load induced 78 upregulated and 91 downregulated phosphopeptides in 5/6Nx rats. The differentially expressed phospholproteins are important transporters, structural molecules, and receptors. Protein-protein interaction analysis revealed that the differentially phosphorylated proteins in 5/6Nx group, Polr2a, Srrm1, Gsta2 and Pxn were the most linked. Salt-induced differential phosphoproteins, Myh6, Lmna and Des were the most linked. Altered phosphorylation levels of lamin A and phospholamban were validated. This study will provide new insight into pathogenetic mechanisms of chronic kidney disease and salt sensitivity.
Collapse
Affiliation(s)
- Shaoling Jiang
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hanchang He
- The First People's Foshan Hospital, Foshan, China
| | - Lishan Tan
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Liangliang Wang
- Division of Nephrology, First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou 510405, P.R. China
| | - Zhengxiu Su
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yufeng Liu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hongguo Zhu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Menghuan Zhang
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fan Fan Hou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Aiqing Li
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
26
|
Dongiovanni P, Valenti L. Genetics of nonalcoholic fatty liver disease. Metabolism 2016; 65:1026-37. [PMID: 26409295 DOI: 10.1016/j.metabol.2015.08.018] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 08/23/2015] [Accepted: 08/26/2015] [Indexed: 02/07/2023]
Abstract
UNLABELLED Epidemiological, familial, and twin studies indicate that non-alcoholic fatty liver disease, now the leading cause of liver damage in developed countries, has a strong heritability. The common I148M variant of PNPLA3 impairing hepatocellular lipid droplets remodeling is the major genetic determinant of hepatic fat content. The I148M variant has a strong impact on the full spectrum of liver damage related to fatty liver, encompassing non-alcoholic steatohepatitis, advanced fibrosis, and hepatocellular carcinoma, and influences the response to therapeutic approaches. Common variants in GCKR enhance de novo hepatic lipogenesis in response to glucose and liver inflammation. Furthermore, the low-frequency E167K variant of TM6SF2 and rare mutations in APOB, which impair very low-density lipoproteins secretion, predispose to progressive fatty liver. CONCLUSIONS These and other recent findings reviewed here indicate that impaired lipid handling by hepatocytes has a major role in the pathogenesis of non-alcoholic fatty liver disease by triggering inflammation, fibrogenesis, and carcinogenesis. These discoveries have provided potential novel biomarkers for clinical use and have revealed intriguing therapeutic targets.
Collapse
Affiliation(s)
- Paola Dongiovanni
- Internal Medicine and Metabolic Diseases, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico
| | - Luca Valenti
- Internal Medicine and Metabolic Diseases, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico; Department of Pathophysiology and Transplantation, Università degli Studi Milano, Milan, Italy.
| |
Collapse
|
27
|
Ruscica M, Ferri N, Macchi C, Meroni M, Lanti C, Ricci C, Maggioni M, Fracanzani AL, Badiali S, Fargion S, Magni P, Valenti L, Dongiovanni P. Liver fat accumulation is associated with circulating PCSK9. Ann Med 2016; 48:384-91. [PMID: 27222915 DOI: 10.1080/07853890.2016.1188328] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) associates with cardiovascular disease independently of classic risk factors. Proprotein Convertase Subtilisin/Kexin type 9 (PCSK9) is secreted by hepatocytes and inhibits the uptake of low-density lipoproteins by targeting the receptor for degradation, and possibly lipogenesis. PCSK9 loss-of-function mutations and anti-PCKS9 drugs reduce LDL-cholesterol. AIM To evaluate whether hepatic fat content is associated with circulating PCSK9. MATERIALS AND METHODS In 201 consecutive patients biopsied for suspected nonalcoholic steatohepatitis, liver damage was quantified by NAFLD activity score, circulating PCSK9 by ELISA, and hepatic mRNA by qRT-PCR in a subset (n = 76). RESULTS Circulating PCSK9 was associated with steatosis grade (p = 0.0011), necroinflammation (p < 0.001), ballooning (p = 0.005), and fibrosis stage (p = 0.001). At multivariate analysis, PCSK9 was associated with steatosis grade (p = 0.012), older age and lower BMI, independently of sex, hyperglycemia, and fibrosis/inflammation. Circulating PCSK9 was associated with hepatic expression of SREBP-1c (p = 0.0002) and FAS (p = 0.03). PCSK9 mRNA levels were also correlated with steatosis severity (p = 0.04) and hepatic APOB (p < 0.001), SREBP-1c (p = 0.047) and FAS expression (p = 0.001). CONCLUSIONS Circulating PCSK9 increases with hepatic fat accumulation and correlates with the severity of steatosis, independently of metabolic confounders and liver damage. Modulation of PCSK9 synthesis and release might be involved in NAFLD pathogenesis. Key Messages Circulating PCSK9 levels increase with hepatic fat accumulation. Circulating PCSK9 levels are associated with increased de novo lipogenesis. Hepatic PCSK9 expression is associated with steatosis severity and activation of lipogenesis.
Collapse
Affiliation(s)
- Massimiliano Ruscica
- a Department of Pharmacological and Biomolecular Sciences , Università Degli Studi Di Milano , Milano , Italy
| | - Nicola Ferri
- b Department of Pharmaceutical and Pharmacological Sciences , Università di Padova , Padova , Italy
| | - Chiara Macchi
- a Department of Pharmacological and Biomolecular Sciences , Università Degli Studi Di Milano , Milano , Italy
| | - Marica Meroni
- c Department of Pathophysiology and Transplantation , Università Degli Studi Di Milano , Milano , Italy
| | - Claudia Lanti
- d Internal Medicine and Metabolic Diseases , Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico , Milano , Italy
| | - Chiara Ricci
- a Department of Pharmacological and Biomolecular Sciences , Università Degli Studi Di Milano , Milano , Italy
| | - Marco Maggioni
- e Department of Pathology , Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico , Milano , Italy
| | - Anna Ludovica Fracanzani
- c Department of Pathophysiology and Transplantation , Università Degli Studi Di Milano , Milano , Italy ;,d Internal Medicine and Metabolic Diseases , Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico , Milano , Italy
| | - Sara Badiali
- f Department of Surgery , Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico , Milano , Italy
| | - Silvia Fargion
- c Department of Pathophysiology and Transplantation , Università Degli Studi Di Milano , Milano , Italy ;,d Internal Medicine and Metabolic Diseases , Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico , Milano , Italy
| | - Paolo Magni
- a Department of Pharmacological and Biomolecular Sciences , Università Degli Studi Di Milano , Milano , Italy
| | - Luca Valenti
- c Department of Pathophysiology and Transplantation , Università Degli Studi Di Milano , Milano , Italy ;,d Internal Medicine and Metabolic Diseases , Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico , Milano , Italy
| | - Paola Dongiovanni
- d Internal Medicine and Metabolic Diseases , Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico , Milano , Italy
| |
Collapse
|
28
|
Petta S, Valenti L, Marra F, Grimaudo S, Tripodo C, Bugianesi E, Cammà C, Cappon A, Di Marco V, Di Maira G, Dongiovanni P, Rametta R, Gulino A, Mozzi E, Orlando E, Maggioni M, Pipitone RM, Fargion S, Craxì A. MERTK rs4374383 polymorphism affects the severity of fibrosis in non-alcoholic fatty liver disease. J Hepatol 2016; 64:682-90. [PMID: 26596542 DOI: 10.1016/j.jhep.2015.10.016] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 09/16/2015] [Accepted: 10/12/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIM Homozygosity for a common non-coding rs4374383 G>A polymorphism in MERTK (myeloid-epithelial-reproductive tyrosine kinase) has been associated with the protection against fibrosis progression in chronic hepatitis C. The main study objective was to assess whether MERTK AA genotype influences liver fibrosis, and secondarily MERTK expression in patients with non-alcoholic fatty liver disease (NAFLD). We also investigated whether MERTK is expressed in human hepatic stellate cells (HSC) and in murine models of fibrogenesis. METHODS We considered 533 consecutive patients who underwent liver biopsy for suspected non-alcoholic steatohepatitis (NASH) without severe obesity from two Italian cohorts. As controls, we evaluated 158 patients with normal liver enzymes and without metabolic disturbances. MERTK rs4374383 genotype was assessed by 5'-nuclease assays. MERTK expression was analysed in mouse models of fibrosis, and the effect of the MERTK ligand GAS6 were investigated in human HSC. RESULTS Clinically significant fibrosis (stage F2-F4) was observed in 19% of patients with MERTK AA compared to 30% in those with MERTK GG/GA (OR 0.43, CI 0.21-0.88, p=0.02; adjusted for centre, and genetic, clinical-metabolic and histological variables). The protective rs4374383 AA genotype was associated with lower MERTK hepatic expression. MERTK was overexpressed in the liver of NAFLD patients with F2-F4 fibrosis and in in vivo models of fibrogenesis. Furthermore, exposure of cultured human HSC to the MERTK ligand GAS6, increased cell migration and induced procollagen expression. These effects were counteracted by inhibition of MERTK activity, which also resulted in apoptotic death of HSC. CONCLUSIONS The rs4374383 AA genotype, associated with lower intrahepatic expression of MERTK, is protective against F2-F4 fibrosis in patients with NAFLD. The mechanism may involve modulation of HSC activation.
Collapse
Affiliation(s)
- Salvatore Petta
- Sezione di Gastroenterologia, DiBiMIS, University of Palermo, Italy.
| | - Luca Valenti
- Department of Pathophysiology and Transplantation, Università degli Studi, Internal Medicine, Fondazione Ca' Granda IRCCS Ospedale Maggiore Policlinico, Milano, Italy
| | - Fabio Marra
- Dipartimento di Medicina Sperimentale e Clinica, Università degli Studi di Firenze, Italy
| | | | - Claudio Tripodo
- Cattedra di Anatomia Patologica, University of Palermo, Italy
| | - Elisabetta Bugianesi
- Division of Gastro-Hepatology, Department of Medical Sciences, San Giovanni Battista Hospital, University of Torino, Torino, Italy
| | - Calogero Cammà
- Sezione di Gastroenterologia, DiBiMIS, University of Palermo, Italy
| | - Andrea Cappon
- Dipartimento di Medicina Sperimentale e Clinica, Università degli Studi di Firenze, Italy
| | - Vito Di Marco
- Sezione di Gastroenterologia, DiBiMIS, University of Palermo, Italy
| | - Giovanni Di Maira
- Dipartimento di Medicina Sperimentale e Clinica, Università degli Studi di Firenze, Italy
| | - Paola Dongiovanni
- Department of Pathophysiology and Transplantation, Università degli Studi, Internal Medicine, Fondazione Ca' Granda IRCCS Ospedale Maggiore Policlinico, Milano, Italy
| | - Raffaela Rametta
- Department of Pathophysiology and Transplantation, Università degli Studi, Internal Medicine, Fondazione Ca' Granda IRCCS Ospedale Maggiore Policlinico, Milano, Italy
| | | | - Enrico Mozzi
- Department of Pathophysiology and Transplantation, Università degli Studi, Internal Medicine, Fondazione Ca' Granda IRCCS Ospedale Maggiore Policlinico, Milano, Italy
| | - Emanuele Orlando
- Sezione di Gastroenterologia, DiBiMIS, University of Palermo, Italy
| | - Marco Maggioni
- Pathology, Fondazione IRCCS Ca' Granda Policlinico, Milano, Italy
| | | | - Silvia Fargion
- Department of Pathophysiology and Transplantation, Università degli Studi, Internal Medicine, Fondazione Ca' Granda IRCCS Ospedale Maggiore Policlinico, Milano, Italy
| | - Antonio Craxì
- Sezione di Gastroenterologia, DiBiMIS, University of Palermo, Italy
| |
Collapse
|
29
|
Donati B, Motta BM, Pingitore P, Meroni M, Pietrelli A, Alisi A, Petta S, Xing C, Dongiovanni P, del Menico B, Rametta R, Mancina RM, Badiali S, Fracanzani AL, Craxì A, Fargion S, Nobili V, Romeo S, Valenti L. The rs2294918 E434K variant modulates patatin-like phospholipase domain-containing 3 expression and liver damage. Hepatology 2016; 63:787-98. [PMID: 26605757 DOI: 10.1002/hep.28370] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 11/23/2015] [Indexed: 12/23/2022]
Abstract
UNLABELLED The patatin-like phosholipase domain-containing 3 (PNPLA3) rs738409 polymorphism (I148M) is a major determinant of hepatic fat and predisposes to the full spectrum of liver damage in nonalcoholic fatty liver disease (NAFLD). The aim of this study was to evaluate whether additional PNPLA3 coding variants contribute to NAFLD susceptibility, first in individuals with contrasting phenotypes (with early-onset NAFLD vs. very low aminotransferases) and then in a large validation cohort. Rare PNPLA3 variants were not detected by sequencing coding regions and intron-exon boundaries either in 142 patients with early-onset NAFLD nor in 100 healthy individuals with alanine aminotransferase <22/20 IU/mL. Besides rs738409 I148M, the rs2294918 G>A polymorphism (E434K sequence variant) was over-represented in NAFLD (adjusted P = 0.01). In 1,447 subjects with and without NAFLD, the 148M-434E (P < 0.0001), but not the 148M-434K, haplotype (P > 0.9), was associated with histological NAFLD and steatohepatitis. Both the I148M (P = 0.0002) and E434K variants (P = 0.044) were associated with serum ALT levels, by interacting with each other, in that the 434K hampered the association with liver damage of the 148M allele (P = 0.006). The E434K variant did not affect PNPLA3 enzymatic activity, but carriers of the rs2294918 A allele (434K) displayed lower hepatic PNPLA3 messenger RNA and protein levels (P < 0.05). CONCLUSIONS Rare loss-of-function PNPLA3 variants were not detected in early-onset NAFLD. However, PNPLA3 rs2294918 E434K decreased PNPLA3 expression, lessening the effect of the I148M variant on the predisposition to steatosis and liver damage. This suggests that the PNPLA3 I148M variant has a codominant negative effect on triglycerides mobilization from lipid droplets, mediated by inhibition of other lipases.
Collapse
Affiliation(s)
- Benedetta Donati
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Policlinico, Milano, Italy
| | - Benedetta Maria Motta
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Policlinico, Milano, Italy.,Sahlgrenska Center for Cardiovascular and Metabolic Research, Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Piero Pingitore
- Sahlgrenska Center for Cardiovascular and Metabolic Research, Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Marica Meroni
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Policlinico, Milano, Italy
| | - Alessandro Pietrelli
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Policlinico, Milano, Italy.,Istituto Nazionale Genetica Molecolare (INGM), "Romeo ed Enrica Invernizzi", Bioinformatic Group, Milano, Italy
| | - Anna Alisi
- Hepato-Metabolic Unit, Ospedale Bambin Gesù, Roma, Italy
| | - Salvatore Petta
- Department of Gastroenterology, Università di Palermo, Palermo, Italy
| | - Chao Xing
- UT Southwestern Medical Center, Dallas, TX
| | - Paola Dongiovanni
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Policlinico, Milano, Italy
| | - Benedetta del Menico
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Policlinico, Milano, Italy
| | - Raffaela Rametta
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Policlinico, Milano, Italy
| | - Rosellina Margherita Mancina
- Sahlgrenska Center for Cardiovascular and Metabolic Research, Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Sara Badiali
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Policlinico, Milano, Italy
| | - Anna Ludovica Fracanzani
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Policlinico, Milano, Italy
| | - Antonio Craxì
- Department of Gastroenterology, Università di Palermo, Palermo, Italy
| | - Silvia Fargion
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Policlinico, Milano, Italy
| | - Valerio Nobili
- Hepato-Metabolic Unit, Ospedale Bambin Gesù, Roma, Italy
| | - Stefano Romeo
- Sahlgrenska Center for Cardiovascular and Metabolic Research, Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden.,Clinical Nutrition Unit, Department of Medical and Surgical Sciences, University Magna Graecia, Catanzaro, Italy.,Department of Cardiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Luca Valenti
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Policlinico, Milano, Italy
| |
Collapse
|
30
|
Canavesi E, Porzio M, Ruscica M, Rametta R, Macchi C, Pelusi S, Fracanzani AL, Dongiovanni P, Fargion S, Magni P, Valenti L. Increased circulating adiponectin in males with chronic HCV hepatitis. Eur J Intern Med 2015; 26:635-9. [PMID: 26293833 DOI: 10.1016/j.ejim.2015.08.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 05/04/2015] [Accepted: 08/03/2015] [Indexed: 12/19/2022]
Abstract
BACKGROUND Increased levels of adiponectin, a major adipokine with insulin sensitizing properties showing a strong sexual dimorphism, have been reported in individuals with chronic HCV infection (CHC), but data are limited by small samples and lack of control for the genetic background and hepatic fibrosis. The aim of this study was to compare adiponectin levels between CHC patients and accurately matched controls. METHODS We considered 184 CHC patients, matched (1:1) for age, gender, body mass index, and Adiponectin genotype (ADIPOQ) with healthy individuals. To control for the severity of liver disease, a second control group consisting of 95 patients with histological nonalcoholic fatty liver disease (NAFLD) further matched (1:1) for severe fibrosis was exploited. ADIPOQ genotype was evaluated by Taqman assays, serum adiponectin measured by ELISA. RESULTS Serum adiponectin was higher in CHC patients than in healthy individuals (9.0±5.0 μg/ml vs. 7.3±4.0 μg/ml; p=0.001; adjusted estimate +1.8, 1.7-2.9; p=0.001), and than in NAFLD patients (8.3±4.5 μg/ml vs. 6.0±4.2 μg/ml; p<0.001; adjusted estimate +0.8, 0.2-1.4, p=0.006). After stratification for sex, serum adiponectin was higher in males with CHC than in healthy individuals and NAFLD patients (p<0.005 for both), whereas the difference was not significant in females. CONCLUSIONS CHC is associated with increased serum adiponectin independently of age, body mass, diabetes, ADIPOQ genotype, and of severe liver fibrosis, particularly in men.
Collapse
Affiliation(s)
- Elena Canavesi
- Internal Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico Milano, Department of Pathophysiology and Transplantation, Centro Malattie Metaboliche del Fegato, Università degli Studi di Milano, Milano, Italy.
| | - Marianna Porzio
- Internal Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico Milano, Department of Pathophysiology and Transplantation, Centro Malattie Metaboliche del Fegato, Università degli Studi di Milano, Milano, Italy.
| | - Massimiliano Ruscica
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, Italy.
| | - Raffaela Rametta
- Internal Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico Milano, Department of Pathophysiology and Transplantation, Centro Malattie Metaboliche del Fegato, Università degli Studi di Milano, Milano, Italy.
| | - Chiara Macchi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, Italy.
| | - Serena Pelusi
- Internal Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico Milano, Department of Pathophysiology and Transplantation, Centro Malattie Metaboliche del Fegato, Università degli Studi di Milano, Milano, Italy.
| | - Anna Ludovica Fracanzani
- Internal Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico Milano, Department of Pathophysiology and Transplantation, Centro Malattie Metaboliche del Fegato, Università degli Studi di Milano, Milano, Italy.
| | - Paola Dongiovanni
- Internal Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico Milano, Department of Pathophysiology and Transplantation, Centro Malattie Metaboliche del Fegato, Università degli Studi di Milano, Milano, Italy.
| | - Silvia Fargion
- Internal Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico Milano, Department of Pathophysiology and Transplantation, Centro Malattie Metaboliche del Fegato, Università degli Studi di Milano, Milano, Italy.
| | - Paolo Magni
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, Italy.
| | - Luca Valenti
- Internal Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico Milano, Department of Pathophysiology and Transplantation, Centro Malattie Metaboliche del Fegato, Università degli Studi di Milano, Milano, Italy.
| |
Collapse
|
31
|
Chettouh H, Lequoy M, Fartoux L, Vigouroux C, Desbois-Mouthon C. Hyperinsulinaemia and insulin signalling in the pathogenesis and the clinical course of hepatocellular carcinoma. Liver Int 2015; 35:2203-17. [PMID: 26123841 DOI: 10.1111/liv.12903] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 06/09/2015] [Indexed: 12/11/2022]
Abstract
Hepatocellular carcinoma (HCC) is the fifth most prevalent cancer and is one of the leading causes of cancer-related death. The risk factors for HCC include cirrhosis, chronic viral hepatitis, heavy alcohol intake and metabolic diseases such as obesity, type 2 diabetes and metabolic syndrome. Insulin resistance is a common denominator of all of these conditions and is tethered to hyperinsulinaemia. Here, we give an overview of the recent advances linking hyperinsulinaemia to HCC development and progression. In particular, we summarise the underlying causes of hyperinsulinaemia in the setting of chronic liver diseases. We present epidemiological evidence linking metabolic diseases to HCC risk and HCC-related mortality, as well as the pathogenic cellular and molecular mechanisms explaining this relation. A better understanding of the mechanisms by which insulin participates in HCC biology might ultimately provide novel opportunities for prevention and treatment.
Collapse
Affiliation(s)
- Hamza Chettouh
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, Paris, France.,INSERM, UMR_S 938, Centre de Recherche Saint-Antoine, Paris, France
| | - Marie Lequoy
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, Paris, France.,INSERM, UMR_S 938, Centre de Recherche Saint-Antoine, Paris, France.,Service d'Hépatologie, AP-HP, Hôpital Saint-Antoine, Paris, France
| | - Laetitia Fartoux
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, Paris, France.,INSERM, UMR_S 938, Centre de Recherche Saint-Antoine, Paris, France.,Service d'Hépatologie, AP-HP, Hôpital Saint-Antoine, Paris, France
| | - Corinne Vigouroux
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, Paris, France.,INSERM, UMR_S 938, Centre de Recherche Saint-Antoine, Paris, France.,Laboratoire Commun de Biologie et Génétique Moléculaires AP-HP, Hôpital Saint-Antoine, Paris, France.,ICAN, Institute of Cardiometabolism and Nutrition, Paris, France
| | - Christèle Desbois-Mouthon
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, Centre de Recherche Saint-Antoine, Paris, France.,INSERM, UMR_S 938, Centre de Recherche Saint-Antoine, Paris, France
| |
Collapse
|
32
|
CHO JINKYUNG, KIM SHINUK, LEE SHINHO, KANG HYUNSIK. Effect of Training Intensity on Nonalcoholic Fatty Liver Disease. Med Sci Sports Exerc 2015; 47:1624-34. [DOI: 10.1249/mss.0000000000000595] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
33
|
Banerji J. Asparaginase treatment side-effects may be due to genes with homopolymeric Asn codons (Review-Hypothesis). Int J Mol Med 2015; 36:607-26. [PMID: 26178806 PMCID: PMC4533780 DOI: 10.3892/ijmm.2015.2285] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 07/15/2015] [Indexed: 12/14/2022] Open
Abstract
The present treatment of childhood T-cell leukemias involves the systemic administration of prokary-otic L-asparaginase (ASNase), which depletes plasma Asparagine (Asn) and inhibits protein synthesis. The mechanism of therapeutic action of ASNase is poorly understood, as are the etiologies of the side-effects incurred by treatment. Protein expression from genes bearing Asn homopolymeric coding regions (N-hCR) may be particularly susceptible to Asn level fluctuation. In mammals, N-hCR are rare, short and conserved. In humans, misfunctions of genes encoding N-hCR are associated with a cluster of disorders that mimic ASNase therapy side-effects which include impaired glycemic control, dislipidemia, pancreatitis, compromised vascular integrity, and neurological dysfunction. This paper proposes that dysregulation of Asn homeostasis, potentially even by ASNase produced by the microbiome, may contribute to several clinically important syndromes by altering expression of N-hCR bearing genes. By altering amino acid abundance and modulating ribosome translocation rates at codon repeats, the microbiomic environment may contribute to genome decoding and to shaping the proteome. We suggest that impaired translation at poly Asn codons elevates diabetes risk and severity.
Collapse
Affiliation(s)
- Julian Banerji
- Center for Computational and Integrative Biology, MGH, Simches Research Center, Boston, MA 02114, USA
| |
Collapse
|
34
|
Fares R, Petta S, Lombardi R, Grimaudo S, Dongiovanni P, Pipitone R, Rametta R, Fracanzani AL, Mozzi E, Craxì A, Fargion S, Sesti G, Valenti L. The UCP2 -866 G>A promoter region polymorphism is associated with nonalcoholic steatohepatitis. Liver Int 2015; 35:1574-80. [PMID: 25351290 DOI: 10.1111/liv.12707] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 10/24/2014] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Uncoupling protein 2 - UCP2 - regulates mitochondrial lipid fluxes and reactive oxygen species production by the respiratory chain. The -866 G>A UCP2 promoter region polymorphism has been linked to insulin resistance and lipid metabolism. The aim of this study was to assess whether the -866 G>A UCP2 polymorphism predisposes to nonalcoholic steatohepatitis in patients at risk, and the relationship with lipid metabolism and hepatic UCP2 expression. METHODS We considered 688 Italian patients who underwent liver biopsy for suspected NASH, and 232 healthy controls. The UCP2 -866 G>A polymorphism was determined by allele specific oligonucleotide probes, hepatic UCP2 mRNA levels by quantitative real-time PCR. RESULTS UCP2 A/A genotype was associated with a reduced risk of nonalcoholic steatohepatitis (Odds Ratio 0.49, 95% C.I. 0.26-0.90; P = 0.02; adjusted for age, sex, BMI, impaired fasting glucose or diabetes, PNPLA3 I148M alleles and recruitment centre). The A/A genotype was associated with reduced risk of steatosis grade G2-G3 and nonalcoholic steatohepatitis in patients without (P = 0.003 and P = 0.01 respectively), but not in those with (P = NS) impaired fasting glucose/diabetes. The UCP2 A/A genotype was associated with higher hepatic UCP2 mRNA levels (adjusted P = 0.008). Concerning the metabolic traits, the UCP2 A/A genotype was associated with higher total serum cholesterol levels (adjusted P = 0.03), but not with serum HDL, triglycerides or impaired fasting glucose/diabetes. CONCLUSIONS UCP2 -866 A/A genotype is associated with increased hepatic UCP2 expression and reduced risk of nonalcoholic steatohepatitis, particularly in subjects with normal fasting glucose.
Collapse
Affiliation(s)
- Roberta Fares
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Policlinico Milano, Milan, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Mancina RM, Matikainen N, Maglio C, Söderlund S, Lundbom N, Hakkarainen A, Rametta R, Mozzi E, Fargion S, Valenti L, Romeo S, Taskinen MR, Borén J. Paradoxical dissociation between hepatic fat content and de novo lipogenesis due to PNPLA3 sequence variant. J Clin Endocrinol Metab 2015; 100:E821-5. [PMID: 25763607 DOI: 10.1210/jc.2014-4464] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
CONTEXT Nonalcoholic fatty liver disease (NAFLD) is an emerging epidemic disease characterized by increased hepatic fat, due to an imbalance between synthesis and removal of hepatic lipids. In particular, increased hepatic de novo lipogenesis (DNL) is a key feature associated with NAFLD. The genetic variations I148M in PNPLA3 and E167K in TM6SF2 confer susceptibility to NAFLD. OBJECTIVE Here we aimed to investigate the contribution of DNL to liver fat accumulation in the PNPLA3 I148M or TM6SF2 E167K genetic determinants of NAFLD. PATIENTS AND METHODS The PNPLA3 I148M and TM6SF2 E167K were genotyped in two well-characterized cohorts of Europeans. In the first cohort (Helsinki cohort; n = 88), we directly quantified hepatic DNL using deuterated water. In the second cohort (Milan cohort; n = 63), we quantified the hepatic expression of SREBP1c that we have found previously associated with increased fat content. Liver fat was measured by magnetic resonance proton spectroscopy in the Helsinki cohort, and by histological assessment of liver biopsies in the Milan cohort. RESULTS PNPLA3 148M was associated with lower DNL and expression of the lipogenic transcription factor SREBP1c despite substantial increased hepatic fat content. CONCLUSIONS Our data show a paradoxical dissociation between hepatic DNL and hepatic fat content due to the PNPLA3 148M allele indicating that increased DNL is not a key feature in all individuals with hepatic steatosis, and reinforces the contribution of decreased mobilization of hepatic triglycerides for hepatic lipid accumulation in subject with the PNPLA3 148M allele.
Collapse
Affiliation(s)
- Rosellina M Mancina
- Department of Molecular and Clinical Medicine (R.M.M, C.M, S.R., J.B.), University of Gothenburg, S-413 45 Gothenburg, Sweden; Department of Medicine, Cardiovascular Research Unit, Diabetes and Obesity Research Program (N.M., S.S., M.-R.T.), Heart and Lung Centre and Division of Endocrinology and Helsinki University Central Hospital, University of Helsinki, 00100 Helsinki, Finland; Department of Radiology, HUS Medical Imaging Center (N.L., A.H.), Helsinki University Central Hospital, University of Helsinki, 00100 Helsinki, Finland; Departments of Internal Medicine (R.R, S.F., L.V.), and General Surgery (E.M.), Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Ca' Granda Ospedale Policlinico, and Department of Pathophysiology and Transplantation Università degli Studi di Milano, 20122 Milano, Italy; and Department of Medical and Surgical Sciences (S.R.), Clinical Nutrition Unit, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Valenti L, Mendoza RM, Rametta R, Maggioni M, Kitajewski C, Shawber CJ, Pajvani UB. Hepatic notch signaling correlates with insulin resistance and nonalcoholic fatty liver disease. Diabetes 2013; 62:4052-62. [PMID: 23990360 PMCID: PMC3837035 DOI: 10.2337/db13-0769] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Hepatic Notch signaling is inappropriately activated in obese/insulin-resistant mouse models. Genetic or pharmacologic inhibition of hepatic Notch signaling in obese mice simultaneously improves glucose tolerance and reduces hepatic triglyceride content. As such, we predicted that Notch signaling in human liver would be positively associated with insulin resistance and hepatic steatosis. Here, we systematically survey Notch signaling in liver biopsy specimens, and show active Notch signaling in lean and obese adults, with expression of multiple Notch receptors and ligands. In morbidly obese patients undergoing bariatric surgery, we show that Notch activation positively correlates with glucose-6-phosphatase (G6PC) and phosphoenolpyruvate carboxykinase (PCK1) expression, key regulators of hepatic glucose output. We used immunofluorescence to identify active Notch signaling in hepatocytes and show highest activity in hyperglycemia, which we confirmed is a direct effect of hyperglycemia and insulin resistance. In a validation cohort of leaner individuals undergoing percutaneous liver biopsy for suspected nonalcoholic fatty liver disease (NAFLD), Notch activity showed independent positive association with insulin resistance and hepatic steatosis. Notably, Notch activity showed stronger correlation with the NAFLD activity score and alanine aminotransferase levels than with steatosis alone, suggesting that Notch activity is associated with nonalcoholic steatohepatitis. In summary, this study establishes that Notch signaling is activated in and may represent a therapeutic target for patients with obesity-related liver disease.
Collapse
Affiliation(s)
- Luca Valenti
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Internal Medicine, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Ca’ Granda, Milano, Italy
| | - Rosa M. Mendoza
- Department of Medicine, Columbia University, New York, New York
| | - Raffaela Rametta
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Internal Medicine, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Ca’ Granda, Milano, Italy
| | - Marco Maggioni
- Department of Pathology, Università degli Studi di Milano, Internal Medicine, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Ca’ Granda, Milano, Italy
| | - Chris Kitajewski
- Department of Obstetrics and Gynecology, Columbia University, New York, New York
| | - Carrie J. Shawber
- Department of Obstetrics and Gynecology, Columbia University, New York, New York
| | - Utpal B. Pajvani
- Department of Medicine, Columbia University, New York, New York
- Corresponding author: Utpal B. Pajvani,
| |
Collapse
|
37
|
Dietary anthocyanins as nutritional therapy for nonalcoholic fatty liver disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2013; 2013:145421. [PMID: 24282628 PMCID: PMC3824564 DOI: 10.1155/2013/145421] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Accepted: 09/03/2013] [Indexed: 02/07/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD), defined by excessive lipid accumulation in the liver, is the hepatic manifestation of insulin resistance and the metabolic syndrome. Due to the epidemics of obesity, NAFLD is rapidly becoming the leading cause of altered liver enzymes in Western countries. NAFLD encompasses a wide spectrum of liver disease ranging from simple uncomplicated steatosis, to steatohepatitis, cirrhosis, and hepatocellular carcinoma. Diet may affect the development of NAFLD either by increasing risk or by providing protective factors. Therefore, it is important to investigate the role of foods and/or food bioactives on the metabolic processes involved in steatohepatitis for preventive strategies. It has been reported that anthocyanins (ACNs) decrease hepatic lipid accumulation and may counteract oxidative stress and hepatic inflammation, but their impact on NAFLD has yet to be fully determined. ACNs are water-soluble bioactive compounds of the polyphenol class present in many vegetable products. Here, we summarize the evidence evaluating the mechanisms of action of ACNs on hepatic lipid metabolism in different experimental setting: in vitro, in vivo, and in human trials. Finally, a working model depicting the possible mechanisms underpinning the beneficial effects of ACNs in NAFLD is proposed, based on the available literature.
Collapse
|