1
|
Ji L, Wei J, Zhang R, Zhang X, Gao Y, Fang M, Yu Z, Cao L, Gao Y, Li M. Bushen Formula promotes the decrease of HBsAg levels in patients with CHB by regulating Tfh cells and B-cell subsets. JOURNAL OF ETHNOPHARMACOLOGY 2024; 328:118072. [PMID: 38508431 DOI: 10.1016/j.jep.2024.118072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 03/07/2024] [Accepted: 03/18/2024] [Indexed: 03/22/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Bushen Formula (BSF) is the effective traditional Chinese medicine (TCM) for chronic hepatitis B (CHB) according to our previous researches. However, the special effectiveness of BSF treating CHB patients in different stages and the immunoregulatory mechanisms remain to be explored. AIM OF THE STUDY To compare the therapeutic effects of BSF in both treatment-naive patients and Peg-IFN-α-treated patients, and explore the potential mechanism of immunomodulation. MATERIALS AND METHODS Ultra-high performance liquid chromatography-quadrupole electrostatic field-orbital trap high resolution mass spectrometry and the TCMSP database were used to determine the main components of BSF. Two hundred and sixty-six patients were enrolled in the retrospective study, and they were divided into the treatment group (T-Group, BSF plus Peg-IFN-α) and the control group (C-Group, Peg-IFN-α monotherapy). Within each group, patients were further grouped into subgroups, namely T1/C1 groups (treatment-naive patients, T1 = 34, C1 = 94) and T2/C2 groups (Peg-IFN-α-treated patients, T2 = 56, C2 = 82). Serum HBV markers, serum HBV DNA levels, serum ALT/AST and TCM symptoms were obtained from the record. Bioinformatics analysis was employed to obtain the potential immunoregulatory mechanisms of BSF treating CHB patients. Among patients in T2 and C2 group, peripheral mononuclear cells from 36 patients were used to analyze the characteristics of peripheral follicular helper T (Tfh) cells and B-cell subtypes by flow cytometry. Preparation of BSF-containing serum in rats. In vitro, the co-culture system of CXCR5+ cells and HepG2.2.15 cells was built to investigate the immunoregulatory effects of BSF. RESULTS A total of 14 main active compounds were detected in BSF, which were deemed critical for the treatment of CHB. Our findings indicated that the T2-Group exhibited the higher percentage of HBsAg decline ≥ 1-log10 IU/ml and rate of HBeAg seroclearance compared to the C2-Group (35.7% vs. 15.9%, P = 0.033; 33.9% vs. 11.0%, P = 0.002). Additionally, the T2-Group demonstrated the higher percentage of HBsAg decline ≥ 1-log10 IU/ml and rate of HBeAg seroclearance compared to the T1-Group (35.7% vs. 14.7%, P = 0.031; 33.9% vs. 2.9%, P = 0.000). The total effective rate based on TCM clinical syndrome in T1-Group and T2-Group were significantly greater than those in C1-Group and C2-Group (85.3% vs. 61.7%, P = 0.012; 89.1% vs. 63.4%, P = 0.000). Bioinformatics analysis indicated that the immunoregulatory mechanisms of BSF treating CHB patients were mainly linked to the growth and stimulation of B-cell, T-cell differentiation, and the signaling pathway of the B-cell receptor. Furthermore, the frequencies of Tfh cells and its IL-21 level, and the IL-21R expressed by B-cell were all increased after BSF treatment. Additionally, in the co-culture system of CXCR5+ cells and HepG2.2.15 cells, HBsAg and HBeAg levels were decreased after BSF-containing serum treatment,as well as the up-regulating of Tfh cell frequencies and down-regulating of B-cell frequencies. CONCLUSIONS BSF have the higher percentage of HBsAg decline and HBeAg seroclearance in Peg-IFN-α-treated patients compared with treatment-naive patients. The potential immunoregulatory mechanism may correlate with promoting the interaction between Tfh cells and B-cell through IL-21/IL-21R signaling pathway.
Collapse
Affiliation(s)
- Longshan Ji
- Laboratory of Cellular Immunity, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education, Shanghai, 201203, China
| | - Jinghan Wei
- Laboratory of Cellular Immunity, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education, Shanghai, 201203, China
| | - Rongjie Zhang
- Laboratory of Cellular Immunity, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education, Shanghai, 201203, China
| | - Xin Zhang
- Laboratory of Cellular Immunity, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education, Shanghai, 201203, China
| | - Yating Gao
- Laboratory of Cellular Immunity, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education, Shanghai, 201203, China
| | - Miao Fang
- Laboratory of Cellular Immunity, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education, Shanghai, 201203, China
| | - Zhuo Yu
- Department of Hepatopathy, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Lin Cao
- Laboratory of Cellular Immunity, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education, Shanghai, 201203, China
| | - Yueqiu Gao
- Laboratory of Cellular Immunity, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education, Shanghai, 201203, China; Department of Hepatopathy, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Institute of Infectious Diseases of Integrated Traditional Chinese and Western Medicine, Shanghai Institute of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Man Li
- Laboratory of Cellular Immunity, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education, Shanghai, 201203, China.
| |
Collapse
|
2
|
Yen TH, Lee SH, Tang CH, Liang HJ, Lin CY. Lipid responses to perfluorooctane sulfonate exposure for multiple rat organs. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 277:116368. [PMID: 38669874 DOI: 10.1016/j.ecoenv.2024.116368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 03/16/2024] [Accepted: 04/20/2024] [Indexed: 04/28/2024]
Abstract
Perfluorooctane sulfonate (PFOS) is a persistent chemical that has long been a threat to human health. However, the molecular effects of PFOS on various organs are not well studied. In this study, male Sprague-Dawley rats were treated with various doses of PFOS through gavage for 21 days. Subsequently, the liver, lung, heart, kidney, pancreas, testis, and serum of the rats were harvested for lipid analysis. We applied a focusing lipidomic analytical strategy to identify key lipid responses of phosphorylcholine-containing lipids, including phosphatidylcholines and sphingomyelins. Partial least squares discriminant analysis revealed that the organs most influenced by PFOS exposure were the liver, kidney, and testis. Changes in the lipid profiles of the rats indicated that after exposure, levels of diacyl-phosphatidylcholines and 22:6-containing phosphatidylcholines in the liver, kidney, and testis of the rats decreased, whereas the level of 20:3-containing phosphatidylcholines increased. Furthermore, levels of polyunsaturated fatty acids-containing plasmenylcholines decreased. Changes in sphingomyelin levels indicated organ-dependent responses. Decreased levels of sphingomyelins in the liver, nonmonotonic dose responses in the kidney, and irregular responses in the testis after PFOS exposure are observed. These lipid responses may be associated with alterations pertaining to phosphatidylcholine synthesis, fatty acid metabolism, membrane properties, and oxidative stress in the liver, kidney, and testis. Lipid responses in the liver could have contributed to the observed increase in liver to body weight ratios. The findings suggest potential toxicity and possible mechanisms associated with PFOS in multiple organs.
Collapse
Affiliation(s)
- Tzu-Hsin Yen
- Institute of Environmental and Occupational Health Sciences, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Sheng-Han Lee
- School of Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Chuan-Ho Tang
- National Museum of Marine Biology and Aquarium, Pingtung, Taiwan; Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Hao-Jan Liang
- Institute of Environmental and Occupational Health Sciences, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Ching-Yu Lin
- Institute of Environmental and Occupational Health Sciences, College of Public Health, National Taiwan University, Taipei, Taiwan; Department of Public Health, College of Public Health, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
3
|
Li H, Liu Y, Meng F, Chen J, Han X. Adrenarche-accompanied rise of adrenal sex steroid precursors prevents NAFLD in Young Female rats by converting into active androgens and inactivating hepatic Srebf1 signaling. BMC Genomics 2024; 25:190. [PMID: 38369486 PMCID: PMC10875776 DOI: 10.1186/s12864-024-10107-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 02/09/2024] [Indexed: 02/20/2024] Open
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) has rapidly become the most common cause of chronic liver disease in children and adolescents, but its etiology remains largely unknown. Adrenarche is a critical phase for hormonal changes, and any disturbance during this period has been linked to metabolic disorders, including obesity and dyslipidemia. However, whether there is a causal linkage between adrenarche disturbance and the increasing prevalence of NAFLD in children remains unclear. RESULTS Using the young female rat as a model, we found that the liver undergoes a transient slowdown period of growth along with the rise of adrenal-derived sex steroid precursors during adrenarche. Specifically blocking androgen actions across adrenarche phase using androgen receptor antagonist flutamide largely increased liver weight by 47.97% and caused marked fat deposition in liver, thus leading to severe NAFLD in young female rats. Conversely, further administrating nonaromatic dihydrotestosterone (DHT) into young female rats across adrenarche phase could effectively reduce liver fat deposition. But, administration of the aromatase inhibitor, formestane across adrenarche had minimal effects on hepatic de novo fatty acid synthesis and liver fat deposition, suggesting adrenal-derived sex steroid precursors exert their anti-NAFLD effects in young females by converting into active androgens rather than into active estrogens. Mechanistically, transcriptomic profiling and integrated data analysis revealed that active androgens converted from the adrenal sex steroid precursors prevent NAFLD in young females primarily by inactivating hepatic sterol regulatory element-binding transcription factor 1 (Srebf1) signaling. CONCLUSIONS We firstly evidenced that adrenarche-accompanied rise of sex steroid precursors plays a predominant role in preventing the incidence of NAFLD in young females by converting into active androgens and inactivating hepatic Srebf1 signaling. Our novel finding provides new insights into the etiology of NAFLD and is crucial in developing effective prevention and management strategies for NAFLD in children.
Collapse
Affiliation(s)
- Haoqing Li
- College of Life Science, Sichuan Agricultural University, Ya'an, 625014, China
| | - Yingyu Liu
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Fengyan Meng
- College of Life Science, Sichuan Agricultural University, Ya'an, 625014, China
| | - Junan Chen
- College of Life Science, Sichuan Agricultural University, Ya'an, 625014, China
| | - Xingfa Han
- College of Life Science, Sichuan Agricultural University, Ya'an, 625014, China.
| |
Collapse
|
4
|
Kaffe E, Tisi A, Magkrioti C, Aidinis V, Mehal WZ, Flavell RA, Maccarrone M. Bioactive signalling lipids as drivers of chronic liver diseases. J Hepatol 2024; 80:140-154. [PMID: 37741346 DOI: 10.1016/j.jhep.2023.08.029] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 08/23/2023] [Accepted: 08/28/2023] [Indexed: 09/25/2023]
Abstract
Lipids are important in multiple cellular functions, with most having structural or energy storage roles. However, a small fraction of lipids exert bioactive roles through binding to G protein-coupled receptors and induce a plethora of processes including cell proliferation, differentiation, growth, migration, apoptosis, senescence and survival. Bioactive signalling lipids are potent modulators of metabolism and energy homeostasis, inflammation, tissue repair and malignant transformation. All these events are involved in the initiation and progression of chronic liver diseases. In this review, we focus specifically on the roles of bioactive lipids derived from phospholipids (lyso-phospholipids) and poly-unsaturated fatty acids (eicosanoids, pro-resolving lipid mediators and endocannabinoids) in prevalent chronic liver diseases (alcohol-associated liver disease, non-alcoholic fatty liver disease, viral hepatitis and hepatocellular carcinoma). We discuss the balance between pathogenic and beneficial bioactive lipids as well as potential therapeutic targets related to the agonism or antagonism of their receptors.
Collapse
Affiliation(s)
- Eleanna Kaffe
- Department of Immunobiology, Yale University School of Medicine, 06511, New Haven, CT, USA.
| | - Annamaria Tisi
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100, L'Aquila, Italy
| | | | - Vassilis Aidinis
- Biomedical Sciences Research Center Alexander Fleming, 16672, Athens, Greece
| | - Wajahat Z Mehal
- Department of Internal Medicine, Section of Digestive Diseases, Yale University, New Haven, CT, 06520, USA; Veterans Affairs Medical Center, West Haven, CT, 06516, USA
| | - Richard A Flavell
- Department of Immunobiology, Yale University School of Medicine, 06511, New Haven, CT, USA; Howard Hughes Medical Institute, Yale School of Medicine, New Haven, CT, 06519, USA
| | - Mauro Maccarrone
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100, L'Aquila, Italy; Laboratory of Lipid Neurochemistry, European Center for Brain Research (CERC), Santa Lucia Foundation IRCCS, 00143 Rome, Italy.
| |
Collapse
|
5
|
Mocciaro G, Allison M, Jenkins B, Azzu V, Huang-Doran I, Herrera-Marcos LV, Hall Z, Murgia A, Susan D, Frontini M, Vidal-Puig A, Koulman A, Griffin JL, Vacca M. Non-alcoholic fatty liver disease is characterised by a reduced polyunsaturated fatty acid transport via free fatty acids and high-density lipoproteins (HDL). Mol Metab 2023; 73:101728. [PMID: 37084865 PMCID: PMC10176260 DOI: 10.1016/j.molmet.2023.101728] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 03/25/2023] [Accepted: 04/13/2023] [Indexed: 04/23/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Non-alcoholic fatty liver disease (NAFLD) develops due to impaired hepatic lipid fluxes and is a risk factor for chronic liver disease and atherosclerosis. Lipidomic studies consistently reported characteristic hepatic/VLDL "lipid signatures" in NAFLD; whole plasma traits are more debated. Surprisingly, the HDL lipid composition by mass spectrometry has not been characterised across the NAFLD spectrum, despite HDL being a possible source of hepatic lipids delivered from peripheral tissues alongside free fatty acids (FFA). This study characterises the HDL lipidomic signature in NAFLD, and its correlation with metabolic and liver disease markers. METHODS We used liquid chromatography-mass spectrometry to determine the whole serum and HDL lipidomic profile in 89 biopsy-proven NAFLD patients and 20 sex and age-matched controls. RESULTS In the whole serum of NAFLD versus controls, we report a depletion in polyunsaturated (PUFA) phospholipids (PL) and FFA; with PUFA PL being also lower in HDL, and negatively correlated with BMI, insulin resistance, triglycerides, and hepatocyte ballooning. In the HDL of the NAFLD group we also describe higher saturated ceramides, which positively correlate with insulin resistance and transaminases. CONCLUSION NAFLD features lower serum lipid species containing polyunsaturated fatty acids; the most affected lipid fractions are FFA and (HDL) phospholipids; our data suggest a possible defect in the transfer of PUFA from peripheral tissues to the liver in NAFLD. Mechanistic studies are required to explore the biological implications of our findings addressing if HDL composition can influence liver metabolism and damage, thus contributing to NAFLD pathophysiology.
Collapse
Affiliation(s)
- Gabriele Mocciaro
- University of Cambridge, Department of Biochemistry, Cambridge, CB2 1GA, United Kingdom; Roger Williams Institute of Hepatology, Foundation for Liver Research, London, SE5 9NT, United Kingdom
| | - Michael Allison
- Addenbrooke's Hospital, Cambridge Biomedical Research Centre, Department of Medicine, United Kingdom
| | - Benjamin Jenkins
- Wellcome Trust-MRC Institute of Metabolic Science Metabolic Research Laboratories, Cambridge, CB2 0QQ, United Kingdom
| | - Vian Azzu
- Addenbrooke's Hospital, Cambridge Biomedical Research Centre, Department of Medicine, United Kingdom; Wellcome Trust-MRC Institute of Metabolic Science Metabolic Research Laboratories, Cambridge, CB2 0QQ, United Kingdom
| | - Isabel Huang-Doran
- Addenbrooke's Hospital, Cambridge Biomedical Research Centre, Department of Medicine, United Kingdom
| | - Luis Vicente Herrera-Marcos
- Department of Biochemistry and Molecular and Cellular Biology, Veterinary Faculty, University of Zaragoza, Zaragoza, 50013, Spain
| | - Zoe Hall
- Biomolecular Medicine, Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, SW7 2AZ, United Kingdom
| | - Antonio Murgia
- University of Cambridge, Department of Biochemistry, Cambridge, CB2 1GA, United Kingdom
| | - Davies Susan
- Addenbrooke's Hospital, Cambridge Biomedical Research Centre, Department of Medicine, United Kingdom
| | - Mattia Frontini
- Faculty of Health and Life Sciences, Clinical and Biomedical Sciences, University of Exeter Medical School, RILD Building, Barrack Road, Exeter, EX2 5DW, United Kingdom
| | - Antonio Vidal-Puig
- Wellcome Trust-MRC Institute of Metabolic Science Metabolic Research Laboratories, Cambridge, CB2 0QQ, United Kingdom
| | - Albert Koulman
- Wellcome Trust-MRC Institute of Metabolic Science Metabolic Research Laboratories, Cambridge, CB2 0QQ, United Kingdom.
| | - Julian L Griffin
- University of Cambridge, Department of Biochemistry, Cambridge, CB2 1GA, United Kingdom; The Rowett Institute, Foresterhill Campus, University of Aberdeen, Aberdeen, AB25 2ZD, United Kingdom.
| | - Michele Vacca
- University of Cambridge, Department of Biochemistry, Cambridge, CB2 1GA, United Kingdom; Roger Williams Institute of Hepatology, Foundation for Liver Research, London, SE5 9NT, United Kingdom; Wellcome Trust-MRC Institute of Metabolic Science Metabolic Research Laboratories, Cambridge, CB2 0QQ, United Kingdom; Aldo Moro University of Bari, Department of Interdisciplinary Medicine, Clinica Medica "C. Frugoni", Bari, 70124, Italy.
| |
Collapse
|
6
|
Ducheix S, Piccinin E, Peres C, Garcia‐Irigoyen O, Bertrand‐Michel J, Fouache A, Cariello M, Lobaccaro J, Guillou H, Sabbà C, Ntambi JM, Moschetta A. Reduction in gut-derived MUFAs via intestinal stearoyl-CoA desaturase 1 deletion drives susceptibility to NAFLD and hepatocarcinoma. Hepatol Commun 2022; 6:2937-2949. [PMID: 35903850 PMCID: PMC9512486 DOI: 10.1002/hep4.2053] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/21/2022] [Accepted: 07/06/2022] [Indexed: 11/15/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is defined by a set of hepatic conditions ranging from steatosis to steatohepatitis (NASH), characterized by inflammation and fibrosis, eventually predisposing to hepatocellular carcinoma (HCC). Together with fatty acids (FAs) originated from adipose lipolysis and hepatic lipogenesis, intestinal-derived FAs are major contributors of steatosis. However, the role of mono-unsaturated FAs (MUFAs) in NAFLD development is still debated. We previously established the intestinal capacity to produce MUFAs, but its consequences in hepatic functions are still unknown. Here, we aimed to determine the role of the intestinal MUFA-synthetizing enzyme stearoyl-CoA desaturase 1 (SCD1) in NAFLD. We used intestinal-specific Scd1-KO (iScd1-/- ) mice and studied hepatic dysfunction in different models of steatosis, NASH, and HCC. Intestinal-specific Scd1 deletion decreased hepatic MUFA proportion. Compared with controls, iScd1-/- mice displayed increased hepatic triglyceride accumulation and derangement in cholesterol homeostasis when fed a MUFA-deprived diet. Then, on Western diet feeding, iScd1-/- mice triggered inflammation and fibrosis compared with their wild-type littermates. Finally, intestinal-Scd1 deletion predisposed mice to liver cancer. Conclusions: Collectively, these results highlight the major importance of intestinal MUFA metabolism in maintaining hepatic functions and show that gut-derived MUFAs are protective from NASH and HCC.
Collapse
Affiliation(s)
- Simon Ducheix
- Department of Interdisciplinary MedicineUniversity of Bari “Aldo Moro”BariItaly
| | - Elena Piccinin
- Department of Basic Medical Science, Neurosciences, and Sense organsUniversity of Bari “Aldo Moro”BariItaly
| | - Claudia Peres
- INBB, National Institute for Biostructures and BiosystemsRomeItaly
| | | | - Justine Bertrand‐Michel
- MetaboHUB‐MetaToul, National Infrastructure of Metabolomics and FluxomicsToulouseFrance
- I2MC, Université de Toulouse, InsermUniversité Toulouse III–Paul SabatierToulouseFrance
| | - Allan Fouache
- INSERM U 1103, CNRS, UMR 6293Université Clermont AuvergneGReDAubièreFrance
- Centre de Recherche en Nutrition Humaine d'AuvergneClermont‐FerrandFrance
| | - Marica Cariello
- Department of Interdisciplinary MedicineUniversity of Bari “Aldo Moro”BariItaly
| | - Jean‐Marc Lobaccaro
- INSERM U 1103, CNRS, UMR 6293Université Clermont AuvergneGReDAubièreFrance
- Centre de Recherche en Nutrition Humaine d'AuvergneClermont‐FerrandFrance
| | - Hervé Guillou
- Integrative Toxicology and Metabolism TeamToxalim (Research Centre in Food Toxicology)Université de Toulouse, INRA, ENVTINP‐Purpan, UPSToulouseFrance
| | - Carlo Sabbà
- Department of Interdisciplinary MedicineUniversity of Bari “Aldo Moro”BariItaly
| | - James M. Ntambi
- Departments of Biochemistry and of Nutritional SciencesUniversity of Wisconsin MadisonMadisonWisconsinUSA
| | - Antonio Moschetta
- Department of Interdisciplinary MedicineUniversity of Bari “Aldo Moro”BariItaly
- INBB, National Institute for Biostructures and BiosystemsRomeItaly
| |
Collapse
|
7
|
Personnaz J, Piccolo E, Dortignac A, Iacovoni JS, Mariette J, Rocher V, Polizzi A, Batut A, Deleruyelle S, Bourdens L, Delos O, Combes-Soia L, Paccoud R, Moreau E, Martins F, Clouaire T, Benhamed F, Montagner A, Wahli W, Schwabe RF, Yart A, Castan-Laurell I, Bertrand-Michel J, Burlet-Schiltz O, Postic C, Denechaud PD, Moro C, Legube G, Lee CH, Guillou H, Valet P, Dray C, Pradère JP. Nuclear HMGB1 protects from nonalcoholic fatty liver disease through negative regulation of liver X receptor. SCIENCE ADVANCES 2022; 8:eabg9055. [PMID: 35333579 PMCID: PMC8956270 DOI: 10.1126/sciadv.abg9055] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 02/03/2022] [Indexed: 06/14/2023]
Abstract
Dysregulations of lipid metabolism in the liver may trigger steatosis progression, leading to potentially severe clinical consequences such as nonalcoholic fatty liver diseases (NAFLDs). Molecular mechanisms underlying liver lipogenesis are very complex and fine-tuned by chromatin dynamics and multiple key transcription factors. Here, we demonstrate that the nuclear factor HMGB1 acts as a strong repressor of liver lipogenesis. Mice with liver-specific Hmgb1 deficiency display exacerbated liver steatosis, while Hmgb1-overexpressing mice exhibited a protection from fatty liver progression when subjected to nutritional stress. Global transcriptome and functional analysis revealed that the deletion of Hmgb1 gene enhances LXRα and PPARγ activity. HMGB1 repression is not mediated through nucleosome landscape reorganization but rather via a preferential DNA occupation in a region carrying genes regulated by LXRα and PPARγ. Together, these findings suggest that hepatocellular HMGB1 protects from liver steatosis development. HMGB1 may constitute a new attractive option to therapeutically target the LXRα-PPARγ axis during NAFLD.
Collapse
Affiliation(s)
- Jean Personnaz
- Institut RESTORE, UMR 1301, Institut National de la Santé et de la Recherche Médicale (INSERM), CNRS-Université Paul Sabatier, Université de Toulouse, Toulouse, France
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1297/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, Toulouse, France
| | - Enzo Piccolo
- Institut RESTORE, UMR 1301, Institut National de la Santé et de la Recherche Médicale (INSERM), CNRS-Université Paul Sabatier, Université de Toulouse, Toulouse, France
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1297/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, Toulouse, France
| | - Alizée Dortignac
- Institut RESTORE, UMR 1301, Institut National de la Santé et de la Recherche Médicale (INSERM), CNRS-Université Paul Sabatier, Université de Toulouse, Toulouse, France
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1297/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, Toulouse, France
| | - Jason S. Iacovoni
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1297/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, Toulouse, France
| | - Jérôme Mariette
- MIAT, Université de Toulouse, INRAE, 31326 Castanet-Tolosan, France
| | - Vincent Rocher
- Molecular, Cellular, and Developmental Biology Unit (MCD), Centre de Biologie Intégrative (CBI), UPS, CNRS, Toulouse, France
| | - Arnaud Polizzi
- Toxalim, INRAE UMR 1331, ENVT, INP-Purpan, University of Toulouse, Paul Sabatier University, F-31027, Toulouse, France
| | - Aurélie Batut
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1297/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, Toulouse, France
| | - Simon Deleruyelle
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1297/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, Toulouse, France
| | - Lucas Bourdens
- Institut RESTORE, UMR 1301, Institut National de la Santé et de la Recherche Médicale (INSERM), CNRS-Université Paul Sabatier, Université de Toulouse, Toulouse, France
| | - Océane Delos
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1297/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, Toulouse, France
- MetaToul-MetaboHUB, Toulouse, France
| | - Lucie Combes-Soia
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Romain Paccoud
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1297/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, Toulouse, France
| | - Elsa Moreau
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1297/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, Toulouse, France
| | - Frédéric Martins
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1297/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, Toulouse, France
- Plateforme GeT, Genotoul, 31100 Toulouse, France
| | - Thomas Clouaire
- Molecular, Cellular, and Developmental Biology Unit (MCD), Centre de Biologie Intégrative (CBI), UPS, CNRS, Toulouse, France
| | - Fadila Benhamed
- Université de Paris, Institut Cochin, CNRS, INSERM, F- 75014 Paris, France
| | - Alexandra Montagner
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1297/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, Toulouse, France
| | - Walter Wahli
- Molecular, Cellular, and Developmental Biology Unit (MCD), Centre de Biologie Intégrative (CBI), UPS, CNRS, Toulouse, France
- Center for Integrative Genomics, University of Lausanne, Le Génopode, CH-1015 Lausanne, Switzerland
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Clinical Sciences Building, 11 Mandalay Road, Singapore 308232, Singapore
| | | | - Armelle Yart
- Institut RESTORE, UMR 1301, Institut National de la Santé et de la Recherche Médicale (INSERM), CNRS-Université Paul Sabatier, Université de Toulouse, Toulouse, France
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1297/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, Toulouse, France
| | - Isabelle Castan-Laurell
- Institut RESTORE, UMR 1301, Institut National de la Santé et de la Recherche Médicale (INSERM), CNRS-Université Paul Sabatier, Université de Toulouse, Toulouse, France
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1297/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, Toulouse, France
| | - Justine Bertrand-Michel
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1297/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, Toulouse, France
- MetaToul-MetaboHUB, Toulouse, France
| | - Odile Burlet-Schiltz
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Catherine Postic
- Université de Paris, Institut Cochin, CNRS, INSERM, F- 75014 Paris, France
| | - Pierre-Damien Denechaud
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1297/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, Toulouse, France
| | - Cédric Moro
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1297/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, Toulouse, France
| | - Gaelle Legube
- Molecular, Cellular, and Developmental Biology Unit (MCD), Centre de Biologie Intégrative (CBI), UPS, CNRS, Toulouse, France
| | - Chih-Hao Lee
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Hervé Guillou
- Toxalim, INRAE UMR 1331, ENVT, INP-Purpan, University of Toulouse, Paul Sabatier University, F-31027, Toulouse, France
| | - Philippe Valet
- Institut RESTORE, UMR 1301, Institut National de la Santé et de la Recherche Médicale (INSERM), CNRS-Université Paul Sabatier, Université de Toulouse, Toulouse, France
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1297/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, Toulouse, France
| | - Cédric Dray
- Institut RESTORE, UMR 1301, Institut National de la Santé et de la Recherche Médicale (INSERM), CNRS-Université Paul Sabatier, Université de Toulouse, Toulouse, France
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1297/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, Toulouse, France
| | - Jean-Philippe Pradère
- Institut RESTORE, UMR 1301, Institut National de la Santé et de la Recherche Médicale (INSERM), CNRS-Université Paul Sabatier, Université de Toulouse, Toulouse, France
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1297/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, Toulouse, France
| |
Collapse
|
8
|
Rives C, Fougerat A, Ellero-Simatos S, Loiseau N, Guillou H, Gamet-Payrastre L, Wahli W. Oxidative Stress in NAFLD: Role of Nutrients and Food Contaminants. Biomolecules 2020; 10:E1702. [PMID: 33371482 PMCID: PMC7767499 DOI: 10.3390/biom10121702] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 12/14/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is often the hepatic expression of metabolic syndrome and its comorbidities that comprise, among others, obesity and insulin-resistance. NAFLD involves a large spectrum of clinical conditions. These range from steatosis, a benign liver disorder characterized by the accumulation of fat in hepatocytes, to non-alcoholic steatohepatitis (NASH), which is characterized by inflammation, hepatocyte damage, and liver fibrosis. NASH can further progress to cirrhosis and hepatocellular carcinoma. The etiology of NAFLD involves both genetic and environmental factors, including an unhealthy lifestyle. Of note, unhealthy eating is clearly associated with NAFLD development and progression to NASH. Both macronutrients (sugars, lipids, proteins) and micronutrients (vitamins, phytoingredients, antioxidants) affect NAFLD pathogenesis. Furthermore, some evidence indicates disruption of metabolic homeostasis by food contaminants, some of which are risk factor candidates in NAFLD. At the molecular level, several models have been proposed for the pathogenesis of NAFLD. Most importantly, oxidative stress and mitochondrial damage have been reported to be causative in NAFLD initiation and progression. The aim of this review is to provide an overview of the contribution of nutrients and food contaminants, especially pesticides, to oxidative stress and how they may influence NAFLD pathogenesis.
Collapse
Affiliation(s)
- Clémence Rives
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRA, EVT, INP-Purpan, UPS, 31300 Toulouse, France; (C.R.); (A.F.); (S.E.-S.); (N.L.); (H.G.)
| | - Anne Fougerat
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRA, EVT, INP-Purpan, UPS, 31300 Toulouse, France; (C.R.); (A.F.); (S.E.-S.); (N.L.); (H.G.)
| | - Sandrine Ellero-Simatos
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRA, EVT, INP-Purpan, UPS, 31300 Toulouse, France; (C.R.); (A.F.); (S.E.-S.); (N.L.); (H.G.)
| | - Nicolas Loiseau
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRA, EVT, INP-Purpan, UPS, 31300 Toulouse, France; (C.R.); (A.F.); (S.E.-S.); (N.L.); (H.G.)
| | - Hervé Guillou
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRA, EVT, INP-Purpan, UPS, 31300 Toulouse, France; (C.R.); (A.F.); (S.E.-S.); (N.L.); (H.G.)
| | - Laurence Gamet-Payrastre
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRA, EVT, INP-Purpan, UPS, 31300 Toulouse, France; (C.R.); (A.F.); (S.E.-S.); (N.L.); (H.G.)
| | - Walter Wahli
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRA, EVT, INP-Purpan, UPS, 31300 Toulouse, France; (C.R.); (A.F.); (S.E.-S.); (N.L.); (H.G.)
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Clinical Sciences Building, 11 Mandalay Road, Singapore 308232, Singapore
- Center for Integrative Genomics, Université de Lausanne, Le Génopode, CH-1015 Lausanne, Switzerland
| |
Collapse
|
9
|
Fougerat A, Montagner A, Loiseau N, Guillou H, Wahli W. Peroxisome Proliferator-Activated Receptors and Their Novel Ligands as Candidates for the Treatment of Non-Alcoholic Fatty Liver Disease. Cells 2020; 9:E1638. [PMID: 32650421 PMCID: PMC7408116 DOI: 10.3390/cells9071638] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 06/26/2020] [Accepted: 07/04/2020] [Indexed: 12/11/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a major health issue worldwide, frequently associated with obesity and type 2 diabetes. Steatosis is the initial stage of the disease, which is characterized by lipid accumulation in hepatocytes, which can progress to non-alcoholic steatohepatitis (NASH) with inflammation and various levels of fibrosis that further increase the risk of developing cirrhosis and hepatocellular carcinoma. The pathogenesis of NAFLD is influenced by interactions between genetic and environmental factors and involves several biological processes in multiple organs. No effective therapy is currently available for the treatment of NAFLD. Peroxisome proliferator-activated receptors (PPARs) are nuclear receptors that regulate many functions that are disturbed in NAFLD, including glucose and lipid metabolism, as well as inflammation. Thus, they represent relevant clinical targets for NAFLD. In this review, we describe the determinants and mechanisms underlying the pathogenesis of NAFLD, its progression and complications, as well as the current therapeutic strategies that are employed. We also focus on the complementary and distinct roles of PPAR isotypes in many biological processes and on the effects of first-generation PPAR agonists. Finally, we review novel and safe PPAR agonists with improved efficacy and their potential use in the treatment of NAFLD.
Collapse
Affiliation(s)
- Anne Fougerat
- Institut National de la Recherche Agronomique (INRAE), ToxAlim, UMR1331 Toulouse, France; (A.M.); (N.L.); (H.G.)
| | - Alexandra Montagner
- Institut National de la Recherche Agronomique (INRAE), ToxAlim, UMR1331 Toulouse, France; (A.M.); (N.L.); (H.G.)
- Institut National de la Santé et de la Recherche Médicale (Inserm), Institute of Metabolic and Cardiovascular Diseases, UMR1048 Toulouse, France
- Institute of Metabolic and Cardiovascular Diseases, University of Toulouse, UMR1048 Toulouse, France
| | - Nicolas Loiseau
- Institut National de la Recherche Agronomique (INRAE), ToxAlim, UMR1331 Toulouse, France; (A.M.); (N.L.); (H.G.)
| | - Hervé Guillou
- Institut National de la Recherche Agronomique (INRAE), ToxAlim, UMR1331 Toulouse, France; (A.M.); (N.L.); (H.G.)
| | - Walter Wahli
- Institut National de la Recherche Agronomique (INRAE), ToxAlim, UMR1331 Toulouse, France; (A.M.); (N.L.); (H.G.)
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Clinical Sciences Building, 11 Mandalay Road, Singapore 308232, Singapore
- Center for Integrative Genomics, Université de Lausanne, Le Génopode, CH-1015 Lausanne, Switzerland
| |
Collapse
|
10
|
Hepatobiliary Involvement in Cystic Fibrosis. Respir Med 2020. [DOI: 10.1007/978-3-030-42382-7_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
11
|
Amiel A, Tremblay-Franco M, Gautier R, Ducheix S, Montagner A, Polizzi A, Debrauwer L, Guillou H, Bertrand-Michel J, Canlet C. Proton NMR Enables the Absolute Quantification of Aqueous Metabolites and Lipid Classes in Unique Mouse Liver Samples. Metabolites 2019; 10:metabo10010009. [PMID: 31877749 PMCID: PMC7023327 DOI: 10.3390/metabo10010009] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 12/16/2019] [Accepted: 12/19/2019] [Indexed: 02/06/2023] Open
Abstract
Hepatic metabolites provide valuable information on the physiological state of an organism, and thus, they are monitored in many clinical situations. Typically, monitoring requires several analyses for each class of targeted metabolite, which is time consuming. The present study aimed to evaluate a proton nuclear magnetic resonance (1H-NMR) method for obtaining quantitative measurements of aqueous and lipidic metabolites. We optimized the extraction protocol, the standard samples, and the organic solvents for the absolute quantification of lipid species. To validate the method, we analyzed metabolic profiles in livers of mice fed three different diets. We compared our results with values obtained with conventional methods and found strong correlations. The 1H-NMR protocol enabled the absolute quantification of 29 aqueous metabolites and eight lipid classes. Results showed that mice fed a diet enriched in saturated fatty acids had higher levels of triglycerides, cholesterol ester, monounsaturated fatty acids, lactate, 3-hydroxy-butyrate, and alanine and lower levels of glucose, compared to mice fed a control diet. In conclusion, proton NMR provided a rapid overview of the main lipid classes (triglycerides, cholesterol, phospholipids, fatty acids) and the most abundant aqueous metabolites in liver.
Collapse
Affiliation(s)
- Aurélien Amiel
- Toxalim-Research Centre in Food Toxicology, Toulouse University, INRAE UMR 1331, ENVT, INP-Purpan, Paul Sabatier University, F-31027 Toulouse, France; (A.A.); (M.T.-F.); (R.G.); (S.D.); (A.M.); (A.P.); (L.D.); (H.G.)
- Metatoul-AXIOM platform, National Infrastructure for Metabolomics and Fluxomics, MetaboHUB, Toxalim, INRAE UMR 1331, F-31027 Toulouse, France
| | - Marie Tremblay-Franco
- Toxalim-Research Centre in Food Toxicology, Toulouse University, INRAE UMR 1331, ENVT, INP-Purpan, Paul Sabatier University, F-31027 Toulouse, France; (A.A.); (M.T.-F.); (R.G.); (S.D.); (A.M.); (A.P.); (L.D.); (H.G.)
- Metatoul-AXIOM platform, National Infrastructure for Metabolomics and Fluxomics, MetaboHUB, Toxalim, INRAE UMR 1331, F-31027 Toulouse, France
| | - Roselyne Gautier
- Toxalim-Research Centre in Food Toxicology, Toulouse University, INRAE UMR 1331, ENVT, INP-Purpan, Paul Sabatier University, F-31027 Toulouse, France; (A.A.); (M.T.-F.); (R.G.); (S.D.); (A.M.); (A.P.); (L.D.); (H.G.)
- Metatoul-AXIOM platform, National Infrastructure for Metabolomics and Fluxomics, MetaboHUB, Toxalim, INRAE UMR 1331, F-31027 Toulouse, France
| | - Simon Ducheix
- Toxalim-Research Centre in Food Toxicology, Toulouse University, INRAE UMR 1331, ENVT, INP-Purpan, Paul Sabatier University, F-31027 Toulouse, France; (A.A.); (M.T.-F.); (R.G.); (S.D.); (A.M.); (A.P.); (L.D.); (H.G.)
| | - Alexandra Montagner
- Toxalim-Research Centre in Food Toxicology, Toulouse University, INRAE UMR 1331, ENVT, INP-Purpan, Paul Sabatier University, F-31027 Toulouse, France; (A.A.); (M.T.-F.); (R.G.); (S.D.); (A.M.); (A.P.); (L.D.); (H.G.)
| | - Arnaud Polizzi
- Toxalim-Research Centre in Food Toxicology, Toulouse University, INRAE UMR 1331, ENVT, INP-Purpan, Paul Sabatier University, F-31027 Toulouse, France; (A.A.); (M.T.-F.); (R.G.); (S.D.); (A.M.); (A.P.); (L.D.); (H.G.)
| | - Laurent Debrauwer
- Toxalim-Research Centre in Food Toxicology, Toulouse University, INRAE UMR 1331, ENVT, INP-Purpan, Paul Sabatier University, F-31027 Toulouse, France; (A.A.); (M.T.-F.); (R.G.); (S.D.); (A.M.); (A.P.); (L.D.); (H.G.)
- Metatoul-AXIOM platform, National Infrastructure for Metabolomics and Fluxomics, MetaboHUB, Toxalim, INRAE UMR 1331, F-31027 Toulouse, France
| | - Hervé Guillou
- Toxalim-Research Centre in Food Toxicology, Toulouse University, INRAE UMR 1331, ENVT, INP-Purpan, Paul Sabatier University, F-31027 Toulouse, France; (A.A.); (M.T.-F.); (R.G.); (S.D.); (A.M.); (A.P.); (L.D.); (H.G.)
| | | | - Cécile Canlet
- Toxalim-Research Centre in Food Toxicology, Toulouse University, INRAE UMR 1331, ENVT, INP-Purpan, Paul Sabatier University, F-31027 Toulouse, France; (A.A.); (M.T.-F.); (R.G.); (S.D.); (A.M.); (A.P.); (L.D.); (H.G.)
- Metatoul-AXIOM platform, National Infrastructure for Metabolomics and Fluxomics, MetaboHUB, Toxalim, INRAE UMR 1331, F-31027 Toulouse, France
- Correspondence:
| |
Collapse
|
12
|
Marchix J, Catheline D, Duby C, Monthéan-Boulier N, Boissel F, Pédrono F, Boudry G, Legrand P. Interactive effects of maternal and weaning high linoleic acid intake on hepatic lipid metabolism, oxylipins profile and hepatic steatosis in offspring. J Nutr Biochem 2019; 75:108241. [PMID: 31715523 DOI: 10.1016/j.jnutbio.2019.108241] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 08/12/2019] [Accepted: 09/09/2019] [Indexed: 02/07/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) has been described as a hepatic manifestation of the metabolic syndrome. When several studies correlated maternal linoleic acid (LA) intake with the development of obesity, only few links have been made between n-6 fatty acid (FA) and NAFLD. Herein, we investigated the influence of both maternal and weaning high LA intake on lipid metabolism and susceptibility to develop later metabolic diseases in offspring. Pregnant rats were fed a control-diet (2% LA) or a LA-rich diet (12% LA) during gestation and lactation. At weaning, offspring was assigned to one of the two diets, i.e., either maintained on the same maternal diet or fed the other diet for 6 months. Physiological, biochemical parameters and hepatic FA metabolism were analyzed. We demonstrated that the interaction between the maternal and weaning LA intake altered metabolism in offspring and could lead to hepatic steatosis. This phenotype was associated with altered hepatic FA content and lipid metabolism. Interaction between maternal and weaning LA intake led to a specific pattern of n-6 and n-3 oxylipins that could participate to the development of hepatic steatosis in offspring. Our findings highlight the significant interaction between maternal and weaning high LA intake to predispose offspring to later metabolic disease and support the predictive adaptive response hypothesis.
Collapse
Affiliation(s)
- Justine Marchix
- Laboratoire de Biochimie et Nutrition Humaine, INRA USC 1378, Agrocampus Ouest, Rennes, France.
| | - Daniel Catheline
- Laboratoire de Biochimie et Nutrition Humaine, INRA USC 1378, Agrocampus Ouest, Rennes, France.
| | - Cécile Duby
- Laboratoire de Biochimie et Nutrition Humaine, INRA USC 1378, Agrocampus Ouest, Rennes, France.
| | | | - Francoise Boissel
- Laboratoire de Biochimie et Nutrition Humaine, INRA USC 1378, Agrocampus Ouest, Rennes, France.
| | - Frédérique Pédrono
- Laboratoire de Biochimie et Nutrition Humaine, INRA USC 1378, Agrocampus Ouest, Rennes, France.
| | - Gaëlle Boudry
- Institut NuMeCan INRA, INSERM, Univ Rennes, Rennes, France.
| | - Philippe Legrand
- Laboratoire de Biochimie et Nutrition Humaine, INRA USC 1378, Agrocampus Ouest, Rennes, France.
| |
Collapse
|
13
|
Nikolaou N, Gathercole LL, Marchand L, Althari S, Dempster NJ, Green CJ, van de Bunt M, McNeil C, Arvaniti A, Hughes BA, Sgromo B, Gillies RS, Marschall HU, Penning TM, Ryan J, Arlt W, Hodson L, Tomlinson JW. AKR1D1 is a novel regulator of metabolic phenotype in human hepatocytes and is dysregulated in non-alcoholic fatty liver disease. Metabolism 2019; 99:67-80. [PMID: 31330134 PMCID: PMC6744372 DOI: 10.1016/j.metabol.2019.153947] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 06/28/2019] [Accepted: 07/18/2019] [Indexed: 01/07/2023]
Abstract
OBJECTIVE Non-alcoholic fatty liver disease (NAFLD) is the hepatic manifestation of metabolic syndrome. Steroid hormones and bile acids are potent regulators of hepatic carbohydrate and lipid metabolism. Steroid 5β-reductase (AKR1D1) is highly expressed in human liver where it inactivates steroid hormones and catalyzes a fundamental step in bile acid synthesis. METHODS Human liver biopsies were obtained from 34 obese patients and AKR1D1 mRNA expression levels were measured using qPCR. Genetic manipulation of AKR1D1 was performed in human HepG2 and Huh7 liver cell lines. Metabolic assessments were made using transcriptome analysis, western blotting, mass spectrometry, clinical biochemistry, and enzyme immunoassays. RESULTS In human liver biopsies, AKR1D1 expression decreased with advancing steatosis, fibrosis and inflammation. Expression was decreased in patients with type 2 diabetes. In human liver cell lines, AKR1D1 knockdown decreased primary bile acid biosynthesis and steroid hormone clearance. RNA-sequencing identified disruption of key metabolic pathways, including insulin action and fatty acid metabolism. AKR1D1 knockdown increased hepatocyte triglyceride accumulation, insulin sensitivity, and glycogen synthesis, through increased de novo lipogenesis and decreased β-oxidation, fueling hepatocyte inflammation. Pharmacological manipulation of bile acid receptor activation prevented the induction of lipogenic and carbohydrate genes, suggesting that the observed metabolic phenotype is driven through bile acid rather than steroid hormone availability. CONCLUSIONS Genetic manipulation of AKR1D1 regulates the metabolic phenotype of human hepatoma cell lines, driving steatosis and inflammation. Taken together, the observation that AKR1D1 mRNA is down-regulated with advancing NAFLD suggests that it may have a crucial role in the pathogenesis and progression of the disease.
Collapse
Affiliation(s)
- Nikolaos Nikolaou
- Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford OX3 7LE, UK
| | - Laura L Gathercole
- Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford OX3 7LE, UK; Department of Biological and Medical Sciences, Oxford Brookes University, Oxford OX3 0BP, UK
| | - Lea Marchand
- Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford OX3 7LE, UK
| | - Sara Althari
- Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford OX3 7LE, UK
| | - Niall J Dempster
- Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford OX3 7LE, UK
| | - Charlotte J Green
- Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford OX3 7LE, UK
| | - Martijn van de Bunt
- Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford OX3 7LE, UK
| | - Catriona McNeil
- Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford OX3 7LE, UK
| | - Anastasia Arvaniti
- Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford OX3 7LE, UK; Department of Biological and Medical Sciences, Oxford Brookes University, Oxford OX3 0BP, UK
| | - Beverly A Hughes
- Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Bruno Sgromo
- Department of Upper GI Surgery, Churchill Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Richard S Gillies
- Department of Upper GI Surgery, Churchill Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Hanns-Ulrich Marschall
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, 413 45 Gothenburg, Sweden
| | - Trevor M Penning
- Department of Systems Pharmacology & Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, 1315 BRB II/III 421 Curie Blvd, Philadelphia, PA 19104-6160, United States of America
| | - John Ryan
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK
| | - Wiebke Arlt
- Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Leanne Hodson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford OX3 7LE, UK
| | - Jeremy W Tomlinson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, NIHR Oxford Biomedical Research Centre, University of Oxford, Churchill Hospital, Oxford OX3 7LE, UK.
| |
Collapse
|
14
|
Han X, Cui ZY, Song J, Piao HQ, Lian LH, Hou LS, Wang G, Zheng S, Dong XX, Nan JX, Wu YL. Acanthoic acid modulates lipogenesis in nonalcoholic fatty liver disease via FXR/LXRs-dependent manner. Chem Biol Interact 2019; 311:108794. [DOI: 10.1016/j.cbi.2019.108794] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 07/24/2019] [Accepted: 08/13/2019] [Indexed: 02/07/2023]
|
15
|
Immunomodulatory Effects of Combination Therapy with Bushen Formula plus Entecavir for Chronic Hepatitis B Patients. J Immunol Res 2019; 2019:8983903. [PMID: 30766891 PMCID: PMC6350602 DOI: 10.1155/2019/8983903] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 10/03/2018] [Indexed: 02/07/2023] Open
Abstract
Aim To compare the clinical efficacy of the combination therapy with Bushen formula (BSF) plus entecavir (ETV) in naïve chronic hepatitis B (CHB) patients and that in CHB patients with partial virological response to ETV and explore the relevant immunoregulatory mechanism. Materials and Methods Two hundred and twenty CHB patients were enrolled in the historical prospective cohort study. Patients were categorized into a treatment group (T-Group: combination therapy with BSF plus ETV) and a control group (C-Group: ETV). Patients in T-Group and C-Group were grouped into T1/C1 (treatment-naïve patients) and T2/C2 (patients with partial virological response to ETV). Biochemical assessment, viral load quantitation, and HBV markers were tested. Chinese medicine symptom complex score was evaluated and recorded as well. In addition, peripheral blood mononuclear cells were separated from blood samples in 56 patients and 11 healthy donors. The frequencies of Th1, Treg, and dendritic cells (DCs) and expression levels of PD-1/PD-L1 were examined by flow cytometry. Results In treatment-naïve CHB patients, complete viral suppression rates in HBeAg(−) patients were higher than those in HBeAg(+) patients in both T and C groups. In patients with partial virological response to ETV, the rate of HBsAg decline ≥ 20% in HBeAg(+) patients of T2-Group was higher than that in HBeAg(+) patients of C2-Group. A significant reduction of Chinese medicine symptom complex score was only observed in T-Group. The study of mechanism showed that, compared with healthy controls, Th1 and DC frequencies were decreased in all CHB patients, while Treg frequency was increased only in treatment-naïve patients. In addition, compared with healthy controls, PD-1 expression levels on Th1 and Treg were increased in all patients and PD-L1 expression levels on DCs were increased only in treatment-naïve patients. In treatment-naïve patients, the combination therapy with BSF plus ETV increased Th1 and DC frequencies and decreased Treg frequency, which was correlated with HBsAg decline. In addition, in patients with partial virological response to ETV, the combination therapy downregulated PD-L1 levels on DCs and the frequency of Treg, which was related with HBsAg decline. Conclusions In patients with partial virological response to ETV, HBeAg(+) patients tend to achieve ideal effects after the combination therapy with BSF plus ETV, which may correlate with the decrease of Treg frequency and the downregulation of PD-L1 levels on DCs.
Collapse
|
16
|
Itsiopoulos C, Marx W, Mayr H, Tatucu-Babet O, Dash S, George E, Trakman G, Kelly J, Thomas C, Brazionis L. The role of omega-3 polyunsaturated fatty acid supplementation in the management of type 2 diabetes mellitus: A narrative review. JOURNAL OF NUTRITION & INTERMEDIARY METABOLISM 2018. [DOI: 10.1016/j.jnim.2018.02.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
17
|
Hepatoprotective and Antioxidant Activities of Oil from Baru Almonds ( Dipteryx alata Vog.) in a Preclinical Model of Lipotoxicity and Dyslipidemia. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:8376081. [PMID: 30369957 PMCID: PMC6189660 DOI: 10.1155/2018/8376081] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Revised: 09/11/2018] [Accepted: 09/19/2018] [Indexed: 12/15/2022]
Abstract
The oil obtained from baru (Dipteryx alata Vog.) almonds exhibits high energy value and is reported in popular medicine for the treatment of rheumatic diseases and reproductive disturbances. Although baru oil is used in domestic cuisine, the chemical characterization of this oil and its effects on lipid metabolism are still poorly understood. Therefore, this study evaluated the fatty acid (FA) profile and the effects of baru oil on liver and aorta in a murine model of dyslipidemia. The chromatographic profile of baru oil showed high levels of unsaturated FAs, especially oleic acid. Saturated FAs, such as palmitic and lignoceric acids, were found in lower amounts. Hypercholesterolemia was induced in male Wistar rats by daily administration of a lipid emulsion by gavage for 15 weeks. Biochemical and histopathological analysis were performed on serum, aorta, and liver. The results demonstrated that animals developed marked hypercholesterolemia, liver steatosis, and increased lipid peroxidation in the aorta. Treatment with baru oil attenuated lipid peroxidation and drastically reduced liver damage, especially ballooning degeneration and steatosis. By restricting vascular and hepatic injury, this oil showed potential applicability as a functional food, reinforcing its use in popular medicine and domestic cuisine.
Collapse
|
18
|
Dual extraction of mRNA and lipids from a single biological sample. Sci Rep 2018; 8:7019. [PMID: 29728575 PMCID: PMC5935724 DOI: 10.1038/s41598-018-25332-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 04/19/2018] [Indexed: 12/11/2022] Open
Abstract
The extraction of RNA and lipids from a large number of biological samples is time-consuming and costly with steps required for both transcriptomic and lipidomic approaches. Most protocols rely on independent extraction of nucleic acids and lipids from a single sample, thereby increasing the need for biological material and inducing variability in data analysis. We investigated whether it is possible to use a standard RNA extraction procedure to analyze not only RNA levels, but also lipids in a single liver sample. We show that the organic phase obtained when using standard reagents for RNA extraction can be used to analyze lipids, including neutral lipids and fatty acids, by gas chromatography. We applied this technique to an analysis of lipids and the associated gene expression pattern in mice with hepatic steatosis induced by pharmacological activation of nuclear receptor LXR.
Collapse
|
19
|
Multi-tissue transcriptomic study reveals the main role of liver in the chicken adaptive response to a switch in dietary energy source through the transcriptional regulation of lipogenesis. BMC Genomics 2018. [PMID: 29514634 PMCID: PMC5842524 DOI: 10.1186/s12864-018-4520-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Background Because the cost of cereals is unstable and represents a large part of production charges for meat-type chicken, there is an urge to formulate alternative diets from more cost-effective feedstuff. We have recently shown that meat-type chicken source is prone to adapt to dietary starch substitution with fat and fiber. The aim of this study was to better understand the molecular mechanisms of this adaptation to changes in dietary energy sources through the fine characterization of transcriptomic changes occurring in three major metabolic tissues – liver, adipose tissue and muscle – as well as in circulating blood cells. Results We revealed the fine-tuned regulation of many hepatic genes encoding key enzymes driving glycogenesis and de novo fatty acid synthesis pathways and of some genes participating in oxidation. Among the genes expressed upon consumption of a high-fat, high-fiber diet, we highlighted CPT1A, which encodes a key enzyme in the regulation of fatty acid oxidation. Conversely, the repression of lipogenic genes by the high-fat diet was clearly associated with the down-regulation of SREBF1 transcripts but was not associated with the transcript regulation of MLXIPL and NR1H3, which are both transcription factors. This result suggests a pivotal role for SREBF1 in lipogenesis regulation in response to a decrease in dietary starch and an increase in dietary PUFA. Other prospective regulators of de novo hepatic lipogenesis were suggested, such as PPARD, JUN, TADA2A and KAT2B, the last two genes belonging to the lysine acetyl transferase (KAT) complex family regulating histone and non-histone protein acetylation. Hepatic glycogenic genes were also down-regulated in chickens fed a high-fat, high-fiber diet compared to those in chickens fed a starch-based diet. No significant dietary-associated variations in gene expression profiles was observed in the other studied tissues, suggesting that the liver mainly contributed to the adaptation of birds to changes in energy source and nutrients in their diets, at least at the transcriptional level. Moreover, we showed that PUFA deposition observed in the different tissues may not rely on transcriptional changes. Conclusion We showed the major role of the liver, at the gene expression level, in the adaptive response of chicken to dietary starch substitution with fat and fiber. Electronic supplementary material The online version of this article (10.1186/s12864-018-4520-5) contains supplementary material, which is available to authorized users.
Collapse
|
20
|
Pachikian BD, Druart C, Catry E, Bindels LB, Neyrinck AM, Larondelle Y, Cani PD, Delzenne NM. Implication of trans-11,trans-13 conjugated linoleic acid in the development of hepatic steatosis. PLoS One 2018; 13:e0192447. [PMID: 29389988 PMCID: PMC5794163 DOI: 10.1371/journal.pone.0192447] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 01/23/2018] [Indexed: 12/15/2022] Open
Abstract
SCOPE Conjugated linoleic acids are linoleic acid isomers found in the diet that can also be produced through bacterial metabolism of polyunsaturated fatty acids. Our objective was to evaluate the contribution of fatty acid metabolites produced from polyunsaturated fatty acids by the gut microbiota in vivo to regulation of hepatic lipid metabolism and steatosis. METHODS AND RESULTS In mice with depleted n-3 polyunsaturated fatty acids, we observed an accumulation of trans-11,trans-13 CLA and cis-9,cis-11 conjugated linoleic acids in the liver tissue that were associated with an increased triglyceride content and expression of lipogenic genes. We used an in vitro model to evaluate the impact of these two conjugated linoleic acids on hepatic lipid metabolism. In HepG2 cells, we observed that only trans-11,trans-13 conjugated linoleic acids recapitulated triglyceride accumulation and increased lipogenic gene expression, which is a phenomenon that may implicate the nuclear factors sterol regulatory element binding protein 1c (SREBP-1c) and carbohydrate-responsive element-binding protein (ChREBP). CONCLUSION The trans-11,trans-13 conjugated linoleic acids can stimulate hepatic lipogenesis, which supports the conclusion that gut microbiota and related metabolites should be considered in the treatment of non-alcoholic liver disease.
Collapse
Affiliation(s)
- Barbara D. Pachikian
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Céline Druart
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Emilie Catry
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Laure B. Bindels
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Audrey M. Neyrinck
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Yvan Larondelle
- Institut des Sciences de la Vie, Université catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Patrice D. Cani
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
- Université catholique de Louvain, WELBIO, WELBIO (Walloon Excellence in Life sciences and BIOtechnology), Louvain Drug Research Institute, Metabolism and Nutrition Research Group, Brussels, Belgium
| | - Nathalie M. Delzenne
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
- * E-mail:
| |
Collapse
|
21
|
Li Y, Zhao F, Wu Q, Li M, Zhu Y, Song S, Zhu J, Ma Y, Li H, Shi X, Xu X, Zhou G, Li C. Fish oil diet may reduce inflammatory levels in the liver of middle-aged rats. Sci Rep 2017; 7:6241. [PMID: 28740245 PMCID: PMC5524965 DOI: 10.1038/s41598-017-06506-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 06/12/2017] [Indexed: 01/23/2023] Open
Abstract
The impact of dietary soybean oil, lard and fish oil on physiological responses in middle age is little studied. In this study, we investigated the changes of oxidative stress, inflammatory cytokines, telomere length, and age-related gene expression in the liver of middle-aged rats in response to the above three fat diets. Male Sprague Dawley rats (12 months old) were fed AIN-93M diets for 3 months, in which soybean oil was equivalently replaced by lard or fish oil. As compared to the lard diet, intake of fish oil diet significantly decreased body weight gain, white blood cell count, and levels of hepatic triacylglycerol, total cholesterol, fat accumulation, low-density lipoprotein, oxidative stress and inflammatory cytokines (P < 0.05), but increased telomere length (P < 0.05). On the other hand, lard diet and soybean oil diet showed great similarity in the above variables. PCR array analysis further indicated that fish oil diet significantly down-regulated gene expression related to inflammatory response, apoptosis, DNA binding, proteostasis and telomere attrition. Differentially expressed genes were enriched in the complement and coagulation cascades pathways. Such physiological and molecular responses could be due to different fatty acid composition in fish oil, lard and soybean oil.
Collapse
Affiliation(s)
- Yingqiu Li
- Key Laboratory of Meat Processing and Quality Control, MOE, Key Laboratory of Meat Processing, MOA, Jiangsu Innovation Center of Meat Production and Processing, Nanjing Agricultural University, Nanjing, 210095, P.R. China
- Guangxi Vocational College of Technology and Business, Nanning, 530008, Guangxi, P.R. China
| | - Fan Zhao
- Key Laboratory of Meat Processing and Quality Control, MOE, Key Laboratory of Meat Processing, MOA, Jiangsu Innovation Center of Meat Production and Processing, Nanjing Agricultural University, Nanjing, 210095, P.R. China
| | - Qiayu Wu
- Key Laboratory of Meat Processing and Quality Control, MOE, Key Laboratory of Meat Processing, MOA, Jiangsu Innovation Center of Meat Production and Processing, Nanjing Agricultural University, Nanjing, 210095, P.R. China
| | - Mengjie Li
- Key Laboratory of Meat Processing and Quality Control, MOE, Key Laboratory of Meat Processing, MOA, Jiangsu Innovation Center of Meat Production and Processing, Nanjing Agricultural University, Nanjing, 210095, P.R. China
| | - Yingying Zhu
- Key Laboratory of Meat Processing and Quality Control, MOE, Key Laboratory of Meat Processing, MOA, Jiangsu Innovation Center of Meat Production and Processing, Nanjing Agricultural University, Nanjing, 210095, P.R. China
| | - Shangxin Song
- Key Laboratory of Meat Processing and Quality Control, MOE, Key Laboratory of Meat Processing, MOA, Jiangsu Innovation Center of Meat Production and Processing, Nanjing Agricultural University, Nanjing, 210095, P.R. China
- School of Food Science, Nanjing Xiaozhuang University, Nanjing, 211171, P.R. China
| | - Jing Zhu
- Key Laboratory of Meat Processing and Quality Control, MOE, Key Laboratory of Meat Processing, MOA, Jiangsu Innovation Center of Meat Production and Processing, Nanjing Agricultural University, Nanjing, 210095, P.R. China
| | - Yafang Ma
- Key Laboratory of Meat Processing and Quality Control, MOE, Key Laboratory of Meat Processing, MOA, Jiangsu Innovation Center of Meat Production and Processing, Nanjing Agricultural University, Nanjing, 210095, P.R. China
| | - He Li
- Key Laboratory of Meat Processing and Quality Control, MOE, Key Laboratory of Meat Processing, MOA, Jiangsu Innovation Center of Meat Production and Processing, Nanjing Agricultural University, Nanjing, 210095, P.R. China
| | - Xuebin Shi
- Key Laboratory of Meat Processing and Quality Control, MOE, Key Laboratory of Meat Processing, MOA, Jiangsu Innovation Center of Meat Production and Processing, Nanjing Agricultural University, Nanjing, 210095, P.R. China
| | - Xinglian Xu
- Key Laboratory of Meat Processing and Quality Control, MOE, Key Laboratory of Meat Processing, MOA, Jiangsu Innovation Center of Meat Production and Processing, Nanjing Agricultural University, Nanjing, 210095, P.R. China
| | - Guanghong Zhou
- Key Laboratory of Meat Processing and Quality Control, MOE, Key Laboratory of Meat Processing, MOA, Jiangsu Innovation Center of Meat Production and Processing, Nanjing Agricultural University, Nanjing, 210095, P.R. China.
| | - Chunbao Li
- Key Laboratory of Meat Processing and Quality Control, MOE, Key Laboratory of Meat Processing, MOA, Jiangsu Innovation Center of Meat Production and Processing, Nanjing Agricultural University, Nanjing, 210095, P.R. China.
| |
Collapse
|
22
|
Ducheix S, Montagner A, Polizzi A, Lasserre F, Régnier M, Marmugi A, Benhamed F, Bertrand-Michel J, Mselli-Lakhal L, Loiseau N, Martin PG, Lobaccaro JM, Ferrier L, Postic C, Guillou H. Dietary oleic acid regulates hepatic lipogenesis through a liver X receptor-dependent signaling. PLoS One 2017; 12:e0181393. [PMID: 28732092 PMCID: PMC5521785 DOI: 10.1371/journal.pone.0181393] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 06/22/2017] [Indexed: 12/13/2022] Open
Abstract
Olive oil consumption is beneficial for health as it is associated with a decreased prevalence of cancer and cardiovascular diseases. Oleic acid is, by far, the most abundant component of olive oil. Since it can be made through de novo synthesis in animals, it is not an essential fatty acid. While it has become clear that dietary oleic acid regulates many biological processes, the signaling pathway involved in these regulations remains poorly defined. In this work we tested the impact of an oleic acid-rich diet on hepatic gene expression. We were particularly interested in addressing the contribution of Liver X Receptors (LXR) in the control of genes involved in hepatic lipogenesis, an essential process in whole body energy homeostasis. We used wild-type mice and transgenic mice deficient for both α and β Liver X Receptor isoforms (LXR-/-) fed a control or an oleate enriched diet. We observed that hepatic-lipid accumulation was enhanced as well as the expression of lipogenic genes in the liver of wild-type mice fed the oleate enriched diet. In contrast, none of these changes occurred in the liver of LXR-/- mice. Strikingly, oleate-rich diet reduced cholesterolemia in wild-type mice and induced signs of liver inflammation and damage in LXR-/- mice but not in wild-type mice. This work suggests that dietary oleic acid reduces cholesterolemia while promoting LXR-dependent hepatic lipogenesis without detrimental effects to the liver.
Collapse
Affiliation(s)
- Simon Ducheix
- INRA, ToxAlim, Toulouse, France.,Université de Toulouse, INP, UPS, ToxAlim, Toulouse, France
| | - Alexandra Montagner
- INRA, ToxAlim, Toulouse, France.,Université de Toulouse, INP, UPS, ToxAlim, Toulouse, France
| | - Arnaud Polizzi
- INRA, ToxAlim, Toulouse, France.,Université de Toulouse, INP, UPS, ToxAlim, Toulouse, France
| | - Frédéric Lasserre
- INRA, ToxAlim, Toulouse, France.,Université de Toulouse, INP, UPS, ToxAlim, Toulouse, France
| | - Marion Régnier
- INRA, ToxAlim, Toulouse, France.,Université de Toulouse, INP, UPS, ToxAlim, Toulouse, France
| | - Alice Marmugi
- INRA, ToxAlim, Toulouse, France.,Université de Toulouse, INP, UPS, ToxAlim, Toulouse, France
| | - Fadila Benhamed
- INSERM, U1016, Institut Cochin, Paris, France.,CNRS, UMR8104, Paris, France
| | | | - Laila Mselli-Lakhal
- INRA, ToxAlim, Toulouse, France.,Université de Toulouse, INP, UPS, ToxAlim, Toulouse, France
| | - Nicolas Loiseau
- INRA, ToxAlim, Toulouse, France.,Université de Toulouse, INP, UPS, ToxAlim, Toulouse, France
| | - Pascal G Martin
- INRA, ToxAlim, Toulouse, France.,Université de Toulouse, INP, UPS, ToxAlim, Toulouse, France
| | - Jean-Marc Lobaccaro
- Clermont Université, Université Blaise Pascal, Génétique Reproduction et Développement, Clermont-Ferrand, France.,CNRS, UMR 6293, GReD, Aubière, France.,INSERM, U1103, GReD, Aubière, France.,Centre de Recherche en Nutrition Humaine d'Auvergne, Clermont-Ferrand, France
| | - Laurent Ferrier
- INRA, ToxAlim, Toulouse, France.,Université de Toulouse, INP, UPS, ToxAlim, Toulouse, France
| | - Catherine Postic
- INSERM, U1016, Institut Cochin, Paris, France.,CNRS, UMR8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Hervé Guillou
- INRA, ToxAlim, Toulouse, France.,Université de Toulouse, INP, UPS, ToxAlim, Toulouse, France
| |
Collapse
|
23
|
Zhang X, Heckmann BL, Campbell LE, Liu J. G0S2: A small giant controller of lipolysis and adipose-liver fatty acid flux. Biochim Biophys Acta Mol Cell Biol Lipids 2017. [PMID: 28645852 DOI: 10.1016/j.bbalip.2017.06.007] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The discovery of adipose triglyceride lipase (ATGL) and its coactivator comparative gene identification-58 (CGI-58) provided a major paradigm shift in the understanding of intracellular lipolysis in both adipocytes and nonadipocyte cells. The subsequent discovery of G0/G1 switch gene 2 (G0S2) as a potent endogenous inhibitor of ATGL revealed a unique mechanism governing lipolysis and fatty acid (FA) availability. G0S2 is highly conserved in vertebrates, and exhibits cyclical expression pattern between adipose tissue and liver that is critical to lipid flux and energy homeostasis in these two tissues. Biochemical and cell biological studies have demonstrated that a direct interaction with ATGL mediates G0S2's inhibitory effects on lipolysis and lipid droplet degradation. In this review we examine evidence obtained from recent in vitro and in vivo studies that lends support to the proof-of-principle concept that G0S2 functions as a master regulator of tissue-specific balance of TG storage vs. mobilization, partitioning of metabolic fuels between adipose and liver, and the whole-body adaptive energy response. This article is part of a Special Issue entitled: Recent Advances in Lipid Droplet Biology edited by Rosalind Coleman and Matthijs Hesselink.
Collapse
Affiliation(s)
- Xiaodong Zhang
- Department of Biochemistry & Molecular Biology, Mayo Clinic College of Medicine, Scottsdale, AZ, United States; HEAL(th) Program, Mayo Clinic, Scottsdale, AZ, United States
| | - Bradlee L Heckmann
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Latoya E Campbell
- School of Life Sciences, Arizona State University, Tempe, AZ, United States
| | - Jun Liu
- Department of Biochemistry & Molecular Biology, Mayo Clinic College of Medicine, Scottsdale, AZ, United States; HEAL(th) Program, Mayo Clinic, Scottsdale, AZ, United States; Division of Endocrinology, Mayo Clinic, Scottsdale, AZ, United States.
| |
Collapse
|
24
|
Prevete N, Liotti F, Illiano A, Amoresano A, Pucci P, de Paulis A, Melillo RM. Formyl peptide receptor 1 suppresses gastric cancer angiogenesis and growth by exploiting inflammation resolution pathways. Oncoimmunology 2017; 6:e1293213. [PMID: 28507800 DOI: 10.1080/2162402x.2017.1293213] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Revised: 02/03/2017] [Accepted: 02/04/2017] [Indexed: 12/14/2022] Open
Abstract
Chronic inflammation can result from inadequate engagement of resolution mechanisms, mainly accomplished by specialized pro-resolving mediators (SPMs) arising from the metabolic activity of lipoxygenases (ALOX5/15) on ω-6 or ω-3 essential polyunsaturated fatty acids (PUFA). We previously demonstrated that formyl peptide receptor 1 (FPR1) suppresses gastric cancer (GC) by inhibiting its inflammatory/angiogenic potential. In this study, we asked whether FPR1 exploits inflammation resolution pathways to suppress GC angiogenesis and growth. Here, we demonstrate that genetic or pharmacologic modulation of FPR1 in GC cells regulated ALOX5/15 expression and production of the SPMs Resolvin D1 (RvD1) and Lipoxin B4 (LXB4). SPM treatment of GC cells abated their angiogenic potential. Genetic deletion of ALOX15 or of the RvD1 receptor GPR32 increased the angiogenic and tumorigenic activity of GC cells thereby mimicking FPR1 loss. Deletion/inhibition of ALOX5/15 or GPR32 blocked FPR1-mediated anti-angiogenic activities, indicating that ALOX5/15 and GPR32 are required for FPR1's pro-resolving action. An ω-3- or ω-6-enriched diet enforced SPM endogenous production in mice and inhibited growth of shFPR1 GC xenografts by suppressing their angiogenic activity. These data implicate that FPR1 and/or pro-resolving pathway components might be used as risk/prognostic markers for GC; ω-6/3-enriched diets, and targeting FPR1 or SPM machinery may be exploited for GC management.
Collapse
Affiliation(s)
- Nella Prevete
- Dipartimento di Scienze Mediche Traslazionali, University of Naples "Federico II," Naples, Italy.,Istituto di Endocrinologia ed Oncologia Sperimentale del CNR "G. Salvatore," Naples, Italy
| | - Federica Liotti
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, University of Naples "Federico II," Naples, Italy
| | - Anna Illiano
- Dipartimento di Scienze Chimiche, University of Naples "Federico II," Naples, Italy
| | - Angela Amoresano
- Dipartimento di Scienze Chimiche, University of Naples "Federico II," Naples, Italy
| | - Piero Pucci
- Dipartimento di Scienze Chimiche, University of Naples "Federico II," Naples, Italy
| | - Amato de Paulis
- Dipartimento di Scienze Mediche Traslazionali, University of Naples "Federico II," Naples, Italy
| | - Rosa Marina Melillo
- Istituto di Endocrinologia ed Oncologia Sperimentale del CNR "G. Salvatore," Naples, Italy.,Dipartimento di Medicina Molecolare e Biotecnologie Mediche, University of Naples "Federico II," Naples, Italy
| |
Collapse
|
25
|
Polizzi A, Fouché E, Ducheix S, Lasserre F, Marmugi AP, Mselli-Lakhal L, Loiseau N, Wahli W, Guillou H, Montagner A. Hepatic Fasting-Induced PPARα Activity Does Not Depend on Essential Fatty Acids. Int J Mol Sci 2016; 17:ijms17101624. [PMID: 27669233 PMCID: PMC5085657 DOI: 10.3390/ijms17101624] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 09/05/2016] [Accepted: 09/15/2016] [Indexed: 12/13/2022] Open
Abstract
The liver plays a central role in the regulation of fatty acid metabolism, which is highly sensitive to transcriptional responses to nutrients and hormones. Transcription factors involved in this process include nuclear hormone receptors. One such receptor, PPARα, which is highly expressed in the liver and activated by a variety of fatty acids, is a critical regulator of hepatic fatty acid catabolism during fasting. The present study compared the influence of dietary fatty acids and fasting on hepatic PPARα-dependent responses. Pparα−/− male mice and their wild-type controls were fed diets containing different fatty acids for 10 weeks prior to being subjected to fasting or normal feeding. In line with the role of PPARα in sensing dietary fatty acids, changes in chronic dietary fat consumption influenced liver damage during fasting. The changes were particularly marked in mice fed diets lacking essential fatty acids. However, fasting, rather than specific dietary fatty acids, induced acute PPARα activity in the liver. Taken together, the data imply that the potent signalling involved in triggering PPARα activity during fasting does not rely on essential fatty acid-derived ligand.
Collapse
Affiliation(s)
- Arnaud Polizzi
- INRA ToxAlim, 180, Chemin de Tournefeuille, 31027 Toulouse Cedex 3, France.
| | - Edwin Fouché
- INRA ToxAlim, 180, Chemin de Tournefeuille, 31027 Toulouse Cedex 3, France.
| | - Simon Ducheix
- INRA ToxAlim, 180, Chemin de Tournefeuille, 31027 Toulouse Cedex 3, France.
| | - Frédéric Lasserre
- INRA ToxAlim, 180, Chemin de Tournefeuille, 31027 Toulouse Cedex 3, France.
| | - Alice P Marmugi
- INRA ToxAlim, 180, Chemin de Tournefeuille, 31027 Toulouse Cedex 3, France.
| | | | - Nicolas Loiseau
- INRA ToxAlim, 180, Chemin de Tournefeuille, 31027 Toulouse Cedex 3, France.
| | - Walter Wahli
- INRA ToxAlim, 180, Chemin de Tournefeuille, 31027 Toulouse Cedex 3, France.
- Lee Kong Chian School of Medicine, Nanyang Technological University, 637553 Singapore, Singapore.
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland.
| | - Hervé Guillou
- INRA ToxAlim, 180, Chemin de Tournefeuille, 31027 Toulouse Cedex 3, France.
| | | |
Collapse
|
26
|
Tan NS, Vázquez-Carrera M, Montagner A, Sng MK, Guillou H, Wahli W. Transcriptional control of physiological and pathological processes by the nuclear receptor PPARβ/δ. Prog Lipid Res 2016; 64:98-122. [PMID: 27665713 DOI: 10.1016/j.plipres.2016.09.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 08/31/2016] [Accepted: 09/20/2016] [Indexed: 12/14/2022]
Affiliation(s)
- Nguan Soon Tan
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, 637551, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Academia, 20 College Road, 169856, Singapore; Institute of Molecular and Cell Biology, 61 Biopolis Drive, Proteos, Agency for Science Technology & Research, 138673, Singapore; KK Research Centre, KK Women's and Children's Hospital, 100 Bukit Timah Road, 229899, Singapore.
| | - Manuel Vázquez-Carrera
- Department of Pharmacology and Therapeutic Chemistry, Faculty of Pharmacy, University of Barcelona, Institute of Biomedicine of the University of Barcelona (IBUB), Pediatric Research Institute-Hospital Sant Joan de Déu, Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Diseases (CIBERDEM), Barcelona, Spain
| | | | - Ming Keat Sng
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, 637551, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Academia, 20 College Road, 169856, Singapore
| | - Hervé Guillou
- INRA ToxAlim, UMR1331, Chemin de Tournefeuille, Toulouse Cedex 3, France
| | - Walter Wahli
- Lee Kong Chian School of Medicine, Nanyang Technological University, Academia, 20 College Road, 169856, Singapore; INRA ToxAlim, UMR1331, Chemin de Tournefeuille, Toulouse Cedex 3, France; Center for Integrative Genomics, University of Lausanne, Le Génopode, CH 1015 Lausanne, Switzerland.
| |
Collapse
|
27
|
Dupuy A, Le Faouder P, Vigor C, Oger C, Galano JM, Dray C, Lee JCY, Valet P, Gladine C, Durand T, Bertrand-Michel J. Simultaneous quantitative profiling of 20 isoprostanoids from omega-3 and omega-6 polyunsaturated fatty acids by LC-MS/MS in various biological samples. Anal Chim Acta 2016; 921:46-58. [PMID: 27126789 DOI: 10.1016/j.aca.2016.03.024] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 03/10/2016] [Accepted: 03/16/2016] [Indexed: 10/22/2022]
Abstract
Isoprostanoids are a group of non-enzymatic oxygenated metabolites of polyunsaturated fatty acids. It belongs to oxylipins group, which are important lipid mediators in biological processes, such as tissue repair, blood clotting, blood vessel permeability, inflammation and immunity regulation. Recently, isoprostanoids from eicosapentaenoic, docosahexaenoic, adrenic and α-linolenic namely F3-isoprostanes, F4-neuroprostanes, F2-dihomo-isoprostanes and F1-phytoprostanes, respectively have attracted attention because of their putative contribution to health. Since isoprostanoids are derived from different substrate of PUFAs and can have similar or opposing biological consequences, a total isoprostanoids profile is essential to understand the overall effect in the testing model. However, the concentration of most isoprostanoids range from picogram to nanogram, therefore a sensitive method to quantify 20 isoprostanoids simultaneously was formulated and measured by liquid chromatography-tandem mass spectrometry (LC-MS/MS). The lipid portion from various biological samples was extracted prior to LC-MS/MS evaluation. For all the isoprostanoids LOD and LOQ, and the method was validated on plasma samples for matrix effect, yield of extraction and reproducibility were determined. The methodology was further tested for the isoprostanoids profiles in brain and liver of LDLR(-/-) mice with and without docosahexaenoic acid (DHA) supplementation. Our analysis showed similar levels of total F2-isoprostanes and F4-neuroprostanes in the liver and brain of non-supplemented LDLR(-/-) mice. The distribution of different F2-isoprostane isomers varied between tissues but not for F4-neuroprostanes which were predominated by the 4(RS)-4-F4t-neuroprostane isomer. DHA supplementation to LDLR(-/-) mice concomitantly increased total F4-neuroprostanes levels compared to F2-isoprostanes but this effect was more pronounced in the liver than brain.
Collapse
Affiliation(s)
- Aude Dupuy
- MetaToul-Lipidomic Core Facility, MetaboHUB, Inserm U1048, Toulouse, France; Institut des Maladies Métaboliques et Cardiovasculaires, Inserm U1048, Toulouse, France
| | - Pauline Le Faouder
- MetaToul-Lipidomic Core Facility, MetaboHUB, Inserm U1048, Toulouse, France
| | - Claire Vigor
- Institut des Biomolécules Max Mousseron IBMM, UMR 5247 CNRS, Université de Montpellier, ENSCM, France
| | - Camille Oger
- Institut des Biomolécules Max Mousseron IBMM, UMR 5247 CNRS, Université de Montpellier, ENSCM, France
| | - Jean-Marie Galano
- Institut des Biomolécules Max Mousseron IBMM, UMR 5247 CNRS, Université de Montpellier, ENSCM, France
| | - Cédric Dray
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm U1048, Toulouse, France
| | | | - Philippe Valet
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm U1048, Toulouse, France
| | - Cécile Gladine
- INRA, UMR1019, UNH, CRNH Auvergne, Clermont Université, Université d'Auvergne, Unité de Nutrition Humaine, BP 10448, F-63000 Clermont-Ferrand, France
| | - Thierry Durand
- Institut des Biomolécules Max Mousseron IBMM, UMR 5247 CNRS, Université de Montpellier, ENSCM, France
| | | |
Collapse
|
28
|
Fell GL, Nandivada P, Gura KM, Puder M. Intravenous Lipid Emulsions in Parenteral Nutrition. Adv Nutr 2015; 6:600-10. [PMID: 26374182 PMCID: PMC4561835 DOI: 10.3945/an.115.009084] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Fat is an important macronutrient in the human diet. For patients with intestinal failure who are unable to absorb nutrients via the enteral route, intravenous lipid emulsions play a critical role in providing an energy-dense source of calories and supplying the essential fatty acids that cannot be endogenously synthesized. Over the last 50 y, lipid emulsions have been an important component of parenteral nutrition (PN), and over the last 10-15 y many new lipid emulsions have been manufactured with the goal of improving safety and efficacy profiles and achieving physiologically optimal formulations. The purpose of this review is to provide a background on the components of lipid emulsions, their role in PN, and to discuss the lipid emulsions available for intravenous use. Finally, the role of parenteral fat emulsions in the pathogenesis and management of PN-associated liver disease in PN-dependent pediatric patients is reviewed.
Collapse
Affiliation(s)
| | | | | | - Mark Puder
- Vascular Biology Program and Departments of Surgery and
| |
Collapse
|
29
|
Tremblay-Franco M, Zerbinati C, Pacelli A, Palmaccio G, Lubrano C, Ducheix S, Guillou H, Iuliano L. Effect of obesity and metabolic syndrome on plasma oxysterols and fatty acids in human. Steroids 2015; 99:287-92. [PMID: 25896943 DOI: 10.1016/j.steroids.2015.03.019] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Revised: 03/19/2015] [Accepted: 03/23/2015] [Indexed: 01/29/2023]
Abstract
BACKGROUND Obesity and the related entity metabolic syndrome are characterized by altered lipid metabolism and associated with increased morbidity risk for cardiovascular disease and cancer. Oxysterols belong to a large family of cholesterol-derived molecules known to play crucial role in many signaling pathways underlying several diseases. Little is known on the potential effect of obesity and metabolic syndrome on oxysterols in human. OBJECTIVES In this work, we questioned whether circulating oxysterols might be significantly altered in obese patients and in patients with metabolic syndrome. We also tested the potential correlation between circulating oxysterols and fatty acids. METHODS 60 obese patients and 75 patients with metabolic syndrome were enrolled in the study along with 210 age- and sex-matched healthy subjects, used as control group. Plasma oxysterols were analyzed by isotope dilution GC/MS, and plasma fatty acids profiling was assessed by gas chromatography coupled with flame ionization detection. RESULTS We found considerable differences in oxysterols profiling in the two disease groups that were gender-related. Compared to controls, males showed significant differences only in 4α- and 4β-hydroxycholesterol levels in obese and metabolic syndrome patients. In contrast, females showed consistent differences in 7-oxocholesterol, 4α-hydroxycholesterol, 25-hydroxycholesterol and triol. Concerning fatty acids, we found minor differences in the levels of these variables in males of the three groups. Significant changes were observed in plasma fatty acid profile of female patients with obesity or metabolic syndrome. We found significant correlations between various oxysterols and fatty acids. In particular, 4β-hydroxycholesterol, which is reduced in obesity and metabolic syndrome, correlated with a number of saturated and mono-unsaturated fatty acids that are end-products of de novo lipogenesis. CONCLUSIONS Our data provide the first evidence that obesity and metabolic syndrome are associated with major, gender-specific, changes in circulating oxysterols and fatty acids. These findings suggest a metabolic link between oxysterols and fatty acids, and that oxysterols may contribute to the epidemic diseases associated with obesity and metabolic syndrome in female.
Collapse
Affiliation(s)
| | - Chiara Zerbinati
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Antonio Pacelli
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Giuseppina Palmaccio
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Carla Lubrano
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - Simon Ducheix
- INRA, ToxAlim UMR1331 (Research Centre in Food Toxicology), Toulouse, France
| | - Hervé Guillou
- INRA, ToxAlim UMR1331 (Research Centre in Food Toxicology), Toulouse, France
| | - Luigi Iuliano
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy.
| |
Collapse
|
30
|
Renga B, Festa C, De Marino S, Di Micco S, D'Auria MV, Bifulco G, Fiorucci S, Zampella A. Molecular decodification of gymnemic acids from Gymnema sylvestre. Discovery of a new class of liver X receptor antagonists. Steroids 2015; 96:121-31. [PMID: 25668616 DOI: 10.1016/j.steroids.2015.01.024] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 12/19/2014] [Accepted: 01/29/2015] [Indexed: 12/19/2022]
Abstract
The individual chemical components of commercial extract of Gymnema sylvestre, a medicinal plant used in the traditional systems of the Indian medicine for its antidiabetic and hypolipidemic properties, were isolated and evaluated for their capability to act as modulators of nuclear and membrane receptors involved in glucose and lipid homeostasis. The study disclosed for the first time that individual gymnemic acids are potent and selective antagonists for the β isoform of LXR. Indeed the above activity was shared by the most abundant aglycone gymnemagenin (10) whereas gymnestrogenin (11) was endowed with a dual LXRα/β antagonistic profile. Deep pharmacological investigation demonstrated that gymnestrogenin, reducing the expression of SREBP1c and ABCA1 in vitro, is able to decrease lipid accumulation in HepG2 cells. The results of this study substantiate the use of G. sylvestre extract in LXR mediated dislypidemic diseases.
Collapse
Affiliation(s)
- Barbara Renga
- Department of Surgery and Biomedical Sciences, Nuova Facoltà di Medicina, P.zza L. Severi, 1-06132 Perugia, Italy
| | - Carmen Festa
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano, 49, I-80131 Naples, Italy
| | - Simona De Marino
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano, 49, I-80131 Naples, Italy
| | - Simone Di Micco
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano (Salerno), Italy
| | - Maria Valeria D'Auria
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano, 49, I-80131 Naples, Italy
| | - Giuseppe Bifulco
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano (Salerno), Italy
| | - Stefano Fiorucci
- Department of Surgery and Biomedical Sciences, Nuova Facoltà di Medicina, P.zza L. Severi, 1-06132 Perugia, Italy
| | - Angela Zampella
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano, 49, I-80131 Naples, Italy.
| |
Collapse
|
31
|
Double-blind randomized placebo-controlled clinical trial of omega 3 fatty acids for the treatment of diabetic patients with nonalcoholic steatohepatitis. J Clin Gastroenterol 2015; 49:137-44. [PMID: 24583757 PMCID: PMC4147029 DOI: 10.1097/mcg.0000000000000099] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
BACKGROUND Nonalcoholic steatohepatitis (NASH) is common and severe in patients with diabetes mellitus. Although, there are no effective treatments for NASH in diabetic patients, preliminary reports suggest that polyunsaturated fatty acids (PUFA) may be beneficial in these patients. AIM A prospective, randomized, double-blind placebo-controlled study (NCT 00323414) was performed in NASH patients with diabetes. Clinicaltrials.gov (NCT 00323414). SUBJECTS AND METHODS A total of 37 patients (50.6 ± 9.8 y) with well-controlled diabetes (HbA1C<8.5%) were randomized to receive either PUFA containing eicosapentaenoic acid (2160 mg) and docosahexaenoic acid (1440 mg) daily or an isocaloric, identical placebo containing corn oil for 48 weeks under CONSORT guidelines. Clinical, demographics, biochemical laboratory tests, body composition using DEXA, and liver biopsy were performed at randomization and at the end of treatment. Liver biopsy was scored by the NASH CRN criteria. An intention-to-treat analysis was performed. RESULTS At inclusion, sex, age, body weight, biochemical tests, glucose control, and liver histology were similar in the 2 treatment groups. There was no change in liver enzymes, body weight, or body composition during the study in either group. At the end of the treatment, hepatic steatosis and the activity score improved (P<0.05) and lobular inflammation worsened (P<0.001) with placebo but was unchanged with PUFA. At the end of the treatment, insulin resistance (serum glucose and HOMA) worsened with PUFA but not placebo. CONCLUSIONS PUFA provided no benefit over placebo in NASH patients with diabetes. The effects of PUFA on histology and insulin resistance were inferior to placebo. These data provide no support for PUFA supplements in NASH.
Collapse
|
32
|
Baiceanu E, Crisan G, Loghin F, Falson P. Modulators of the human ABCC2: hope from natural sources? Future Med Chem 2015; 7:2041-63. [PMID: 26496229 DOI: 10.4155/fmc.15.131] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Human ABCC2 is an ATP-binding cassette transporter involved in the export of endobiotics and xenobiotics. It is involved in cisplatin resistance in cancer cells, particularly in ovarian cancer. The few known ABCC2 modulators are poorly efficient, so it is necessary to explore new ways to select and optimize efficient compounds ABCC2. Natural products offer an original scaffold for such a strategy and brings hope for this aim. This review covers basic knowledge about ABCC2, from distribution and topology aspects to physiological and pathological functions. It summarizes the effect of natural products as ABCC2 modulators. Certain plant metabolites act on different ABCC2 regulation levels and therefore are promising candidates to block the multidrug resistance mediated by ABCC2 in cancer cells.
Collapse
Affiliation(s)
- Elisabeta Baiceanu
- Drug Resistance Modulation & Membrane Proteins Laboratory, Molecular & Structural Basis of Infectious Systems, Mixed Research Unit between the National Centre for Scientific Research & Lyon I University n 5086, Institute of Biology & Chemistry of Proteins, 7 passage du Vercors 69367, Lyon, Cedex, France
- Pharmaceutical Botany Department, Faculty of Pharmacy, University of Medicine & Pharmacy 'Iuliu Haţieganu' Cluj-Napoca, 23 Marinescu Street, Cluj-Napoca, Romania
| | - Gianina Crisan
- Pharmaceutical Botany Department, Faculty of Pharmacy, University of Medicine & Pharmacy 'Iuliu Haţieganu' Cluj-Napoca, 23 Marinescu Street, Cluj-Napoca, Romania
| | - Felicia Loghin
- Toxicology Department, Faculty of Pharmacy, University of Medicine & Pharmacy 'Iuliu Haţieganu' Cluj-Napoca, 5-9 Louis Pasteur Street, Cluj-Napoca, Romania
| | - Pierre Falson
- Drug Resistance Modulation & Membrane Proteins Laboratory, Molecular & Structural Basis of Infectious Systems, Mixed Research Unit between the National Centre for Scientific Research & Lyon I University n 5086, Institute of Biology & Chemistry of Proteins, 7 passage du Vercors 69367, Lyon, Cedex, France
| |
Collapse
|
33
|
Gobbetti T, Ducheix S, le Faouder P, Perez T, Riols F, Boue J, Bertrand-Michel J, Dubourdeau M, Guillou H, Perretti M, Vergnolle N, Cenac N. Protective effects of n-6 fatty acids-enriched diet on intestinal ischaemia/reperfusion injury involve lipoxin A4 and its receptor. Br J Pharmacol 2014; 172:910-23. [PMID: 25296998 DOI: 10.1111/bph.12957] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 09/12/2014] [Accepted: 09/22/2014] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND AND PURPOSE Long-term intake of dietary fatty acids is known to predispose to chronic inflammation, but their effects on acute intestinal ischaemia/reperfusion (I/R) injury is unknown. The aim of this study was to determine the consequences of a diet rich in n-3 or n-6 polyunsaturated fatty acids (PUFA) on intestinal I/R-induced damage. EXPERIMENTAL APPROACH Mice were fed three different isocaloric diets: a balanced diet used as a control and two different PUFA-enriched diets, providing either high levels of n-3 or of n-6 PUFA. Intestinal injury was evaluated after intestinal I/R. PUFA metabolites were quantitated in intestinal tissues by LC-MS/MS. KEY RESULTS In control diet-fed mice, intestinal I/R caused inflammation and increased COX and lipoxygenase-derived metabolites compared with sham-operated animals. Lipoxin A4 (LxA4 ) was significantly and selectively increased after ischaemia. Animals fed a high n-3 diet did not display a different inflammatory profile following intestinal I/R compared with control diet-fed animals. In contrast, intestinal inflammation was decreased in the I/R group fed with high n-6 diet and level of LxA4 was increased post-ischaemia compared with control diet-fed mice. Blockade of the LxA4 receptor (Fpr2), prevented the anti-inflammatory effects associated with the n-6 rich diet. CONCLUSIONS AND IMPLICATIONS This study indicates that high levels of dietary n-6, but not n-3, PUFAs provides significant protection against intestinal I/R-induced damage and demonstrates that the endogenous production of LxA4 can be influenced by diet.
Collapse
Affiliation(s)
- T Gobbetti
- Inserm, U1043, Toulouse, France; CNRS, U5282, Toulouse, France; Centre de Physiopathologie de Toulouse Purpan (CPTP), Université de Toulouse, Université Paul Sabatier, Toulouse, France; WHRI, Queen Mary University, London, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Expression of liver X receptor correlates with intrahepatic inflammation and fibrosis in patients with nonalcoholic fatty liver disease. Dig Dis Sci 2014; 59:2975-82. [PMID: 25102981 DOI: 10.1007/s10620-014-3289-x] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 07/11/2014] [Indexed: 01/20/2023]
Abstract
BACKGROUND Liver X receptor (LXR) is an oxysterol-activated nuclear receptor involved in the control of major metabolic pathways for cholesterol homeostasis and lipogenesis. Although the role of LXR in hepatic steatosis is well known, its correlation with intrahepatic inflammation and fibrosis has not been thoroughly studied. We investigated the association between LXRα, hepatic inflammation, and fibrosis, as well as its correlation with other intrahepatic lipid transporters in patients with nonalcoholic fatty liver disease (NAFLD). METHODS We evaluated clinical characteristics including sex, age, body mass index, and laboratory findings from 40 NAFLD and 16 control patients. Immunohistochemical staining was carried out on liver biopsy samples from all patients. RESULTS The positive rate of LXRα expression was 30 % in the control group, 50 % in the NAFLD group, and 97 % in NASH groups. LXRα expression was positively correlated with not only the amount of intrahepatic fat, but also with intrahepatic inflammation and hepatic fibrosis. LXRα expression showed positive correlation with intrahepatic expression of ABCG5/8, CD36, and SREBP-1c. The expression of ABCA1, ABCG5/8, SREBP-1c, and CD36 was higher in NAFLD than in controls and there was no further increase in the NASH group. NPC1L1 was abundant in human liver. Expression of NPC1L1 was negatively correlated with intrahepatic inflammation and LXRα intensity. CONCLUSION LXR expression correlated with the degree of hepatic fat deposition, as well as with hepatic inflammation and fibrosis in NAFLD patients. Our research suggests that LXR is an attractive target for treatment and regulation of hepatic inflammation and fibrosis.
Collapse
|
35
|
Sim WC, Park S, Lee KY, Je YT, Yin HQ, Choi YJ, Sung SH, Park SJ, Park HJ, Shin KJ, Lee BH. LXR-α antagonist meso-dihydroguaiaretic acid attenuates high-fat diet-induced nonalcoholic fatty liver. Biochem Pharmacol 2014; 90:414-24. [PMID: 24955981 DOI: 10.1016/j.bcp.2014.06.013] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Revised: 06/14/2014] [Accepted: 06/16/2014] [Indexed: 01/17/2023]
Abstract
Collaborative regulation of liver X receptor (LXR) and sterol regulatory element binding protein (SREBP)-1 are main determinants in hepatic steatosis, as shown in both animal models and human patients. Recent studies indicate that selective intervention of overly functional LXRα in the liver shows promise in treatment of fatty liver disease. In the present study, we evaluated the effects of meso-dihydroguaiaretic acid (MDGA) on LXRα activation and its ability to attenuate fatty liver in mice. MDGA inhibited activation of the LXRα ligand-binding domain by competitively binding to the pocket for agonist T0901317 and decreased the luciferase activity in LXRE-tk-Luc-transfected cells. MDGA significantly attenuated hepatic neutral lipid accumulation in T0901317- and high fat diet (HFD)-induced fatty liver. The effect of MDGA was so potent that treatment with 1mg/kg for 2 weeks completely reversed the lipid accumulation induced by HFD feeding. MDGA reduced the expression of LXRα co-activator protein RIP140 and LXRα target gene products associated with lipogenesis in HFD-fed mice. These results demonstrate that MDGA has the potential to attenuate nonalcoholic steatosis mediated by selective inhibition of LXRα in the liver in mice.
Collapse
Affiliation(s)
- Woo-Cheol Sim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Gwanak-ro 1, Gwanak-gu, Seoul 151-742, Republic of Korea
| | - Sora Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Gwanak-ro 1, Gwanak-gu, Seoul 151-742, Republic of Korea
| | - Kang-Yo Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Gwanak-ro 1, Gwanak-gu, Seoul 151-742, Republic of Korea
| | - Young-Tae Je
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Gwanak-ro 1, Gwanak-gu, Seoul 151-742, Republic of Korea
| | - Hu-Quan Yin
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Gwanak-ro 1, Gwanak-gu, Seoul 151-742, Republic of Korea
| | - You-Jin Choi
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Gwanak-ro 1, Gwanak-gu, Seoul 151-742, Republic of Korea
| | - Sang Hyun Sung
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Gwanak-ro 1, Gwanak-gu, Seoul 151-742, Republic of Korea
| | - So-Jung Park
- School of Pharmacy, Sungkyunkwan University, Seobu-ro 2066, Jangan-gu, Suwon 440-746, Republic of Korea
| | - Hyun-Ju Park
- School of Pharmacy, Sungkyunkwan University, Seobu-ro 2066, Jangan-gu, Suwon 440-746, Republic of Korea
| | - Kye Jung Shin
- College of Pharmacy, The Catholic University, Bucheon 420-743, Republic of Korea
| | - Byung-Hoon Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Gwanak-ro 1, Gwanak-gu, Seoul 151-742, Republic of Korea.
| |
Collapse
|
36
|
Harvatine KJ, Boisclair YR, Bauman DE. Liver x receptors stimulate lipogenesis in bovine mammary epithelial cell culture but do not appear to be involved in diet-induced milk fat depression in cows. Physiol Rep 2014; 2:e00266. [PMID: 24760520 PMCID: PMC4002246 DOI: 10.1002/phy2.266] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Abstract Milk fat synthesis of ruminants can be inhibited by intermediates of ruminal fatty acid biohydrogenation including trans-10, cis-12 conjugated linoleic acid (CLA). These biohydrogenation intermediates signal a coordinated downregulation of genes involved in mammary FA synthesis, transport, and esterification. We have previously reported decreased mammary expression of sterol response element-binding protein 1 (SREBP1), SREBP1-activating proteins, and thyroid hormone-responsive spot 14 (S14) in the cow during diet-induced milk fat depression (MFD), and treatment with trans-10, cis-12 CLA. Liver x receptors (LXR) and retinoid x receptors (RXR) regulate lipogenesis and are known to bind polyunsaturated FA and LXR agonist increases lipid synthesis in mammary epithelial cell culture. The current studies investigated if biohydrogenation products of rumen origin inhibit mammary lipogenesis through LXR and/or RXR. Expression of LXRs was not different in lactating compared to nonlactating bovine mammary tissue, and expression of LXRs, RXRα, and selected LXR and RXR target genes was not changed in mammary tissue during diet-induced or CLA-induced MFD in the cow. In bovine mammary epithelial cell culture, LXR agonist stimulated lipogenesis and expression of LXRß, ATP-binding cassette 1 (ABCA1), SREBP1c, and S14, but LXR activation did not overcome CLA inhibition of lipogenesis and downregulation of LXRß, SREBP1c, and S14 expression. Lastly, expression of the LXR-regulated carbohydrate-responsive element-binding protein (ChREBP) was higher in lactating than nonlactating tissue and was decreased during CLA-induced MFD. We conclude that changes in mammary LXR expression in dairy cows are not involved in MFD and that trans-10, cis-12 CLA inhibition of lipogenesis and diet-induced MFD appears independent of direct LXR signaling.
Collapse
Affiliation(s)
- Kevin J Harvatine
- Department of Animal Science, Penn State University, University Park, Pennsylvania
| | | | | |
Collapse
|
37
|
Ren S, Ning Y. Sulfation of 25-hydroxycholesterol regulates lipid metabolism, inflammatory responses, and cell proliferation. Am J Physiol Endocrinol Metab 2014; 306:E123-30. [PMID: 24302009 PMCID: PMC3920008 DOI: 10.1152/ajpendo.00552.2013] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Intracellular lipid accumulation, inflammatory responses, and subsequent apoptosis are the major pathogenic events of metabolic disorders, including atherosclerosis and nonalcoholic fatty liver diseases. Recently, a novel regulatory oxysterol, 5-cholesten-3b, 25-diol 3-sulfate (25HC3S), has been identified, and hydroxysterol sulfotransferase 2B1b (SULT2B1b) has been elucidated as the key enzyme for its biosynthesis from 25-hydroxycholesterol (25HC) via oxysterol sulfation. The product 25HC3S and the substrate 25HC have been shown to coordinately regulate lipid metabolism, inflammatory responses, and cell proliferation in vitro and in vivo. 25HC3S decreases levels of the nuclear liver oxysterol receptor (LXR) and sterol regulatory element-binding proteins (SREBPs), inhibits SREBP processing, subsequently downregulates key enzymes in lipid biosynthesis, decreases intracellular lipid levels in hepatocytes and THP-1-derived macrophages, prevents apoptosis, and promotes cell proliferation in liver tissues. Furthermore, 25HC3S increases nuclear PPARγ and cytosolic IκBα and decreases nuclear NF-κB levels and proinflammatory cytokine expression and secretion when cells are challenged with LPS and TNFα. In contrast to 25HC3S, 25HC, a known LXR ligand, increases nuclear LXR and decreases nuclear PPARs and cytosol IκBα levels. In this review, we summarize our recent findings, including the discovery of the regulatory oxysterol sulfate, its biosynthetic pathway, and its functional mechanism. We also propose that oxysterol sulfation functions as a regulatory signaling pathway.
Collapse
Affiliation(s)
- Shunlin Ren
- Departments of Medicine, McGuire Veterans Affairs Medical Center/Virginia Commonwealth University, Richmond, Virginia
| | | |
Collapse
|
38
|
The liver X receptor: A master regulator of the gut–liver axis and a target for non alcoholic fatty liver disease. Biochem Pharmacol 2013; 86:96-105. [DOI: 10.1016/j.bcp.2013.03.016] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Revised: 03/21/2013] [Accepted: 03/21/2013] [Indexed: 12/15/2022]
|