1
|
Mignini I, Piccirilli G, Di Vincenzo F, Covello C, Pizzoferrato M, Esposto G, Galasso L, Borriello R, Gabrielli M, Ainora ME, Gasbarrini A, Zocco MA. Intestinal-Failure-Associated Liver Disease: Beyond Parenteral Nutrition. Biomolecules 2025; 15:388. [PMID: 40149924 PMCID: PMC11939910 DOI: 10.3390/biom15030388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/05/2025] [Accepted: 03/06/2025] [Indexed: 03/29/2025] Open
Abstract
Short bowel syndrome (SBS), usually resulting from massive small bowel resections or congenital defects, may lead to intestinal failure (IF), requiring intravenous fluids and parenteral nutrition to preserve patients' nutritional status. Approximately 15% to 40% of subjects with SBS and IF develop chronic hepatic damage during their life, a condition referred to as intestinal-failure-associated liver disease (IFALD), which ranges from steatosis to fibrosis or end-stage liver disease. Parenteral nutrition has been largely pointed out as the main pathogenetic factor for IFALD. However, other elements, such as inflammation, bile acid metabolism, bacterial overgrowth and gut dysbiosis also contribute to the development of liver damage and may deserve specific treatment strategies. Indeed, in our review, we aim to explore IFALD pathogenesis beyond parenteral nutrition. By critically analyzing recent literature, we seek to delve with molecular mechanisms and metabolic pathways underlying liver damage in such a complex set of patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Maria Assunta Zocco
- CEMAD Digestive Diseases Center, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168 Rome, Italy; (I.M.); (G.P.); (F.D.V.); (C.C.); (M.P.); (G.E.); (L.G.); (R.B.); (M.G.); (M.E.A.); (A.G.)
| |
Collapse
|
2
|
Zhang W, Guo J, Miao G, Chen J, Xu Y, Lai P, Zhang L, Han Y, Lam SM, Shui G, Wang Y, Huang W, Xian X. Fat-1 Ameliorates Metabolic Dysfunction-Associated Fatty Liver Disease and Atherosclerosis through Promoting the Nuclear Localization of PPARα in Hamsters. RESEARCH (WASHINGTON, D.C.) 2025; 8:0577. [PMID: 40052160 PMCID: PMC11884683 DOI: 10.34133/research.0577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 12/16/2024] [Accepted: 12/20/2024] [Indexed: 03/09/2025]
Abstract
Fat-1, an enzyme encoded by the fat-1 gene, is responsible for the conversion of endogenous omega-6 polyunsaturated fatty acids into omega-3 polyunsaturated fatty acids in Caenorhabditis elegans. To better investigate whether the expression of Fat-1 will exert a beneficial function in dyslipidemia and metabolic dysfunction-associated fatty liver disease (MAFLD), we established an adeno-associated virus 9 expressing Fat-1. We found that adeno-associated-virus-mediated expression of Fat-1 markedly reduced the levels of plasma triglycerides and total cholesterol but increased high-density lipoprotein levels in male wild-type hamsters on both chow diet and high-fat diet as well as in chow-diet-fed male LDLR-/- hamsters. Fat-1 ameliorated diet-induced MAFLD in wild-type hamsters by enhancing fatty acid oxidation through the hepatic peroxisome proliferator-activated receptor α (PPARα)-dependent pathway. Mechanistically, Fat-1 increased the levels of multiple lipid derivatives as ligands for PPARα and simultaneously facilitated the nuclear localization of PPARα. Our results provide new insights into the multiple therapeutic potentials of Fat-1 to treat dyslipidemia, MAFLD, and atherosclerosis.
Collapse
Affiliation(s)
- Wenxi Zhang
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences,
Peking University, Beijing 100191, China
| | - Jiabao Guo
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences,
Peking University, Beijing 100191, China
| | - Guolin Miao
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences,
Peking University, Beijing 100191, China
| | - Jingxuan Chen
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences,
Peking University, Beijing 100191, China
| | - Yitong Xu
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences,
Peking University, Beijing 100191, China
| | - Pingping Lai
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences,
Peking University, Beijing 100191, China
| | - Lianxin Zhang
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences,
Peking University, Beijing 100191, China
| | - Yufei Han
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences,
Peking University, Beijing 100191, China
| | - Sin Man Lam
- State Key Laboratory of Molecular Developmental Biology,
Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- LipidALL Technologies Company Limited, Changzhou 213022, Jiangsu Province, China
| | - Guanghou Shui
- State Key Laboratory of Molecular Developmental Biology,
Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yuhui Wang
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences,
Peking University, Beijing 100191, China
| | - Wei Huang
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences,
Peking University, Beijing 100191, China
| | - Xunde Xian
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences,
Peking University, Beijing 100191, China
- Beijing Key Laboratory of Cardiovascular Receptors Research,
Peking University Third Hospital, Beijing 100191, China
| |
Collapse
|
3
|
Bril F, Berg G, Barchuk M, Nogueira JP. Practical Approaches to Managing Dyslipidemia in Patients With Metabolic Dysfunction-Associated Steatotic Liver Disease. J Lipid Atheroscler 2025; 14:5-29. [PMID: 39911965 PMCID: PMC11791423 DOI: 10.12997/jla.2025.14.1.5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 02/15/2024] [Accepted: 03/10/2024] [Indexed: 02/07/2025] Open
Abstract
Dyslipidemia is a major risk factor for cardiovascular disease, and its impact may be exacerbated when accompanied by metabolic dysfunction-associated steatotic liver disease (MASLD). The simultaneous management of these conditions poses multiple challenges for healthcare providers. Insulin resistance has been implicated in the pathogenesis of both dyslipidemia and MASLD, necessitating a holistic approach to managing dyslipidemia, glucose levels, body weight, and MASLD. This review explores the intricate pathophysiological relationship between MASLD and dyslipidemia. It also examines current guidance regarding the use of lipid-lowering agents (including statins, ezetimibe, fibrates, omega-3 polyunsaturated fatty acids, and proprotein convertase subtilisin/kexin type 9 inhibitors) as well as glucose-lowering medications (such as pioglitazone, glucagon-like peptide-1 receptor agonists, and sodium-glucose cotransporter 2 inhibitors) in patients with MASLD, with or without metabolic dysfunction-associated steatohepatitis (MASH), and dyslipidemia. Additionally, the review addresses the potential of emerging drugs to concurrently target both MASLD/MASH and dyslipidemia. Our hope is that a deeper understanding of the mechanisms underlying MASLD and dyslipidemia may assist clinicians in the management of these complex cases.
Collapse
Affiliation(s)
- Fernando Bril
- Division of Endocrinology, Diabetes and Metabolism, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Gabriela Berg
- Facultad de Farmacia y Bioquímica, Departamento de Bioquímica Clínica, Cátedra de Bioquímica Clínica I, Laboratorio de Lípidos y Aterosclerosis, Universidad de Buenos Aires, Buenos Aires, Argentina
- CONICET, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Magali Barchuk
- Facultad de Farmacia y Bioquímica, Departamento de Bioquímica Clínica, Cátedra de Bioquímica Clínica I, Laboratorio de Lípidos y Aterosclerosis, Universidad de Buenos Aires, Buenos Aires, Argentina
- CONICET, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Juan Patricio Nogueira
- Centro de Investigación en Endocrinología, Nutrición y Metabolismo (CIENM), Facultad de Ciencias de la Salud, Universidad Nacional de Formosa, Formosa, Argentina
- Universidad Internacional de las Américas, San José, Costa Rica
| |
Collapse
|
4
|
Peng L, Shen J, Li L, Liu J, Jiang X, Zhang G, Li Y. Birthweight influences liver structure, function and disease risk: Evidence of a causal association. Diabetes Obes Metab 2024; 26:4976-4988. [PMID: 39228281 DOI: 10.1111/dom.15910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/16/2024] [Accepted: 08/16/2024] [Indexed: 09/05/2024]
Abstract
AIM Low birthweight is an issue during pregnancy associated with an increased risk of developing liver disease later in life. Previous Mendelian randomisation (MR) studies which explored this issue have not isolated the direct impact of the foetus on birthweight. In the present study, MR was used to assess whether direct foetal effects on birthweight were causally associated with liver structure, function and disease risk independent of intrauterine effects. MATERIALS AND METHODS We extracted single nucleotide polymorphisms (SNPs) from genome-wide association studies (GWAS) about direct foetal-affected birthweight (321 223 cases) to conduct univariable and multivariable MR analyses to explore the relationships between birthweight and 4 liver structure measures, 9 liver function measures and 18 liver diseases. A two-step MR analysis was used to further assess and quantify the mediating effects of the mediators. RESULTS When isolating direct foetal effects, genetically predicted lower birthweight was associated with a higher risk of non-alcoholic fatty liver disease (NAFLD) (odds ratios [OR], 95% confidence interval [CI]: 1.61, 1.29-2.02, p < 0.001), higher magnetic resonance imaging [MRI] proton density fat fraction (PDFF) and higher serum gamma glutamyltransferase (GGT). Two-step MR identified two candidate mediators that partially mediate the direct foetal effect of lower birthweight on NAFLD, including fasting insulin (proportion mediated: 22.29%) and triglycerides (6.50%). CONCLUSIONS Our MR analysis reveals a direct causal association between lower birthweight and liver MRI PDFF, as well as the development of NAFLD, which persisted even after accounting for the potential influence of maternal factors. In addition, we identified fasting insulin and triglycerides as mediators linking birthweight and hepatic outcomes, providing insights for early clinical interventions.
Collapse
Affiliation(s)
- Lei Peng
- Department of Gastroenterology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jiajia Shen
- Department of General Surgery, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Lurong Li
- Department of Gastroenterology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jiahao Liu
- Department of Gastroenterology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xingzhou Jiang
- Department of Gastroenterology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Guoxin Zhang
- Department of Gastroenterology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yuanyuan Li
- Department of Endocrinology, Children's Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
5
|
Xie P, Wu Y, Lee YY, Wang Y, Zhang Z. Asterias Rolleston starfish gonad lipids: A novel source of Omega-3 fatty acids - assessment of major components and their antioxidant activities. Food Chem 2024; 456:140005. [PMID: 38870815 DOI: 10.1016/j.foodchem.2024.140005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 05/31/2024] [Accepted: 06/06/2024] [Indexed: 06/15/2024]
Abstract
The major lipids and antioxidant activities of Asterias rolleston gonad lipids were evaluated systematically. Major lipids of A. Rolleston gonad lipids were triacylglycerols (TAGs) and phospholipids (PLs). Total lipids were composed of 15.62% of polyunsaturated fatty acids (PUFAs), and 40.81% of monounsaturated fatty acids (MUFAs). The most abundant PUFA were C20:5n-3 (EPA) (6.28%) and C22:6n-3 (DHA) (5.80%). Predominantly composed of phosphatidylcholine (PC) and phosphatidylethanolamine (PE), polar lipids were rich in PUFAs and could contain up to 34.59% EPA and DHA, and PE and PI (phosphatidylinositol) were also found to be the main carriers of EPA and ARA (arachidonic acid) in polar lipids. The MUFA and PUFA of Sn-2 in TAG are 39.72% and 30.37%, respectively. A total of 64 TAG species were identified, with Eo-P-M, Eo-Eo-M, and M-M-Eo being the main TAGs components. Moreover, A. rollestoni gonad lipids exhibited potent radical scavenging activities and reducing power in a dose-dependent manner.
Collapse
Affiliation(s)
- Pengkai Xie
- JNU-UPM International Joint Laboratory on Plant Oil Processing and Safety, Department of Food Science and Engineering, Jinan University, Guangzhou, Guangdong 510632, China
| | - Yuxin Wu
- JNU-UPM International Joint Laboratory on Plant Oil Processing and Safety, Department of Food Science and Engineering, Jinan University, Guangzhou, Guangdong 510632, China
| | - Yee-Ying Lee
- School of Science, Monash University Malaysia, Bandar Sunway 47500, Selangor, Malaysia
| | - Yong Wang
- JNU-UPM International Joint Laboratory on Plant Oil Processing and Safety, Department of Food Science and Engineering, Jinan University, Guangzhou, Guangdong 510632, China.
| | - Zhen Zhang
- JNU-UPM International Joint Laboratory on Plant Oil Processing and Safety, Department of Food Science and Engineering, Jinan University, Guangzhou, Guangdong 510632, China.
| |
Collapse
|
6
|
Zhao J, Gao G, Ding J, Liu W, Wang T, Zhao L, Xu J, Zhang Z, Zhang X, Xie Z. Astragaloside I Promotes Lipophagy and Mitochondrial Biogenesis to Improve Hyperlipidemia by Regulating Akt/mTOR/TFEB Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:21548-21559. [PMID: 39226078 DOI: 10.1021/acs.jafc.4c03172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
The simultaneous enhancement of lipophagy and mitochondrial biogenesis has emerged as a promising strategy for lipid lowering. The transcription factor EB (TFEB) exhibits a dual role, whereby it facilitates the degradation of lipid droplets (LDs) through the process of lipophagy while simultaneously stimulating mitochondrial biogenesis to support the utilization of lipophagy products. The purpose of this study was to explore the effect of astragaloside I (AS I) on hyperlipidemia and elucidate its underlying mechanism. AS I improved serum total cholesterol and triglyceride levels and reduced hepatic steatosis and lipid accumulation in db/db mice. AS I enhanced the fluorescence colocalization of LDs and autophagosomes and promoted the proteins and genes related to the autolysosome. Moreover, AS I increased the expression of mitochondrial biogenesis-related proteins and genes, indicating that AS I promoted lipophagy and mitochondrial biogenesis. Mechanistically, AS I inhibits the protein level of p-TFEB (ser211) expression and promotes TFEB nuclear translocation. The activation of TFEB by AS I was impeded upon the introduction of the mammalian target of rapamycin (mTOR) agonist MHY1485. The inhibition of p-mTOR by AS I and the activation of TFEB were no longer observed after administration of the Akt agonist SC-79, which indicated that AS I activated TFEB to promote lipophagy-dependent on the Akt/mTOR pathway and may be a potentially effective pharmaceutical and food additive for the treatment of hyperlipidemia.
Collapse
Affiliation(s)
- Jie Zhao
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Gai Gao
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Jing Ding
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Wei Liu
- Department of Pharmacy, The SATCM Third Grade Laboratory of Traditional Chinese Medicine Preparations, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200021, China
| | - Tao Wang
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Liang Zhao
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Jiangyan Xu
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Zhenqiang Zhang
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Xiaowei Zhang
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Zhishen Xie
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan, Henan University of Chinese Medicine, Zhengzhou 450046, China
| |
Collapse
|
7
|
Xue X, Wang L, Wu R, Li Y, Liu R, Ma Z, Jia K, Zhang Y, Li X. Si-Wu-Tang alleviates metabolic dysfunction-associated fatty liver disease by inhibiting ACSL4-mediated arachidonic acid metabolism and ferroptosis in MCD diet-fed mice. Chin Med 2024; 19:79. [PMID: 38844978 PMCID: PMC11157816 DOI: 10.1186/s13020-024-00953-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 05/31/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND Metabolic dysfunction-associated fatty liver disease (MAFLD) is a prevalent chronic liver disease worldwide. Si-Wu-Tang (SWT), a traditional Chinese medicine decoction has shown therapeutic effects on various liver diseases. However, the hepatoprotective effects and underlying mechanism of SWT on MAFLD remain unclear. METHODS First, a methionine-choline-deficient (MCD) diet-fed mice model was used and lipidomic analysis and transcriptomic analysis were performed. The contents of total iron ions, ferrous ions, and lipid peroxidation were detected and Prussian blue staining was performed to confirm the protective effects of SWT against ferroptosis. Finally, chemical characterization and network pharmacological analysis were employed to identify the potential active ingredients. RESULTS Serological and hepatic histopathological findings indicated SWT's discernible therapeutic impact on MCD diet-induced MAFLD. Lipidomic analysis revealed that SWT improved intrahepatic lipid accumulation by inhibiting TG synthesis and promoting TG transport. Transcriptomic analysis suggested that SWT ameliorated abnormal FA metabolism by inhibiting FA synthesis and promoting FA β-oxidation. Then, ferroptosis phenotype experiments revealed that SWT could effectively impede hepatocyte ferroptosis, which was induced by long-chain acyl-CoA synthetase 4 (ACSL4)-mediated esterification of arachidonic acid (AA). Finally, chemical characterization and network pharmacological analysis identified that paeoniflorin and other active ingredients might be responsible for the regulative effects against ferroptosis and MAFLD. CONCLUSION In conclusion, our study revealed the intricate mechanism through which SWT improved MCD diet-induced MAFLD by targeting FA metabolism and ferroptosis in hepatocytes, thus offering a novel therapeutic approach for the treatment of MAFLD and its complications.
Collapse
Affiliation(s)
- Xiaoyong Xue
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Le Wang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Ruiyu Wu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11 Bei San Huan Dong Lu, Beijing, 100029, China
| | - Yufei Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11 Bei San Huan Dong Lu, Beijing, 100029, China
| | - Runping Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11 Bei San Huan Dong Lu, Beijing, 100029, China
| | - Zhi Ma
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Kexin Jia
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yinhao Zhang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xiaojiaoyang Li
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
8
|
Šmíd V, Dvořák K, Stehnová K, Strnad H, Rubert J, Stříteský J, Staňková B, Stránská M, Hajšlová J, Brůha R, Vítek L. The Ameliorating Effects of n-3 Polyunsaturated Fatty Acids on Liver Steatosis Induced by a High-Fat Methionine Choline-Deficient Diet in Mice. Int J Mol Sci 2023; 24:17226. [PMID: 38139055 PMCID: PMC10743075 DOI: 10.3390/ijms242417226] [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: 10/31/2023] [Revised: 12/02/2023] [Accepted: 12/03/2023] [Indexed: 12/24/2023] Open
Abstract
The pathogenesis of non-alcoholic fatty liver disease (NAFLD) is associated with abnormalities of liver lipid metabolism. On the contrary, a diet enriched with n-3 polyunsaturated fatty acids (n-3-PUFAs) has been reported to ameliorate the progression of NAFLD. The aim of our study was to investigate the impact of dietary n-3-PUFA enrichment on the development of NAFLD and liver lipidome. Mice were fed for 6 weeks either a high-fat methionine choline-deficient diet (MCD) or standard chow with or without n-3-PUFAs. Liver histology, serum biochemistry, detailed plasma and liver lipidomic analyses, and genome-wide transcriptome analysis were performed. Mice fed an MCD developed histopathological changes characteristic of NAFLD, and these changes were ameliorated with n-3-PUFAs. Simultaneously, n-3-PUFAs decreased serum triacylglycerol and cholesterol concentrations as well as ALT and AST activities. N-3-PUFAs decreased serum concentrations of saturated and monounsaturated free fatty acids (FAs), while increasing serum concentrations of long-chain PUFAs. Furthermore, in the liver, the MCD significantly increased the hepatic triacylglycerol content, while the administration of n-3-PUFAs eliminated this effect. Administration of n-3-PUFAs led to significant beneficial differences in gene expression within biosynthetic pathways of cholesterol, FAs, and pro-inflammatory cytokines (IL-1 and TNF-α). To conclude, n-3-PUFA supplementation appears to represent a promising nutraceutical approach for the restoration of abnormalities in liver lipid metabolism and the prevention and treatment of NAFLD.
Collapse
Affiliation(s)
- Václav Šmíd
- 4th Department of Internal Medicine, 1st Faculty of Medicine, Charles University in Prague and General University Hospital, 128 08 Prague, Czech Republic (R.B.); (L.V.)
| | - Karel Dvořák
- 4th Department of Internal Medicine, 1st Faculty of Medicine, Charles University in Prague and General University Hospital, 128 08 Prague, Czech Republic (R.B.); (L.V.)
| | - Kamila Stehnová
- Department of Food Analysis and Nutrition, University of Chemistry and Technology, 166 28 Prague, Czech Republic; (K.S.); (J.R.); (J.H.)
| | - Hynek Strnad
- Laboratory of Genomics and Bioinformatics, Institute of Molecular Genetics of the Czech Academy of Sciences, 142 20 Prague, Czech Republic
| | - Josep Rubert
- Department of Food Analysis and Nutrition, University of Chemistry and Technology, 166 28 Prague, Czech Republic; (K.S.); (J.R.); (J.H.)
| | - Jan Stříteský
- Institute of Pathology, 1st Faculty of Medicine, Charles University in Prague and General University Hospital, 128 00 Prague, Czech Republic;
| | - Barbora Staňková
- 4th Department of Internal Medicine, 1st Faculty of Medicine, Charles University in Prague and General University Hospital, 128 08 Prague, Czech Republic (R.B.); (L.V.)
- Institute of Medical Biochemistry and Laboratory Diagnostics, 1st Faculty of Medicine, Charles University in Prague and General University Hospital, 128 08 Prague, Czech Republic
| | - Milena Stránská
- Department of Food Analysis and Nutrition, University of Chemistry and Technology, 166 28 Prague, Czech Republic; (K.S.); (J.R.); (J.H.)
| | - Jana Hajšlová
- Department of Food Analysis and Nutrition, University of Chemistry and Technology, 166 28 Prague, Czech Republic; (K.S.); (J.R.); (J.H.)
| | - Radan Brůha
- 4th Department of Internal Medicine, 1st Faculty of Medicine, Charles University in Prague and General University Hospital, 128 08 Prague, Czech Republic (R.B.); (L.V.)
| | - Libor Vítek
- 4th Department of Internal Medicine, 1st Faculty of Medicine, Charles University in Prague and General University Hospital, 128 08 Prague, Czech Republic (R.B.); (L.V.)
- Institute of Medical Biochemistry and Laboratory Diagnostics, 1st Faculty of Medicine, Charles University in Prague and General University Hospital, 128 08 Prague, Czech Republic
| |
Collapse
|
9
|
Cho AR, Kwon YJ, Lee JH. Oxidative balance score is inversely associated with the incidence of non-alcoholic fatty liver disease. Clin Nutr 2023; 42:1292-1300. [PMID: 37352819 DOI: 10.1016/j.clnu.2023.06.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 05/22/2023] [Accepted: 06/13/2023] [Indexed: 06/25/2023]
Abstract
BACKGROUND & AIMS The contribution of the balance between overall oxidative stress and antioxidant status in non-alcoholic fatty liver disease (NAFLD) is unclear. The oxidative balance score (OBS) comprises dietary and lifestyle pro- and antioxidant components that indicate the overall oxidative stress burden. We investigated the association between OBS and the incidence of NAFLD in middle-aged and older Korean adults. METHODS Among the 10,030 participants of the Korean Genome and Epidemiology Study_Ansan and Ansung cohort, 5065 without NAFLD at baseline were selected for secondary analysis. OBS was categorized into quartiles. Multiple Cox proportional hazard regression analyses were performed to estimate the hazard ratio (HR) and 95% confidence interval (CI) for NAFLD incidence by sex-specific OBS quartile groups after adjusting for confounders. RESULTS During the follow-up period (median 13.4 years), 913 (43.2%) men and 1288 (43.9%) women were newly diagnosed with NAFLD. Compared with that of the lowest quartile (Q1) group, the adjusted HR (95% CI) for NAFLD incidence in Q2, Q3, and Q4 groups was 0.85 (0.71-1.00), 0.65 (0.54-0.78), and 0.50 (0.40-0.62) in men, and 0.85 (0.73-0.99), 0.66 (0.56-0.77), and 0.48 (0.40-0.59) in women, respectively, and for NAFLD incidence, per incremental change in OBS, was 0.90 (0.87-0.92) in men and 0.88 (0.86-0.90) in women. CONCLUSIONS A higher OBS was significantly associated with a lower risk for NAFLD incidence. Maintaining a healthy lifestyle and an antioxidant-rich diet is a potentially viable strategy for preventing NAFLD.
Collapse
Affiliation(s)
- A-Ra Cho
- Department of Family Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, 06273, Republic of Korea
| | - Yu-Jin Kwon
- Department of Family Medicine, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin, 16995, Republic of Korea
| | - Jun-Hyuk Lee
- Department of Family Medicine, Nowon Eulji Medical Center, Eulji University School of Medicine, Seoul, 01830, Republic of Korea; Department of Medicine, Graduate School of Hanyang University, Seoul, 04763, Republic of Korea.
| |
Collapse
|
10
|
Yuan C, Fan J, Jiang L, Ye W, Chen Z, Wu W, Huang Q, Qian L. Integrated Analysis of Gut Microbiome and Liver Metabolome to Evaluate the Effects of Fecal Microbiota Transplantation on Lipopolysaccharide/D-galactosamine-Induced Acute Liver Injury in Mice. Nutrients 2023; 15:nu15051149. [PMID: 36904149 PMCID: PMC10005546 DOI: 10.3390/nu15051149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 02/16/2023] [Accepted: 02/22/2023] [Indexed: 03/03/2023] Open
Abstract
Acute liver failure (ALF) refers to the occurrence of massive hepatocyte necrosis in a short time, with multiple complications, including inflammatory response, hepatic encephalopathy, and multiple organ failure. Additionally, effective therapies for ALF are lacking. There exists a relationship between the human intestinal microbiota and liver, so intestinal microbiota modulation may be a strategy for therapy of hepatic diseases. In previous studies, fecal microbiota transplantation (FMT) from fit donors has been used to modulate intestinal microbiota widely. Here, we established a mouse model of lipopolysaccharide (LPS)/D-galactosamine (D-gal) induced ALF to explore the preventive and therapeutic effects of FMT, and its mechanism of action. We found that FMT decreased hepatic aminotransferase activity and serum total bilirubin levels, and decreased hepatic pro-inflammatory cytokines in LPS/D-gal challenged mice (p < 0.05). Moreover, FMT gavage ameliorated LPS/D-gal induced liver apoptosis and markedly reduced cleaved caspase-3 levels, and improved histopathological features of the liver. FMT gavage also restored LPS/D-gal-evoked gut microbiota dysbiosis by modifying the colonic microbial composition, improving the abundance of unclassified_o_Bacteroidales (p < 0.001), norank_f_Muribaculaceae (p < 0.001), and Prevotellaceae_UCG-001 (p < 0.001), while reducing that of Lactobacillus (p < 0.05) and unclassified_f_Lachnospiraceae (p < 0.05). Metabolomics analysis revealed that FMT significantly altered LPS/D-gal induced disordered liver metabolites. Pearson's correlation revealed strong correlations between microbiota composition and liver metabolites. Our findings suggest that FMT ameliorate ALF by modulating gut microbiota and liver metabolism, and can used as a potential preventive and therapeutic strategy for ALF.
Collapse
Affiliation(s)
- Chunchun Yuan
- Key Laboratory of Animal Nutrition and Feed Science in East China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jinghui Fan
- Hangzhou Academy of Agricultural Sciences, Hangzhou 310004, China
| | - Lai Jiang
- Key Laboratory of Animal Nutrition and Feed Science in East China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Wenxin Ye
- Hainan Institute of Zhejiang University, Sanya 572025, China
| | - Zhuo Chen
- Hainan Institute of Zhejiang University, Sanya 572025, China
| | - Wenzi Wu
- Hainan Institute of Zhejiang University, Sanya 572025, China
| | - Qixin Huang
- Key Laboratory of Animal Nutrition and Feed Science in East China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Lichun Qian
- Key Laboratory of Animal Nutrition and Feed Science in East China, Ministry of Agriculture, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
- Correspondence: ; Tel.: +86-571-88982171
| |
Collapse
|
11
|
Lopez-Pentecost M, Hallmark B, Thomson CA, Chilton F, Garcia DO. Association between Dietary Fatty Acid Intake and Liver Steatosis and Fibrosis in a Sample of Mexican-Origin Hispanic Adults with Overweight or Obesity. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:3103. [PMID: 36833798 PMCID: PMC9960945 DOI: 10.3390/ijerph20043103] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/07/2023] [Accepted: 02/09/2023] [Indexed: 06/18/2023]
Abstract
Rates of non-alcoholic fatty liver disease (NAFLD) vary dramatically among Hispanic subpopulations, with Mexican-origin (MO) Hispanics experiencing a disproportionate burden. This study examined dietary fatty acid (FA) intake among overweight and obese MO Hispanic adults in the United States (US) and evaluated its association with liver steatosis and fibrosis. Participants (N = 285, MO Hispanic adults) completed 24-h dietary recalls to assess dietary FA exposure. Liver steatosis and fibrosis were estimated using transient elastography (FibroScan®). Multiple regression analysis tested relationships between FA intakes and liver steatosis or fibrosis, adjusting for age, sex, body mass index (BMI) and total energy. A total of 51% (n = 145) of participants were suspected to have NAFLD and 20% self-reported a type 2 diabetes diagnosis. No significant association was observed between Linoleic Acid and α-Linolenic Acid (LA:ALA) ratio, or omega-6 to omega-3 (n-6:n-3) ratio and liver steatosis. However, a one-point increase in the LA:ALA ratio resulted in a 1.01% increase in the liver fibrosis scores (95% CI: [1.00, 1.03]; p = 0.03), and a one-point increase in the n-6:n-3 ratio resulted in a 1.02% increase in liver fibrosis score (95% CI: [1.01, 1.03]; p = 0.01). Further research is needed to determine if modulation of FA intake could reduce NAFLD risk in this high-risk population.
Collapse
Affiliation(s)
- Melissa Lopez-Pentecost
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Brian Hallmark
- The BIO5 Institute, University of Arizona, Tucson, AZ 85721, USA
| | - Cynthia A. Thomson
- Department of Health Promotion Sciences Tucson, University of Arizona, Tucson, AZ 85721, USA
| | - Floyd Chilton
- The BIO5 Institute, University of Arizona, Tucson, AZ 85721, USA
- Department of Nutritional Sciences, University of Arizona, Tucson, AZ 85721, USA
| | - David O. Garcia
- Department of Health Promotion Sciences Tucson, University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
12
|
Jiang X, Yang Q, Qu H, Chen Y, Zhu S. Endogenous n-3 PUFAs Improve Non-Alcoholic Fatty Liver Disease through FFAR4-Mediated Gut-Liver Crosstalk. Nutrients 2023; 15:nu15030586. [PMID: 36771292 PMCID: PMC9919706 DOI: 10.3390/nu15030586] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/17/2023] [Accepted: 01/19/2023] [Indexed: 01/24/2023] Open
Abstract
The gut-liver axis plays a key role in the development and progression of non-alcoholic fatty liver disease (NAFLD). Due to the complexity and incomplete understanding of the cross-talk between the gut and liver, effective therapeutic targets are largely unknown. Free fatty acid receptors (FFARs) may bridge the cross-talk between the gut and liver. FFAR4 has received considerable attention due to its important role in lipid metabolism. However, the role of FFAR4 in this cross talk in NAFLD remains unclear. In this study, mice with high endogenous n-3 PUFAs but FFAR4 deficiency were generated by crossbreeding Fat-1 and FFAR4 knockout mice. FFAR4 deficiency blocked the protective effects of high endogenous n-3 PUFAs on intestinal barrier dysfunction and hepatic steatosis. In addition, FFAR4 deficiency decreased gut microbiota diversity and enriched Rikenella, Anaerotruncus, and Enterococcus, and reduced Dubosiella, Ruminococcaceae UCG-010, Ruminococcaceae UCG-014, Coriobacteriaceae UCG-002, Faecalibaculum, Ruminococcaceae UCG-009, and Akkermansia. Notably, FFAR4 deficiency co-regulated pantothenic acid and CoA biosynthesis, β-alanine metabolism, and sphingolipid metabolism pathways in the gut and liver, potentially associated with the aggravation of NAFLD. Together, the beneficial effects of n-3 PUFAs on the gut and liver were mediated by FFAR4, providing insights on the role of FFAR4 in the treatment of NAFLD through the gut-liver axis.
Collapse
Affiliation(s)
- Xuan Jiang
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Qin Yang
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Hongyan Qu
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
| | - Yongquan Chen
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- Wuxi Translational Medicine Research Center and School of Translational Medicine, Jiangnan University, Wuxi 214122, China
| | - Shenglong Zhu
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China
- Wuxi Translational Medicine Research Center and School of Translational Medicine, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
13
|
Willis SA, Malaikah S, Parry S, Bawden S, Ennequin G, Sargeant JA, Yates T, Webb DR, Davies MJ, Stensel DJ, Aithal GP, King JA. The effect of acute and chronic exercise on hepatic lipid composition. Scand J Med Sci Sports 2023; 33:550-568. [PMID: 36610000 DOI: 10.1111/sms.14310] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 12/06/2022] [Accepted: 01/04/2023] [Indexed: 01/09/2023]
Abstract
Exercise is recommended for those with, or at risk of nonalcoholic fatty liver disease (NAFLD), owing to beneficial effects on hepatic steatosis and cardiometabolic risk. Whilst exercise training reduces total intrahepatic lipid in people with NAFLD, accumulating evidence indicates that exercise may also modulate hepatic lipid composition. This metabolic influence is important as the profile of saturated (SFA), monounsaturated (MUFA), and polyunsaturated fatty acids (PUFA) dramatically affect the metabolic consequences of hepatic lipid accumulation; with SFA being especially lipotoxic. Relatedly, obesity and NAFLD are associated with hepatic PUFA depletion and elevated SFA. This review summarizes the acute (single bout) and chronic (exercise training) effects of exercise on hepatic lipid composition in rodents (acute studies: n = 3, chronic studies: n = 13) and humans (acute studies: n = 1, chronic studies: n = 3). An increased proportion of hepatic PUFA after acute and chronic exercise is the most consistent finding of this review. Mechanistically, this may relate to an enhanced uptake of adipose-derived PUFA (reflecting habitual diet), particularly in rodents. A relative decrease in the proportion of hepatic MUFA after chronic exercise is also documented repeatedly, particularly in rodent models with elevated hepatic MUFA. This outcome is related to decreased hepatic stearoyl-CoA desaturase-1 activity in some studies. Findings regarding hepatic SFA are less consistent and limited by the absence of metabolic challenge in rodent models. These findings require confirmation in well-controlled interventions in people with NAFLD. These studies will be facilitated by recently validated magnetic resonance spectroscopy techniques, able to precisely quantify hepatic lipid composition in vivo.
Collapse
Affiliation(s)
- Scott A Willis
- National Centre for Sport and Exercise Medicine, School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK.,NIHR Leicester Biomedical Research Centre, University Hospitals of Leicester NHS Trust and the University of Leicester, Leicester, UK
| | - Sundus Malaikah
- National Centre for Sport and Exercise Medicine, School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK.,NIHR Leicester Biomedical Research Centre, University Hospitals of Leicester NHS Trust and the University of Leicester, Leicester, UK
| | - Siôn Parry
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism, University Hospitals Coventry and Warwickshire NHS Trust, Coventry, UK
| | - Stephen Bawden
- Sir Peter Mansfield Imaging Centre, University of Nottingham, Nottingham, UK.,NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham, UK
| | - Gaël Ennequin
- Laboratory of Metabolic Adaptations to Exercise Under Physiological and Pathological Conditions (AME2P), Université of Clermont Auvergne, Clermont-Ferrand, France
| | - Jack A Sargeant
- NIHR Leicester Biomedical Research Centre, University Hospitals of Leicester NHS Trust and the University of Leicester, Leicester, UK.,Diabetes Research Centre, University of Leicester, Leicester, UK
| | - Thomas Yates
- NIHR Leicester Biomedical Research Centre, University Hospitals of Leicester NHS Trust and the University of Leicester, Leicester, UK.,Diabetes Research Centre, University of Leicester, Leicester, UK
| | - David R Webb
- NIHR Leicester Biomedical Research Centre, University Hospitals of Leicester NHS Trust and the University of Leicester, Leicester, UK.,Diabetes Research Centre, University of Leicester, Leicester, UK
| | - Melanie J Davies
- NIHR Leicester Biomedical Research Centre, University Hospitals of Leicester NHS Trust and the University of Leicester, Leicester, UK.,Diabetes Research Centre, University of Leicester, Leicester, UK
| | - David J Stensel
- National Centre for Sport and Exercise Medicine, School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK.,NIHR Leicester Biomedical Research Centre, University Hospitals of Leicester NHS Trust and the University of Leicester, Leicester, UK.,Faculty of Sport Sciences, Waseda University, Tokorozawa, Japan
| | - Guruprasad P Aithal
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham, UK.,Nottingham Digestive Diseases Centre, School of Medicine, University of Nottingham, Nottingham, UK
| | - James A King
- National Centre for Sport and Exercise Medicine, School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK.,NIHR Leicester Biomedical Research Centre, University Hospitals of Leicester NHS Trust and the University of Leicester, Leicester, UK
| |
Collapse
|
14
|
Shi H, Prough RA, McClain CJ, Song M. Different Types of Dietary Fat and Fructose Interactions Result in Distinct Metabolic Phenotypes in Male Mice. J Nutr Biochem 2023; 111:109189. [PMID: 36272691 DOI: 10.1016/j.jnutbio.2022.109189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 07/19/2022] [Accepted: 09/20/2022] [Indexed: 11/06/2022]
Abstract
Fat and fructose are the two major components over-represented in the Western diet. The aim of this study was to determine the combined effects of different types of dietary fat and fructose on the development of nonalcoholic fatty liver disease (NAFLD) in a murine model. Eight-week-old male C57BL/6J mice were fed with high-fat diet enriched with saturated fat (HSF), or omega-6 polyunsaturated fat (n6HUSF), or omega-3 polyunsaturated fat (n3HUSF) with 42% of calories derived from the fat. Fructose supplementation was given via 10% fructose (w/v) in the drinking water ad libitum for 20 weeks. While both HSF and n6HUSF fed mice developed obesity, HSF fed mice exhibited severe hepatic steatosis associated with hepatomegaly and liver injury. Fructose feeding promotes the development of liver fibrosis in HSF fed mice. n6HUSF fed mice were characterized with moderate hepatic steatosis, accompanied with hypertriglyceridemia and hyperlipidemia. Notably, fructose supplementation led to remarkable glucose intolerance in n6HUSF fed mice compared to controls. Hepatic lipidomic analysis revealed that the total saturated fatty acids and total monounsaturated fatty acids were significantly increased by fructose in the free fatty acid pool in HSF fed mice. Moreover, fructose supplementation increased hepatic and plasma cholesterol levels in the HSF fed mice. Our data suggest that excess energy from HSF intake results in fat storage in the liver, likely due to impaired triglyceride secretion; whereas excess energy from n6HUSF diet is stored in the periphery. Both effects are exacerbated by fructose supplementation. n3HUSF is beneficial, even consumed with fructose.
Collapse
Affiliation(s)
- Hongxue Shi
- Department of Pharmacology and Toxicology; Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - Russell A Prough
- Hepatobiology and Toxicology Center; Department of Biochemistry and Molecular Genetics
| | - Craig J McClain
- Department of Pharmacology and Toxicology; Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition; Hepatobiology and Toxicology Center; University of Louisville Alcohol Research Center, University of Louisville School of Medicine, Louisville, Kentucky, USA; Robley Rex Veterans Affairs Medical Center, Louisville, Kentucky, USA
| | - Ming Song
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition; Hepatobiology and Toxicology Center.
| |
Collapse
|
15
|
Heinzer K, Lang S, Farowski F, Wisplinghoff H, Vehreschild MJGT, Martin A, Nowag A, Kretzschmar A, Scholz CJ, Roderburg C, Mohr R, Tacke F, Kasper P, Goeser T, Steffen HM, Demir M. Dietary omega-6/omega-3 ratio is not associated with gut microbiota composition and disease severity in patients with nonalcoholic fatty liver disease. Nutr Res 2022; 107:12-25. [PMID: 36162275 DOI: 10.1016/j.nutres.2022.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 07/24/2022] [Accepted: 07/30/2022] [Indexed: 12/27/2022]
Abstract
In this cross-sectional study, we hypothesized that a high dietary ratio of omega-6 (n-6) to omega-3 (n-3) fatty acids could be associated with an altered gut bacterial composition and with the disease severity in patients with nonalcoholic fatty liver disease (NAFLD). A total of 101 NAFLD patients were included in the study, of which 63 underwent a liver biopsy. All 101 patients completed a 14-day food and activity record. Ebispro 2016 professional software was used to calculate individual macronutrients and micronutrients consumed. Patients were grouped into 3 quantiles (Q) according to a low (Q1: <6.1, n = 34), moderate (Q2: 6.1-7.8, n = 33), or high (Q3: >7.8, n = 34) dietary n-6/n-3 ratio. Stool samples were analyzed using 16S rRNA gene sequencing. Spearman correlation coefficients and principal coordinate analysis were used to detect differences in the bacterial composition of the gut microbiota. The median dietary n-6/n-3 ratio of all patients was 6.7 (range, 3.1-14.9). No significant associations between the dietary n-6/n-3 ratio and the gut microbiota composition or disease severity were observed. However, the abundance of specific bacteria such as Catenibacterium or Lactobacillus ruminis were found to be positively correlated and the abundance of Clostridium were negatively correlated with dietary n-6 fatty acid intake. The results indicate that a high dietary n-6/n-3 ratio is probably not a highly relevant factor in the pathogenesis of human NAFLD. Further studies are needed to clarify the importance of interactions between gut bacterial taxa and n-6 fatty acids in the pathophysiology of NAFLD.
Collapse
Affiliation(s)
- Kathrin Heinzer
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Gastroenterology and Hepatology, Cologne, Germany
| | - Sonja Lang
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Gastroenterology and Hepatology, Cologne, Germany; Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Fedja Farowski
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Cologne, Germany; German Centre for Infection Research (DZIF), partner site Bonn/Cologne; Department of Internal Medicine, Infectious Diseases, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Hilmar Wisplinghoff
- Wisplinghoff Laboratories, Cologne, Germany; University of Cologne, Faculty of Medicine, Institute for Medical Microbiology, Immunology and Hygiene, University Hospital of Cologne, Cologne, Germany; Institute for Virology and Medical Microbiology, University Witten/Herdecke, Witten, Germany
| | - Maria J G T Vehreschild
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Cologne, Germany; German Centre for Infection Research (DZIF), partner site Bonn/Cologne; Department of Internal Medicine, Infectious Diseases, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Anna Martin
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Gastroenterology and Hepatology, Cologne, Germany
| | - Angela Nowag
- Wisplinghoff Laboratories, Cologne, Germany; University of Cologne, Faculty of Medicine, Institute for Medical Microbiology, Immunology and Hygiene, University Hospital of Cologne, Cologne, Germany
| | | | | | - Christoph Roderburg
- Department of Hepatology and Gastroenterology, Campus Virchow Clinic and Campus Charité Mitte, Charité Universitätsmedizin, Berlin, Germany
| | - Raphael Mohr
- Department of Hepatology and Gastroenterology, Campus Virchow Clinic and Campus Charité Mitte, Charité Universitätsmedizin, Berlin, Germany
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Campus Virchow Clinic and Campus Charité Mitte, Charité Universitätsmedizin, Berlin, Germany
| | - Philipp Kasper
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Gastroenterology and Hepatology, Cologne, Germany
| | - Tobias Goeser
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Gastroenterology and Hepatology, Cologne, Germany
| | - Hans-Michael Steffen
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Gastroenterology and Hepatology, Cologne, Germany
| | - Münevver Demir
- Department of Hepatology and Gastroenterology, Campus Virchow Clinic and Campus Charité Mitte, Charité Universitätsmedizin, Berlin, Germany.
| |
Collapse
|
16
|
Deng KQ, Huang X, Lei F, Zhang XJ, Zhang P, She ZG, Cai J, Ji YX, Li H. Role of hepatic lipid species in the progression of nonalcoholic fatty liver disease. Am J Physiol Cell Physiol 2022; 323:C630-C639. [PMID: 35759443 DOI: 10.1152/ajpcell.00123.2022] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 06/21/2022] [Accepted: 06/21/2022] [Indexed: 11/22/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) has become the most common liver disease due to the global pandemic of metabolic diseases. Dysregulation of hepatic lipid metabolism plays a central role in the initiation and progression of NAFLD. With the advancement of lipidomics, an increasing number of lipid species and underlying mechanisms associating hepatic lipid components have been revealed. Therefore, the focus of this review is to highlight the links between hepatic lipid species and their mechanisms mediating the pathogenesis of NAFLD. We first summarized the interplay between NAFLD and hepatic lipid disturbances. Next, we focused on reviewing the role of saturated fatty acids, cholesterol, oxidized phospholipids, and their respective intermediates in the pathogenesis of NAFLD. The mechanisms by which monounsaturated fatty acids and other pro-resolving mediators exert protective effects are also addressed. Finally, we further discussed the implication of different analysis approaches in lipidomics. Evolving insights into the pathophysiology of NAFLD will provide the opportunity for drug development.
Collapse
Affiliation(s)
- Ke-Qiong Deng
- Department of Cardiology, Huanggang Central Hospital, Huanggang, China
- Huanggang Institute of Translation Medicine, Huanggang, China
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xuewei Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Institute of Model Animal, Wuhan University, Wuhan, China
| | - Fang Lei
- Institute of Model Animal, Wuhan University, Wuhan, China
- School of Basic Medical Science, Wuhan University, Wuhan, China
| | - Xiao-Jing Zhang
- Institute of Model Animal, Wuhan University, Wuhan, China
- School of Basic Medical Science, Wuhan University, Wuhan, China
| | - Peng Zhang
- Institute of Model Animal, Wuhan University, Wuhan, China
- School of Basic Medical Science, Wuhan University, Wuhan, China
| | - Zhi-Gang She
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Institute of Model Animal, Wuhan University, Wuhan, China
| | - Jingjing Cai
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yan-Xiao Ji
- Institute of Model Animal, Wuhan University, Wuhan, China
- School of Basic Medical Science, Wuhan University, Wuhan, China
| | - Hongliang Li
- Huanggang Institute of Translation Medicine, Huanggang, China
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Institute of Model Animal, Wuhan University, Wuhan, China
| |
Collapse
|
17
|
Zhang K, Yuan Y, Dawa Z, Liu F, Yao Y, Wang M, Zhu C, Lin C. Integrating metabolomics and network pharmacology to reveal the mechanisms of Delphinium brunonianum extract against nonalcoholic steatohepatitis. JOURNAL OF ETHNOPHARMACOLOGY 2022; 293:115268. [PMID: 35398502 DOI: 10.1016/j.jep.2022.115268] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 04/04/2022] [Accepted: 04/04/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Herba Delphinii Brunoniani, a Tibetan Material Medica, derived from the aerial parts of Delphinium brunonianum Royle, possesses efficacy of cooling blood to remove apthogentic heat, and dispelling wind to arrest itching, and has been used for the treatment for liver disease according to Tibetan Medicine Theories in Shel Gong Shel Phreng. However, the mechanisms of action remain unclear. AIM OF THE STUDY This work aimed to investigate the efficacy mechanism of Delphinium brunonianum extract (DBE) on nonalcoholic steatohepatitis (NASH), a kind of liver disease by integrating serum metabolomics and network pharmacology analysis. MATERIALS AND METHODS In this study, NASH model mice were established by a high-fat diet. The indexes of lipid accumulation, insulin resistance, and inflammatory reaction were used to evaluate the efficacy of DBE. A combination of UHPLC-QTOF-MS based metabolomics and network pharmacology was established to illustrate the serum biomarkers of NASH mice and to demonstrate the anti-NASH mechanisms of DBE. Serum metabolomics demonstrated potential metabolites and the corresponding metabolic pathways in the efficacy of DBE. Network pharmacology screened the targets of DBE against NASH. Finally, the mechanisms of DBE against NASH were verified by in-vivo pharmacology. RESULTS Metabolomics revealed that DBE significantly regulated the abnormal levels of twenty-two metabolites, which involved the biosynthesis of unsaturated fatty acids and steroid hormone, linoleic acid metabolism, arachidonic acid metabolism, and alpha-Linolenic acid metabolism pathways. Network pharmacology showed that DBE exhibited anti-NASH effects through regulating the targets of PTGS2, PLA2, ALOX5, ALOX15, FASN, and CYP450. Finally, united pharmacological verification result, we found that the mechanisms of DBE against NASH may be related to the regulation of the unsaturated fatty acids biosynthesis and the arachidonic acid metabolism pathway. CONCLUSIONS Integrating serum metabolomic and network analysis, we found that DBE might inhibit the pathological process of NASH by regulating the relative targets and the metabolic pathways, which may be a potential mechanism for the anti-NASH efficacy of DBE. This integrated strategy also provided a rational way for revealing the pharmacodynamic mechanisms of multi-components, multi-targets, and multi-pathways in Traditional Chinese Medicine (TCM).
Collapse
Affiliation(s)
- Kaihui Zhang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China
| | - Yue Yuan
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China
| | - Zeren Dawa
- University of Tibetan Medicine, Lasa, 850000, PR China
| | - Fangle Liu
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China
| | - Yufeng Yao
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China
| | - Meiqi Wang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China.
| | - Chenchen Zhu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China.
| | - Chaozhan Lin
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China.
| |
Collapse
|
18
|
de Jesus RP, de Carvalho JF, de Oliveira LPM, Cunha CDM, Alves TCHS, Vieira STB, Figueiredo VM, Bueno AA. Metabolic and nutritional triggers associated with increased risk of liver complications in SARS-CoV-2. World J Hepatol 2022; 14:80-97. [PMID: 35126841 PMCID: PMC8790394 DOI: 10.4254/wjh.v14.i1.80] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/28/2021] [Accepted: 12/22/2022] [Indexed: 02/06/2023] Open
Abstract
Obesity, diabetes, cardiovascular and respiratory diseases, cancer and smoking are risk factors for negative outcomes in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which can quickly induce severe respiratory failure in 5% of cases. Coronavirus disease-associated liver injury may occur during progression of SARS-CoV-2 in patients with or without pre-existing liver disease, and damage to the liver parenchyma can be caused by infection of hepatocytes. Cirrhosis patients may be particularly vulnerable to SARS-CoV-2 if suffering with cirrhosis-associated immune dysfunction. Furthermore, pharmacotherapies including macrolide or quinolone antibiotics and steroids can also induce liver damage. In this review we addressed nutritional status and nutritional interventions in severe SARS-CoV-2 liver patients. As guidelines for SARS-CoV-2 in intensive care (IC) specifically are not yet available, strategies for management of sepsis and SARS are suggested in SARS-CoV-2. Early enteral nutrition (EN) should be started soon after IC admission, preferably employing iso-osmolar polymeric formula with initial protein content at 0.8 g/kg per day progressively increasing up to 1.3 g/kg per day and enriched with fish oil at 0.1 g/kg per day to 0.2 g/kg per day. Monitoring is necessary to identify signs of intolerance, hemodynamic instability and metabolic disorders, and transition to parenteral nutrition should not be delayed when energy and protein targets cannot be met via EN. Nutrients including vitamins A, C, D, E, B6, B12, folic acid, zinc, selenium and ω-3 fatty acids have in isolation or in combination shown beneficial effects upon immune function and inflammation modulation. Cautious and monitored supplementation up to upper limits may be beneficial in management strategies for SARS-CoV-2 liver patients.
Collapse
Affiliation(s)
- Rosangela Passos de Jesus
- Postgraduate Program in Food, Nutrition and Health at the School of Nutrition of the Federal University of Bahia, Salvador 40.110-150, Bahia, Brazil
| | | | | | - Carla de Magalhães Cunha
- Postgraduate Program in Food, Nutrition and Health at the School of Nutrition of the Federal University of Bahia, Salvador 40.110-150, Bahia, Brazil
| | - Thaisy Cristina Honorato Santos Alves
- Postgraduate Program in Food, Nutrition and Health at the School of Nutrition of the Federal University of Bahia, Salvador 40.110-150, Bahia, Brazil
| | - Sandra Tavares Brito Vieira
- Postgraduate Program in Food, Nutrition and Health at the School of Nutrition of the Federal University of Bahia, Salvador 40.110-150, Bahia, Brazil
| | - Virginia Maria Figueiredo
- Department of Gastroenterology, IPEMED, Ipemed Faculty of Medical Sciences, Salvador 40170-110, Bahia, Brazil
| | - Allain Amador Bueno
- College of Health, Life and Environmental Sciences, University of Worcester, Worcester WR2 6AJ, United Kingdom
| |
Collapse
|
19
|
Lima Rocha JÉ, Mendes Furtado M, Mello Neto RS, da Silva Mendes AV, Brito AKDS, Sena de Almeida JOC, Rodrigues Queiroz EI, de Sousa França JV, Silva Primo MG, Cunha Sales ALDC, Gomes Vasconcelos A, Felix Cabral W, Souza Kückelhaus SA, de Souza de Almeida Leite JR, Fortes Lustosa AKM, Lucarini M, Durazzo A, Arcanjo DDR, Martins MDCDCE. Effects of Fish Oil Supplementation on Oxidative Stress Biomarkers and Liver Damage in Hypercholesterolemic Rats. Nutrients 2022; 14:nu14030426. [PMID: 35276784 PMCID: PMC8839313 DOI: 10.3390/nu14030426] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/13/2022] [Accepted: 01/14/2022] [Indexed: 01/27/2023] Open
Abstract
Metabolic syndrome, especially its component related to dyslipidemia, is related to the development of nonalcoholic fatty liver disease (NAFLD), which is a disease with a significant global prevalence. Supplementation with omega-3 polyunsaturated fatty acids emerged as a complementary therapeutic possibility for dyslipidemia, but its benefits are questioned. This paper aims at evaluating the effects of fish oil supplementation in rats with hypercholesterolemia induced by hypercholesterolemic diet (HD). The study design is based on an experimental model in which the animals were randomly divided into 3 groups: G1 (standard commercial feed + saline solution); G2 (hypercholesterolemic diet + saline solution) and G3 (hypercholesterolemic diet + fish oil) over a period of 16 weeks. Metabolic control parameters and oxidative stress biomarkers were evaluated according to standardized methodologies. The G3 group showed significantly lower values of plasma concentrations of TG, and hepatic myeloperoxidase as well as higher erythrocyte superoxide dismutase activity (p < 0.05). Regarding histopathological analysis, there was lipid accumulation in the liver of animals from group G2; meanwhile, hepatocytes reorganization and expressive reduction of lipid vacuoles and hepatic TG content was observed in group G3. This study demonstrated how fish oil supplementation reduced the plasma concentration and hepatic content of triglycerides, as well as liver tissue damage in histopathological analysis.
Collapse
Affiliation(s)
- Joana Érica Lima Rocha
- Department of Biophysics and Physiology, Federal University of Piauí, Teresina 64049-550, Brazil; (J.É.L.R.); (M.M.F.); (R.S.M.N.); (A.V.d.S.M.); (A.K.d.S.B.); (J.O.C.S.d.A.); (E.I.R.Q.); (J.V.d.S.F.); (M.G.S.P.); (A.L.d.C.C.S.); (D.D.R.A.)
| | - Mariely Mendes Furtado
- Department of Biophysics and Physiology, Federal University of Piauí, Teresina 64049-550, Brazil; (J.É.L.R.); (M.M.F.); (R.S.M.N.); (A.V.d.S.M.); (A.K.d.S.B.); (J.O.C.S.d.A.); (E.I.R.Q.); (J.V.d.S.F.); (M.G.S.P.); (A.L.d.C.C.S.); (D.D.R.A.)
| | - Renato Sampaio Mello Neto
- Department of Biophysics and Physiology, Federal University of Piauí, Teresina 64049-550, Brazil; (J.É.L.R.); (M.M.F.); (R.S.M.N.); (A.V.d.S.M.); (A.K.d.S.B.); (J.O.C.S.d.A.); (E.I.R.Q.); (J.V.d.S.F.); (M.G.S.P.); (A.L.d.C.C.S.); (D.D.R.A.)
| | - Ana Victória da Silva Mendes
- Department of Biophysics and Physiology, Federal University of Piauí, Teresina 64049-550, Brazil; (J.É.L.R.); (M.M.F.); (R.S.M.N.); (A.V.d.S.M.); (A.K.d.S.B.); (J.O.C.S.d.A.); (E.I.R.Q.); (J.V.d.S.F.); (M.G.S.P.); (A.L.d.C.C.S.); (D.D.R.A.)
| | - Ana Karolinne da Silva Brito
- Department of Biophysics and Physiology, Federal University of Piauí, Teresina 64049-550, Brazil; (J.É.L.R.); (M.M.F.); (R.S.M.N.); (A.V.d.S.M.); (A.K.d.S.B.); (J.O.C.S.d.A.); (E.I.R.Q.); (J.V.d.S.F.); (M.G.S.P.); (A.L.d.C.C.S.); (D.D.R.A.)
| | - José Otávio Carvalho Sena de Almeida
- Department of Biophysics and Physiology, Federal University of Piauí, Teresina 64049-550, Brazil; (J.É.L.R.); (M.M.F.); (R.S.M.N.); (A.V.d.S.M.); (A.K.d.S.B.); (J.O.C.S.d.A.); (E.I.R.Q.); (J.V.d.S.F.); (M.G.S.P.); (A.L.d.C.C.S.); (D.D.R.A.)
| | - Emerson Iuri Rodrigues Queiroz
- Department of Biophysics and Physiology, Federal University of Piauí, Teresina 64049-550, Brazil; (J.É.L.R.); (M.M.F.); (R.S.M.N.); (A.V.d.S.M.); (A.K.d.S.B.); (J.O.C.S.d.A.); (E.I.R.Q.); (J.V.d.S.F.); (M.G.S.P.); (A.L.d.C.C.S.); (D.D.R.A.)
| | - José Vinícius de Sousa França
- Department of Biophysics and Physiology, Federal University of Piauí, Teresina 64049-550, Brazil; (J.É.L.R.); (M.M.F.); (R.S.M.N.); (A.V.d.S.M.); (A.K.d.S.B.); (J.O.C.S.d.A.); (E.I.R.Q.); (J.V.d.S.F.); (M.G.S.P.); (A.L.d.C.C.S.); (D.D.R.A.)
| | - Maísa Guimarães Silva Primo
- Department of Biophysics and Physiology, Federal University of Piauí, Teresina 64049-550, Brazil; (J.É.L.R.); (M.M.F.); (R.S.M.N.); (A.V.d.S.M.); (A.K.d.S.B.); (J.O.C.S.d.A.); (E.I.R.Q.); (J.V.d.S.F.); (M.G.S.P.); (A.L.d.C.C.S.); (D.D.R.A.)
| | - Ana Lina de Carvalho Cunha Sales
- Department of Biophysics and Physiology, Federal University of Piauí, Teresina 64049-550, Brazil; (J.É.L.R.); (M.M.F.); (R.S.M.N.); (A.V.d.S.M.); (A.K.d.S.B.); (J.O.C.S.d.A.); (E.I.R.Q.); (J.V.d.S.F.); (M.G.S.P.); (A.L.d.C.C.S.); (D.D.R.A.)
| | - Andreanne Gomes Vasconcelos
- Research Center in Morphology and Applied Immunology, Faculty of Medicine, University of Brasília, Brasília 70910-900, Brazil; (A.G.V.); (W.F.C.); (S.A.S.K.); (J.R.d.S.d.A.L.)
| | - Wanessa Felix Cabral
- Research Center in Morphology and Applied Immunology, Faculty of Medicine, University of Brasília, Brasília 70910-900, Brazil; (A.G.V.); (W.F.C.); (S.A.S.K.); (J.R.d.S.d.A.L.)
| | - Selma Aparecida Souza Kückelhaus
- Research Center in Morphology and Applied Immunology, Faculty of Medicine, University of Brasília, Brasília 70910-900, Brazil; (A.G.V.); (W.F.C.); (S.A.S.K.); (J.R.d.S.d.A.L.)
| | - José Roberto de Souza de Almeida Leite
- Research Center in Morphology and Applied Immunology, Faculty of Medicine, University of Brasília, Brasília 70910-900, Brazil; (A.G.V.); (W.F.C.); (S.A.S.K.); (J.R.d.S.d.A.L.)
| | | | - Massimo Lucarini
- CREA-Research Centre for Food and Nutrition, Via Ardeatina 546, 00178 Rome, Italy; (M.L.); (A.D.)
| | - Alessandra Durazzo
- CREA-Research Centre for Food and Nutrition, Via Ardeatina 546, 00178 Rome, Italy; (M.L.); (A.D.)
| | - Daniel Dias Rufino Arcanjo
- Department of Biophysics and Physiology, Federal University of Piauí, Teresina 64049-550, Brazil; (J.É.L.R.); (M.M.F.); (R.S.M.N.); (A.V.d.S.M.); (A.K.d.S.B.); (J.O.C.S.d.A.); (E.I.R.Q.); (J.V.d.S.F.); (M.G.S.P.); (A.L.d.C.C.S.); (D.D.R.A.)
| | - Maria do Carmo de Carvalho e Martins
- Department of Biophysics and Physiology, Federal University of Piauí, Teresina 64049-550, Brazil; (J.É.L.R.); (M.M.F.); (R.S.M.N.); (A.V.d.S.M.); (A.K.d.S.B.); (J.O.C.S.d.A.); (E.I.R.Q.); (J.V.d.S.F.); (M.G.S.P.); (A.L.d.C.C.S.); (D.D.R.A.)
- Correspondence:
| |
Collapse
|
20
|
Stephen NM, Maradagi T, Kavalappa YP, Sharma H, Ponesakki G. Seafood nutraceuticals: Health benefits and functional properties. RESEARCH AND TECHNOLOGICAL ADVANCES IN FOOD SCIENCE 2022:109-139. [DOI: 10.1016/b978-0-12-824369-5.00012-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
21
|
Lee SM, Kim HK, Lee HB, Kwon OD, Lee EB, Bok JD, Cho CS, Choi YJ, Kang SK. Effects of flaxseed supplementation on omega-6 to omega-3 fatty acid ratio, lipid mediator profile, proinflammatory cytokines and stress indices in laying hens. JOURNAL OF APPLIED ANIMAL RESEARCH 2021. [DOI: 10.1080/09712119.2021.2000416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Sang-Mok Lee
- Department of Agricultural Biotechnology, Seoul National University, Seoul, Republic of Korea
| | - Hee Kyum Kim
- Department of Agricultural Biotechnology, Seoul National University, Seoul, Republic of Korea
| | - Ho-Bin Lee
- Institute of Green-Bio Science & Technology, Seoul National University, Pyeongchang, Republic of Korea
- Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang, Republic of Korea
| | - Oh-Dae Kwon
- Institute of Green-Bio Science & Technology, Seoul National University, Pyeongchang, Republic of Korea
- Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang, Republic of Korea
| | - Eun-Bi Lee
- Institute of Green-Bio Science & Technology, Seoul National University, Pyeongchang, Republic of Korea
- Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang, Republic of Korea
| | - Jin-Duck Bok
- Institute of Green-Bio Science & Technology, Seoul National University, Pyeongchang, Republic of Korea
- Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang, Republic of Korea
| | - Chong-Su Cho
- Department of Agricultural Biotechnology, Seoul National University, Seoul, Republic of Korea
- Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Yun-Jaie Choi
- Department of Agricultural Biotechnology, Seoul National University, Seoul, Republic of Korea
- Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Sang-Kee Kang
- Institute of Green-Bio Science & Technology, Seoul National University, Pyeongchang, Republic of Korea
- Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang, Republic of Korea
- Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
22
|
The Interplay between Insulin Resistance, Inflammation, Oxidative Stress, Base Excision Repair and Metabolic Syndrome in Nonalcoholic Fatty Liver Disease. Int J Mol Sci 2021; 22:ijms222011128. [PMID: 34681787 PMCID: PMC8537238 DOI: 10.3390/ijms222011128] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/11/2021] [Accepted: 10/12/2021] [Indexed: 02/06/2023] Open
Abstract
One of the most common chronic liver disorders, affecting mainly people in Western countries, is nonalcoholic fatty liver disease (NAFLD). Unfortunately, its pathophysiological mechanism is not fully understood, and no dedicated treatment is available. Simple steatosis can lead to nonalcoholic steatohepatitis and even to fibrosis, cancer, and cirrhosis of the liver. NAFLD very often occurs in parallel with type 2 diabetes mellitus and in obese people. Furthermore, it is much more likely to develop in patients with metabolic syndrome (MS), whose criteria include abdominal obesity, elevated blood triacylglycerol level, reduced high-density lipoprotein cholesterol level, increased blood pressure, and high fasting glucose. An important phenomenon in MS is also insulin resistance (IR), which is very common in NAFLD. Liver IR and NAFLD development are linked through an interaction between the accumulation of free fatty acids, hepatic inflammation, and increased oxidative stress. The liver is particularly exposed to elevated levels of reactive oxygen species due to a large number of mitochondria in hepatocytes. In these organelles, the main DNA repair pathway is base excision repair (BER). The present article will illustrate how impairment of BER may be related to the development of NAFLD.
Collapse
|
23
|
Di Pasqua LG, Berardo C, Cagna M, Mannucci B, Milanesi G, Croce AC, Ferrigno A, Vairetti M. Long-term cold storage preservation does not affect fatty livers from rats fed with a methionine and choline deficient diet. Lipids Health Dis 2021; 20:78. [PMID: 34320998 PMCID: PMC8317281 DOI: 10.1186/s12944-021-01503-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 07/13/2021] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Waiting lists that continue to grow and the lack of organs available for transplantation necessitate the use of marginal livers, such as fatty livers. Since steatotic livers are more susceptible to damage from ischemia and reperfusion, it was investigated whether fatty livers with different lipidomic profiles show a different outcome when subjected to long-term cold storage preservation. METHODS Eight-week-old male Wistar rats fed for 2 weeks by a methionine-choline-deficient (MCD) diet or control diet were employed in this study. Livers were preserved in a University of Wisconsin (UW) solution at 4 °C for 6, 12 or 24 h and, after washout, reperfused for 2 h with a Krebs-Henseleit buffer at 37 °C. Hepatic enzyme release, bile production, O2-uptake, and portal venous pressure (PVP) were evaluated. The liver fatty acid profile was evaluated by a gas chromatography-mass spectrometry (GC/MS). RESULTS MCD rats showed higher LDH and AST levels with respect to the control group. When comparing MCD livers preserved for 6, 12 or 24 h, no differences in enzyme release were found during both the washout or the reperfusion period. The same trend occurred for O2-uptake, PVP, and bile flow. A general decrease in SFA and MUFA, except for oleic acid, and a decrease in PUFA, except for arachidonic, eicosadienoic, and docosahexanaeoic acids, were found in MCD rats when compared with control rats. Moreover, the ratio between SFA and the various types of unsaturated fatty acids (UFA) was significantly lower in MCD rats. CONCLUSIONS Although prolonged cold ischemia negatively affects the graft outcome, our data suggest that the quality of lipid constituents could influence liver injury during cold storage: the lack of an increased hepatic injury in MCD may be justified by low SFA, which likely reduces the deleterious tendency toward lipid crystallization occurring under cold ischemia.
Collapse
Affiliation(s)
| | - Clarissa Berardo
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100, Pavia, Italy.
| | - Marta Cagna
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100, Pavia, Italy
| | | | - Gloria Milanesi
- Department of Biology and Biotechnology "Lazzaro Spallanzani", University of Pavia, Pavia, Italy
| | - Anna Cleta Croce
- Institute of Molecular Genetics, Italian National Research Council (CNR), Pavia, Italy
| | - Andrea Ferrigno
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100, Pavia, Italy.
| | - Mariapia Vairetti
- Department of Internal Medicine and Therapeutics, University of Pavia, 27100, Pavia, Italy
| |
Collapse
|
24
|
Warner DR, Warner JB, Hardesty JE, Song YL, Chen CY, Chen Z, Kang JX, McClain CJ, Kirpich IA. Beneficial effects of an endogenous enrichment in n3-PUFAs on Wnt signaling are associated with attenuation of alcohol-mediated liver disease in mice. FASEB J 2021; 35:e21377. [PMID: 33481293 DOI: 10.1096/fj.202001202r] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 12/30/2020] [Accepted: 01/04/2021] [Indexed: 12/13/2022]
Abstract
Alcohol-associated liver disease (ALD) is a major human health issue for which there are limited treatment options. Experimental evidence suggests that nutrition plays an important role in ALD pathogenesis, and specific dietary fatty acids, for example, n6 or n3-PUFAs, may exacerbate or attenuate ALD, respectively. The purpose of the current study was to determine whether the beneficial effects of n3-PUFA enrichment in ALD were mediated, in part, by improvement in Wnt signaling. Wild-type (WT) and fat-1 transgenic mice (that endogenously convert n6-PUFAs to n3) were fed ethanol (EtOH) for 6 weeks followed by a single LPS challenge. fat-1 mice had less severe liver damage than WT littermates as evidenced by reduced plasma alanine aminotransferase, hepatic steatosis, liver tissue neutrophil infiltration, and pro-inflammatory cytokine expression. WT mice had a greater downregulation of Axin2, a key gene in the Wnt pathway, than fat-1 mice in response to EtOH and LPS. Further, there were significant differences between WT and fat-1 EtOH+LPS-challenged mice in the expression of five additional genes linked to the Wnt signaling pathway, including Apc, Fosl1/Fra-1, Mapk8/Jnk-1, Porcn, and Nkd1. Compared to WT, primary hepatocytes isolated from fat-1 mice exhibited more effective Wnt signaling and were more resistant to EtOH-, palmitic acid-, or TNFα-induced cell death. Further, we demonstrated that the n3-PUFA-derived lipid mediators, resolvins D1 and E1, can regulate hepatocyte expression of several Wnt-related genes that were differentially expressed between WT and fat-1 mice. These data demonstrate a novel mechanism by which n3-PUFAs can ameliorate ALD.
Collapse
Affiliation(s)
- Dennis R Warner
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, Louisville, KY, USA
| | - Jeffrey B Warner
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, Louisville, KY, USA.,Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Josiah E Hardesty
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, Louisville, KY, USA.,Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Ying L Song
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, Louisville, KY, USA
| | - Chi-Yu Chen
- Laboratory for Lipid Medicine and Technology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Zoe Chen
- Laboratory for Lipid Medicine and Technology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Jing X Kang
- Laboratory for Lipid Medicine and Technology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Craig J McClain
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, Louisville, KY, USA.,Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA.,University of Louisville Alcohol Center, University of Louisville School of Medicine, Louisville, KY, USA.,Hepatobiology & Toxicology Center, University of Louisville School of Medicine, Louisville, KY, USA.,Robley Rex Veterans Medical Center, Louisville, KY, USA
| | - Irina A Kirpich
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, Louisville, KY, USA.,Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA.,University of Louisville Alcohol Center, University of Louisville School of Medicine, Louisville, KY, USA.,Hepatobiology & Toxicology Center, University of Louisville School of Medicine, Louisville, KY, USA
| |
Collapse
|
25
|
Fousekis FS, Mitselos IV, Christodoulou DK. New insights into intestinal failure-associated liver disease in adults: A comprehensive review of the literature. Saudi J Gastroenterol 2021; 27:3-12. [PMID: 33642350 PMCID: PMC8083246 DOI: 10.4103/sjg.sjg_551_20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Intestinal failure-associated liver disease (IFALD) remains one of the most common and serious complications of parenteral nutrition (PN), causing a wide spectrum of hepatic manifestations from steatosis and mild cholestasis to portal hypertension and end-stage liver failure. The prevalence of IFALD depends on the diagnostic criteria and ranges from 4.3% to 65%. Moreover, many factors are shown to contribute to its development, including nutrient deficiencies, toxicity of PN, infections, and alterations of bile acid metabolism and gut microbiota. Prevention and management of IFALD aim at ameliorating or eliminating the risk factors associated with IFALD. The use of PN formulations with a lower ratio omega-6-to-omega-3 polyunsaturated fatty acids, cycle PN, optimization of enteral stimulation and prevention and early treatment of infections constitute the main therapeutic targets. However, failure of improvement and severe IFALD with end-stage liver failure should be considered as the indications of intestinal transplantation. The aim of this review is to provide an update of the epidemiology, pathophysiology, and diagnosis of IFALD in the adult population as well as to present a clinical approach of the therapeutic strategies of IFALD and present novel therapeutic targets.
Collapse
Affiliation(s)
- Fotios S. Fousekis
- Department of Gastroenterology and Hepatology, School of Health Sciences, University Hospital of Ioannina, Faculty of Medicine, University of Ioannina, Ioannina, Greece
| | - Ioannis V. Mitselos
- Department of Gastroenterology and Hepatology, School of Health Sciences, University Hospital of Ioannina, Faculty of Medicine, University of Ioannina, Ioannina, Greece
| | - Dimitrios K. Christodoulou
- Department of Gastroenterology and Hepatology, School of Health Sciences, University Hospital of Ioannina, Faculty of Medicine, University of Ioannina, Ioannina, Greece,Address for correspondence: Prof. Dimitrios K. Christodoulou, Professor of Gastroenterology, Department of Gastroenterology and Hepatology, School of Health Sciences, University Hospital of Ioannina, Faculty of Medicine, University of Ioannina, POBox 1186, Ioannina 45110, Greece. E-mail:
| |
Collapse
|
26
|
Rozner R, Vernikov J, Griess-Fishheimer S, Travinsky T, Penn S, Schwartz B, Mesilati-Stahy R, Argov-Argaman N, Shahar R, Monsonego-Ornan E. The Role of Omega-3 Polyunsaturated Fatty Acids from Different Sources in Bone Development. Nutrients 2020; 12:nu12113494. [PMID: 33202985 PMCID: PMC7697266 DOI: 10.3390/nu12113494] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 11/05/2020] [Accepted: 11/10/2020] [Indexed: 01/01/2023] Open
Abstract
N-3 polyunsaturated fatty acids (PUFAs) are essential nutrients that must be obtained from the diet. We have previously showed that endogenous n-3 PUFAs contribute to skeletal development and bone quality in fat-1 mice. Unlike other mammals, these transgenic mice, carry the n-3 desaturase gene and thus can convert n-6 to n-3 PUFAs endogenously. Since this model does not mimic dietary exposure to n-3 PUFAs, diets rich in fish and flaxseed oils were used to further elucidate the role of n-3 PUFAs in bone development. Our investigation reveals that dietary n-3 PUFAs decrease fat accumulation in the liver, lower serum fat levels, and alter fatty acid (FA) content in liver and serum. Bone analyses show that n-3 PUFAs improve mechanical properties, which were measured using a three-point bending test, but exert complex effects on bone structure that vary according to its source. In a micro-CT analysis, we found that the flaxseed oil diet improves trabecular bone micro-architecture, whereas the fish oil diet promotes higher bone mineral density (BMD) with no effect on trabecular bone. The transcriptome characterization of bone by RNA-seq identified regulatory mechanisms of n-3 PUFAs via modulation of the cell cycle and peripheral circadian rhythm genes. These results extend our knowledge and provide insights into the molecular mechanisms of bone remodeling regulation induced by different sources of dietary n-3 PUFAs.
Collapse
Affiliation(s)
- Reut Rozner
- The Robert H. Smith Faculty of Agriculture, Food and Environment, Institute of Biochemistry and Nutrition, The Hebrew University of Jerusalem, Rehovot 7610001, Israel; (R.R.); (J.V.); (S.G.-F.); (T.T.); (S.P.); (B.S.)
| | - Janna Vernikov
- The Robert H. Smith Faculty of Agriculture, Food and Environment, Institute of Biochemistry and Nutrition, The Hebrew University of Jerusalem, Rehovot 7610001, Israel; (R.R.); (J.V.); (S.G.-F.); (T.T.); (S.P.); (B.S.)
| | - Shelley Griess-Fishheimer
- The Robert H. Smith Faculty of Agriculture, Food and Environment, Institute of Biochemistry and Nutrition, The Hebrew University of Jerusalem, Rehovot 7610001, Israel; (R.R.); (J.V.); (S.G.-F.); (T.T.); (S.P.); (B.S.)
| | - Tamar Travinsky
- The Robert H. Smith Faculty of Agriculture, Food and Environment, Institute of Biochemistry and Nutrition, The Hebrew University of Jerusalem, Rehovot 7610001, Israel; (R.R.); (J.V.); (S.G.-F.); (T.T.); (S.P.); (B.S.)
| | - Svetlana Penn
- The Robert H. Smith Faculty of Agriculture, Food and Environment, Institute of Biochemistry and Nutrition, The Hebrew University of Jerusalem, Rehovot 7610001, Israel; (R.R.); (J.V.); (S.G.-F.); (T.T.); (S.P.); (B.S.)
| | - Betty Schwartz
- The Robert H. Smith Faculty of Agriculture, Food and Environment, Institute of Biochemistry and Nutrition, The Hebrew University of Jerusalem, Rehovot 7610001, Israel; (R.R.); (J.V.); (S.G.-F.); (T.T.); (S.P.); (B.S.)
| | - Ronit Mesilati-Stahy
- Animal Science, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 7610001, Israel; (R.M.-S.); (N.A.-A.)
| | - Nurit Argov-Argaman
- Animal Science, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 7610001, Israel; (R.M.-S.); (N.A.-A.)
| | - Ron Shahar
- The Robert H. Smith Faculty of Agriculture, Food and Environment, Koret School of Veterinary Medicine, The Hebrew University of Jerusalem, Rehovot 7610001, Israel;
| | - Efrat Monsonego-Ornan
- The Robert H. Smith Faculty of Agriculture, Food and Environment, Institute of Biochemistry and Nutrition, The Hebrew University of Jerusalem, Rehovot 7610001, Israel; (R.R.); (J.V.); (S.G.-F.); (T.T.); (S.P.); (B.S.)
- Correspondence:
| |
Collapse
|
27
|
DHA Supplementation Attenuates MI-Induced LV Matrix Remodeling and Dysfunction in Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:7606938. [PMID: 32832005 PMCID: PMC7424392 DOI: 10.1155/2020/7606938] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 03/24/2020] [Accepted: 04/07/2020] [Indexed: 01/12/2023]
Abstract
Objective Myocardial ischemia and reperfusion (I/R) injury is associated with oxidative stress and inflammation, leading to scar development and malfunction. The marine omega-3 fatty acids (ω-3 FA), eicosapentaenoic acid (EPA), and docosahexaenoic acid (DHA) are mediating cardioprotection and improving clinical outcomes in patients with heart disease. Therefore, we tested the hypothesis that docosahexaenoic acid (DHA) supplementation prior to LAD occlusion-induced myocardial injury (MI) confers cardioprotection in mice. Methods C57BL/6N mice were placed on DHA or control diets (CD) beginning 7 d prior to 60 min LAD occlusion-induced MI or sham surgery. The expression of inflammatory mediators was measured via RT-qPCR. Besides FACS analysis for macrophage quantification and subtype evaluation, macrophage accumulation as well as collagen deposition was quantified in histological sections. Cardiac function was assessed using a pressure-volume catheter for up to 14 d. Results DHA supplementation significantly attenuated the induction of peroxisome proliferator-activated receptor-α (PPAR-α) (2.3 ± 0.4 CD vs. 1.4 ± 0.3 DHA) after LAD occlusion. Furthermore, TNF-α (4.0 ± 0.6 CD vs. 1.5 ± 0.2 DHA), IL-1β (60.7 ± 7.0 CD vs. 11.6 ± 1.9 DHA), and IL-10 (223.8 ± 62.1 CD vs. 135.5 ± 38.5 DHA) mRNA expression increase was diminished in DHA-supplemented mice after 72 h reperfusion. These changes were accompanied by a less prominent switch in α/β myosin heavy chain isoforms. Chemokine mRNA expression was stronger initiated (CCL2 6 h: 32.8 ± 11.5 CD vs. 78.8 ± 13.6 DHA) but terminated earlier (CCL2 72 h: 39.5 ± 7.8 CD vs. 8.2 ± 1.9 DHA; CCL3 72 h: 794.3 ± 270.9 CD vs. 258.2 ± 57.8 DHA) in DHA supplementation compared to CD mice after LAD occlusion. Correspondingly, DHA supplementation was associated with a stronger increase of predominantly alternatively activated Ly6C-positive macrophage phenotype, being associated with less collagen deposition and better LV function (EF 14 d: 17.6 ± 2.6 CD vs. 31.4 ± 1.5 DHA). Conclusion Our data indicate that DHA supplementation mediates cardioprotection from MI via modulation of the inflammatory response with timely and attenuated remodeling. DHA seems to attenuate MI-induced cardiomyocyte injury partly by transient PPAR-α downregulation, diminishing the need for antioxidant mechanisms including mitochondrial function, or α- to β-MHC isoform switch.
Collapse
|
28
|
Agarwal P, Combes TW, Shojaee-Moradie F, Fielding B, Gordon S, Mizrahi V, Martinez FO. Foam Cells Control Mycobacterium tuberculosis Infection. Front Microbiol 2020; 11:1394. [PMID: 32754123 PMCID: PMC7381311 DOI: 10.3389/fmicb.2020.01394] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 05/29/2020] [Indexed: 12/27/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb) infects macrophages and macrophage-derived foam cells, a hallmark of granulomata in tuberculous lesions. We analyzed the effects of lipid accumulation in human primary macrophages and quantified strong triglyceride and phospholipid remodeling which depended on the dietary fatty acid used for the assay. The enrichment of >70% in triglyceride and phospholipids can alter cell membrane properties, signaling and phagocytosis in macrophages. In conventional macrophage cultures, cells are heterogeneous, small or large macrophages. In foam cells, a third population of 30% of cells with increased granularity can be detected. We found that foam cell formation is heterogenous and that lipid accumulation and foam cell formation reduces the phagocytosis of Mtb. Under the conditions tested, cell death was highly prevalent in macrophages, whereas foam cells were largely protected from this effect. Foam cells also supported slower Mtb replication, yet this had no discernible impact on the intracellular efficacy of four different antitubercular drugs. Foam cell formation had a significant impact in the inflammatory potential of the cells. TNF-α, IL-1β, and prototypical chemokines were increased. The ratio of inflammatory IL-1β, TNF-α, and IL-6 vs. anti-inflammatory IL-10 was significantly higher in response to Mtb vs. LPS, and was increased in foam cells compared to macrophages, suggestive of increased pro-inflammatory properties. Cytokine production correlated with NF-κB activation in our models. We conclude that foam cell formation reduces the host cell avidity for, and phagocytosis of, Mtb while protecting the cells from death. This protective effect is associated with enhanced inflammatory potential of foam cells and restricted intracellular growth of Mtb.
Collapse
Affiliation(s)
- Pooja Agarwal
- South African Medical Research Council/National Health Laboratory Service/University of Cape Town, Molecular Mycobacteriology Research Unit, Division of Medical Microbiology, Department of Pathology, Department of Science and Innovation/National Research Foundation, Centre of Excellence for Biomedical TB Research and Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Theo W Combes
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | | | - Barbara Fielding
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Siamon Gordon
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan City, Taiwan.,Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Valerie Mizrahi
- South African Medical Research Council/National Health Laboratory Service/University of Cape Town, Molecular Mycobacteriology Research Unit, Division of Medical Microbiology, Department of Pathology, Department of Science and Innovation/National Research Foundation, Centre of Excellence for Biomedical TB Research and Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Fernando O Martinez
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| |
Collapse
|
29
|
Khadge S, Sharp JG, Thiele GM, McGuire TR, Talmadge JE. Fatty Acid Mediators in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1259:125-153. [PMID: 32578175 DOI: 10.1007/978-3-030-43093-1_8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Patients with cancer frequently overexpress inflammatory cytokines with an associated neutrophilia both of which may be downregulated by diets with high omega-3 polyunsaturated fatty acids (ω-3 PUFA). The anti-inflammatory activity of dietary ω-3 PUFA has been suggested to have anticancer properties and to improve survival of cancer patients. Currently, the majority of dietary research efforts do not differentiate between obesity and dietary fatty acid consumption as mediators of inflammatory cell expansion and tumor microenvironmental infiltration, initiation, and progression. In this chapter, we discuss the relationships between dietary lipids, inflammation, neoplasia and strategies to regulate these relationships. We posit that dietary composition, notably the ratio of ω-3 vs. ω-6 PUFA, regulates tumor initiation and progression and the frequency and sites of metastasis that, together, impact overall survival (OS). We focus on three broad topics: first, the role of dietary lipids in chronic inflammation and tumor initiation, progression, and regression; second, lipid mediators linking inflammation and cancer; and third, dietary lipid regulation of murine and human tumor initiation, progression, and metastasis.
Collapse
Affiliation(s)
- Saraswoti Khadge
- Department of Pathology and Microbiology and Immunology, University of Nebraska Medical Center, Omaha, NE, USA.,Vanderbilt University, Nashville, TN, USA
| | - John Graham Sharp
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Geoffrey M Thiele
- Department of Pathology and Microbiology and Immunology, University of Nebraska Medical Center, Omaha, NE, USA.,Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA.,Veteran Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Timothy R McGuire
- Department of Pharmacy Practice, University of Nebraska Medical Center, Omaha, NE, USA
| | - James E Talmadge
- Department of Pathology and Microbiology and Immunology, University of Nebraska Medical Center, Omaha, NE, USA. .,Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
30
|
Soluble Epoxide Hydrolase Inhibition in Liver Diseases: A Review of Current Research and Knowledge Gaps. BIOLOGY 2020; 9:biology9060124. [PMID: 32545637 PMCID: PMC7345757 DOI: 10.3390/biology9060124] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/08/2020] [Accepted: 06/10/2020] [Indexed: 12/14/2022]
Abstract
Emerging evidence suggests that soluble epoxide hydrolase (sEH) inhibition is a valuable therapeutic strategy for the treatment of numerous diseases, including those of the liver. sEH rapidly degrades cytochrome P450-produced epoxygenated lipids (epoxy-fatty acids), which are synthesized from omega-3 and omega-6 polyunsaturated fatty acids, that generally exert beneficial effects on several cellular processes. sEH hydrolysis of epoxy-fatty acids produces dihydroxy-fatty acids which are typically less biologically active than their parent epoxide. Efforts to develop sEH inhibitors have made available numerous compounds that show therapeutic efficacy and a wide margin of safety in a variety of different diseases, including non-alcoholic fatty liver disease, liver fibrosis, portal hypertension, and others. This review summarizes research efforts which characterize the applications, underlying effects, and molecular mechanisms of sEH inhibitors in these liver diseases and identifies gaps in knowledge for future research.
Collapse
|
31
|
Madnawat H, Welu AL, Gilbert EJ, Taylor DB, Jain S, Manithody C, Blomenkamp K, Jain AK. Mechanisms of Parenteral Nutrition-Associated Liver and Gut Injury. Nutr Clin Pract 2019; 35:63-71. [PMID: 31872510 DOI: 10.1002/ncp.10461] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Parenteral nutrition (PN) has revolutionized the care of patients with intestinal failure by providing nutrition intravenously. Worldwide, PN remains a standard tool of nutrition delivery in neonatal, pediatric, and adult patients. Though the benefits are evident, patients receiving PN can suffer serious cholestasis due to lack of enteral feeding and sometimes have fatal complications from liver injury and gut atrophy, including PN-associated liver disease or intestinal failure-associated liver disease. Recent studies into gut-systemic cross talk via the bile acid-regulated farnesoid X receptor (FXR)-fibroblast growth factor 19 (FGF19) axis, gut microbial control of the TGR5-glucagon-like peptide (GLP) axis, sepsis, and role of prematurity of hepatobiliary receptors are greatly broadening our understanding of PN-associated injury. It has also been shown that the composition of ω-6/ω-3 polyunsaturated fatty acids given parenterally as lipid emulsions can variably drive damage to hepatocytes and cell integrity. This manuscript reviews the mechanisms for the multifactorial pathogenesis of liver disease and gut injury with PN and discusses novel ameliorative strategies.
Collapse
Affiliation(s)
- Himani Madnawat
- Department of Pediatrics, St. Louis University School of Medicine, Cardinal Glennon Children's Medical Center, St. Louis, Missouri, USA
| | - Adam L Welu
- Department of Pediatrics, St. Louis University School of Medicine, Cardinal Glennon Children's Medical Center, St. Louis, Missouri, USA
| | - Ester J Gilbert
- Department of Pediatrics, St. Louis University School of Medicine, Cardinal Glennon Children's Medical Center, St. Louis, Missouri, USA
| | - Derian B Taylor
- Department of Pediatrics, St. Louis University School of Medicine, Cardinal Glennon Children's Medical Center, St. Louis, Missouri, USA
| | - Sonali Jain
- Department of Pediatrics, St. Louis University School of Medicine, Cardinal Glennon Children's Medical Center, St. Louis, Missouri, USA
| | - Chandrashekhara Manithody
- Department of Pediatrics, St. Louis University School of Medicine, Cardinal Glennon Children's Medical Center, St. Louis, Missouri, USA
| | - Keith Blomenkamp
- Department of Pediatrics, St. Louis University School of Medicine, Cardinal Glennon Children's Medical Center, St. Louis, Missouri, USA
| | - Ajay K Jain
- Department of Pediatrics, St. Louis University School of Medicine, Cardinal Glennon Children's Medical Center, St. Louis, Missouri, USA
| |
Collapse
|
32
|
Hao H, Fu M, Yan R, He B, Li M, Liu Q, Cai Y, Zhang X, Huang R. Chemical composition and immunostimulatory properties of green alga Caulerpa racemosa var peltata. FOOD AGR IMMUNOL 2019. [DOI: 10.1080/09540105.2019.1646216] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Affiliation(s)
- Huili Hao
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou, People’s Republic of China
| | - Manqin Fu
- Sericultural & Agri-Food Research Institute Guangdong Academy of Agricultural Sciences/Key Laboratory of Functional Foods, Ministry of Agriculture/Guangdong Key Laboratory of Agricultural Products Processing, Guangzhou, People’s Republic of China
| | - Ru Yan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, People’s Republic of China
| | - Baolin He
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou, People’s Republic of China
| | - Meiying Li
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou, People’s Republic of China
| | - Qiabiao Liu
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou, People’s Republic of China
| | - Yimian Cai
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou, People’s Republic of China
| | - Xiaoyong Zhang
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, People’s Republic of China
| | - Riming Huang
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou, People’s Republic of China
| |
Collapse
|
33
|
de Bus I, Witkamp R, Zuilhof H, Albada B, Balvers M. The role of n-3 PUFA-derived fatty acid derivatives and their oxygenated metabolites in the modulation of inflammation. Prostaglandins Other Lipid Mediat 2019; 144:106351. [PMID: 31260750 DOI: 10.1016/j.prostaglandins.2019.106351] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 06/27/2019] [Indexed: 12/14/2022]
Abstract
Notwithstanding the ongoing debate on their full potential in health and disease, there is general consensus that n-3 PUFAs play important physiological roles. Increasing dietary n-3 PUFA intake results in increased DHA and EPA content in cell membranes as well as an increase in n-3 derived oxylipin and -endocannabinoid concentrations, like fatty acid amides and glycerol-esters. These shifts are believed to (partly) explain the pharmacological and anti-inflammatory effects of n-3 PUFAs. Recent studies discovered that n-3 PUFA-derived endocannabinoids can be further metabolized by the oxidative enzymes CYP-450, LOX and COX, similar to the n-6 derived endocannabinoids. Interestingly, these oxidized n-3 PUFA derived endocannabinoids of eicosapentaenoyl ethanolamide (EPEA) and docosahexaenoyl ethanolamide (DHEA) have higher anti-inflammatory and anti-proliferative potential than their precursors. In this review, an overview of recently discovered n-3 PUFA derived endocannabinoids and their metabolites is provided. In addition, the use of chemical probes will be presented as a promising technique to study the n-3 PUFA and n-3 PUFA metabolism within the field of lipid biochemistry.
Collapse
Affiliation(s)
- Ian de Bus
- Nutrition and Pharmacology Group, Division of Human Nutrition, Wageningen University & Research, Stippeneng 4, 6708 WE, Wageningen, the Netherlands; Laboratory of Organic Chemistry, Wageningen University & Research, Stippeneng 4, 6708 WE, Wageningen, the Netherlands
| | - Renger Witkamp
- Nutrition and Pharmacology Group, Division of Human Nutrition, Wageningen University & Research, Stippeneng 4, 6708 WE, Wageningen, the Netherlands
| | - Han Zuilhof
- Laboratory of Organic Chemistry, Wageningen University & Research, Stippeneng 4, 6708 WE, Wageningen, the Netherlands; School of Pharmaceutical Sciences and Technology, Tianjin University, 92 Weijin Road, Tianjin, PR China; Department of Chemical and Materials Engineering, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Bauke Albada
- Laboratory of Organic Chemistry, Wageningen University & Research, Stippeneng 4, 6708 WE, Wageningen, the Netherlands.
| | - Michiel Balvers
- Nutrition and Pharmacology Group, Division of Human Nutrition, Wageningen University & Research, Stippeneng 4, 6708 WE, Wageningen, the Netherlands.
| |
Collapse
|
34
|
Unsaturated fatty acids from flaxseed oil and exercise modulate GPR120 but not GPR40 in the liver of obese mice: a new anti-inflammatory approach. J Nutr Biochem 2018; 66:52-62. [PMID: 30771734 DOI: 10.1016/j.jnutbio.2018.12.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 09/19/2018] [Accepted: 12/05/2018] [Indexed: 12/18/2022]
Abstract
GPR120 and GPR40 were recently reported as omega-3 (ω3) receptors with anti-inflammatory properties. Physical exercise could increase the expression of these receptors in the liver, improving hepatic metabolism in obesity and type 2 diabetes. Our aim was to investigate GPR120/40 in the liver of lean and obese mice after acute or chronic physical exercise, with or without the supplementation of ω3 rich flaxseed oil (FS), as well as assess the impact of exercise and FS on insulin signaling and inflammation. Mice were fed a high-fat diet (HF) for 4 weeks to induce obesity and subsequently subjected to exercise with or without FS, or FS alone. Insulin signaling, inflammatory markers and GPR120/40 and related cascades were measured. Chronic, but not acute, exercise and FS increased GPR120, but not GPR40, activating β-arrestin-2 and decreasing the inflammatory response, as well as reducing fat depots in liver and adipose tissue. Exercise or a source of ω3 led to a higher tolerance to fatigue and an increased running distance and speed. The combination of physical exercise and ω3 food sources could provide a new strategy against obesity through the modulation of hepatic GPR120 and an increase in exercise performance.
Collapse
|
35
|
Immune regulation and anti-cancer activity by lipid inflammatory mediators. Int Immunopharmacol 2018; 65:580-592. [PMID: 30447537 DOI: 10.1016/j.intimp.2018.10.026] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 10/02/2018] [Accepted: 10/17/2018] [Indexed: 12/19/2022]
Abstract
Rodent and clinical studies have documented that myeloid cell infiltration of tumors is associated with poor outcomes, neutrophilia and lymphocytopenia. This contrasts with increased lymphocyte infiltration of tumors, which is correlated with improved outcomes. Lifestyle parameters, such as obesity and diets with high levels of saturated fat and/or omega (ω)-6 polyunsaturated fatty acids (PUFAs), can influence these inflammatory parameters, including an increase in extramedullary myelopoiesis (EMM). While tumor secretion of growth factors (GFs) and chemokines regulate tumor-immune-cell crosstalk, lifestyle choices also contribute to inflammation, abnormal pathology and leukocyte infiltration of tumors. A relationship between obesity and high-fat diets (notably saturated fats in Western diets) and inflammation, tumor incidence, metastasis and poor outcomes is generally accepted. However, the mechanisms of dietary promotion of an inflammatory microenvironment and targeted drugs to inhibit the clinical sequelae are poorly understood. Thus, modifications of obesity and dietary fat may provide preventative or therapeutic approaches to control tumor-associated inflammation and disease progression. Currently, the majority of basic and clinical research does not differentiate between obesity and fatty acid consumption as mediators of inflammatory and neoplastic processes. In this review, we discuss the relationships between dietary PUFAs, inflammation and neoplasia and experimental strategies to improve our understanding of these relationships. We conclude that dietary composition, notably the ratio of ω-3 vs ω-6 PUFA regulates tumor growth and the frequency and sites of metastasis that together, impact overall survival (OS) in mice.
Collapse
|
36
|
Liu L, Hu Q, Wu H, Wang X, Gao C, Chen G, Yao P, Gong Z. Dietary DHA/EPA Ratio Changes Fatty Acid Composition and Attenuates Diet-Induced Accumulation of Lipid in the Liver of ApoE -/- Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:6256802. [PMID: 30538803 PMCID: PMC6261399 DOI: 10.1155/2018/6256802] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 09/25/2018] [Indexed: 02/07/2023]
Abstract
Diets containing various docosahexaenoic acid (DHA)/eicosapentaenoic acid (EPA) ratios protect against liver damage in mice fed with a high-fat diet (HFD). However, it is unclear whether these beneficial roles of DHA and EPA are associated with alterations of fatty acid (FA) composition in the liver. This study evaluated the positive impacts of n-6/n-3 polyunsaturated fatty acids (PUFAs) containing different DHA/EPA ratios on HFD-induced liver disease and alterations of the hepatic FA composition. ApoE-/- mice were fed with HFDs with various ratios of DHA/EPA (2 : 1, 1 : 1, and 1 : 2) and an n-6/n-3 ratio of 4 : 1 for 12 weeks. After treatment, the serum and hepatic FA compositions, serum biochemical parameters, liver injury, and hepatic lipid metabolism-related gene expression were determined. Our results demonstrated that dietary DHA/EPA changed serum and hepatic FA composition by increasing contents of n-6 and n-3 PUFAs and decreasing amounts of monounsaturated fatty acids (MUFAs) and the n-6/n-3 ratio. Among the three DHA/EPA groups, the DHA/EPA 2 : 1 group tended to raise n-3 PUFAs concentration and lower the n-6/n-3 ratio in the liver, whereas DHA/EPA 1 : 2 tended to raise n-6 PUFAs concentration and improve the n-6/n-3 ratio. DHA/EPA supplementation reduced the hepatic impairment of lipid homeostasis, oxidative stress, and the inflammatory responses in HFD-fed mice. The DHA/EPA 2 : 1 group had lower serum levels of total cholesterol, triglycerides, and low-density lipoprotein cholesterol and higher levels of adiponectin than HFD group. The DHA/EPA 1 : 2 group had elevated serum levels of aspartate aminotransferase, alanine aminotransferase, and alkaline phosphatase, without significant change the expression of genes for inflammation or hepatic lipid metabolism among the three DHA/EPA groups. The results suggest that DHA/EPA-enriched diet with an n-6/n-3 ratio of 4 : 1 may reverse HFD-induced nonalcoholic fatty liver disease to some extent by increasing n-6 and n-3 PUFAs and decreasing the amount of MUFAs and the n-6/n-3 ratio.
Collapse
Affiliation(s)
- Liang Liu
- Key Laboratory for Deep Processing of Major Grain and Oil, Ministry of Education, Wuhan 430023, China
- College of Food Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, China
- Hubei Key Laboratory for Processing and Transformation of Agricultural Products (Wuhan Polytechnic University), Wuhan 430023, China
| | - Qinling Hu
- Key Laboratory for Deep Processing of Major Grain and Oil, Ministry of Education, Wuhan 430023, China
- College of Food Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Huihui Wu
- Key Laboratory for Deep Processing of Major Grain and Oil, Ministry of Education, Wuhan 430023, China
- College of Food Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Xiujing Wang
- Key Laboratory for Deep Processing of Major Grain and Oil, Ministry of Education, Wuhan 430023, China
- College of Food Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Chao Gao
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Guoxun Chen
- Department of Nutrition, University of Tennessee at Knoxville, Knoxville 37996, USA
| | - Ping Yao
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhiyong Gong
- Key Laboratory for Deep Processing of Major Grain and Oil, Ministry of Education, Wuhan 430023, China
- College of Food Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, China
- Hubei Key Laboratory for Processing and Transformation of Agricultural Products (Wuhan Polytechnic University), Wuhan 430023, China
| |
Collapse
|
37
|
Khadge S, Thiele GM, Sharp JG, McGuire TR, Klassen LW, Black PN, DiRusso CC, Cook L, Talmadge JE. Long-chain omega-3 polyunsaturated fatty acids decrease mammary tumor growth, multiorgan metastasis and enhance survival. Clin Exp Metastasis 2018; 35:797-818. [PMID: 30327985 DOI: 10.1007/s10585-018-9941-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 10/04/2018] [Indexed: 02/07/2023]
Abstract
Epidemiological studies show a reduced risk of breast cancer (BC) in women consuming high levels of long-chain (LC) omega-3 (ω-3) fatty acids (FAs) compared with women who consumed low levels. However, the regulatory and mechanistic roles of dietary ω-6 and LC-ω-3 FAs on tumor progression, metastasis and survival are poorly understood. Female BALB/c mice (10-week old) were pair-fed with a diet containing ω-3 or an isocaloric, isolipidic ω-6 diet for 16 weeks prior to the orthotopic implantation of 4T1 mammary tumor cells. Major outcomes studied included: mammary tumor growth, survival analysis, and metastases analyses in multiple organs including pulmonary, hepatic, bone, cardiac, renal, ovarian, and contralateral MG (CMG). The dietary regulation of the tumor microenvironment was evaluated in mice autopsied on day-35 post tumor injection. In mice fed the ω-3 containing diet, there was a significant delay in tumor initiation and prolonged survival relative to the ω-6 diet-fed group. The tumor size on day 35 post tumor injection in the ω-3 group was 50% smaller and the frequencies of pulmonary and bone metastases were significantly lower relative to the ω-6 group. Similarly, the incidence/frequencies and/or size of cardiac, renal, ovarian metastases were significantly lower in mice fed the ω-3 diet. The analyses of the tumor microenvironment showed that tumors in the ω-3 group had significantly lower numbers of proliferating tumor cells (Ki67+)/high power field (HPF), and higher numbers of apoptotic tumor cells (TUNEL+)/HPF, lower neo-vascularization (CD31+ vessels/HPF), infiltration by neutrophil elastase+ cells, and macrophages (F4/80+) relative to the tumors from the ω-6 group. Further, in tumors from the ω-3 diet-fed mice, T-cell infiltration was 102% higher resulting in a neutrophil to T-lymphocyte ratio (NLR) that was 76% lower (p < 0.05). Direct correlations were observed between NLR with tumor size and T-cell infiltration with the number of apoptotic tumor cells. qRT-PCR analysis revealed that tumor IL10 mRNA levels were significantly higher (six-fold) in the tumors from mice fed the ω-3 diet and inversely correlated with the tumor size. Our data suggest that dietary LC-ω-3FAs modulates the mammary tumor microenvironment slowing tumor growth, and reducing metastases to both common and less preferential organs resulting in prolonged survival. The surrogate analyses undertaken support a mechanism of action by dietary LC-ω-3FAs that includes, but is not limited to decreased infiltration by myeloid cells (neutrophils and macrophages), an increase in CD3+ lymphocyte infiltration and IL10 associated anti-inflammatory activity.
Collapse
Affiliation(s)
- Saraswoti Khadge
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, 68198-6495, USA
| | - Geoffrey M Thiele
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, 68198-6495, USA.,Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198-6495, USA.,Veteran Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - John Graham Sharp
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Timothy R McGuire
- Department of Pharmacy Practice, University of Nebraska Medical Center, Omaha, NE, USA
| | - Lynell W Klassen
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198-6495, USA.,Veteran Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Paul N Black
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Concetta C DiRusso
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Leah Cook
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, 68198-6495, USA
| | - James E Talmadge
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, 68198-6495, USA. .,Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198-6495, USA.
| |
Collapse
|
38
|
Kühn G, Pallauf K, Schulz C, Rimbach G. Flavonoids as putative modulators of Δ4-, Δ5-, and Δ6-desaturases: Studies in cultured hepatocytes, myocytes, and adipocytes. Biofactors 2018; 44:485-495. [PMID: 30365230 DOI: 10.1002/biof.1443] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 06/07/2018] [Accepted: 06/27/2018] [Indexed: 12/19/2022]
Abstract
This study was conducted to screen flavonoids for affecting expression of desaturases involved in omega-3 fatty acid synthesis and ceramide (CER) metabolism. To this end, cultured HepG2 hepatocytes, C2C12 myocytes, and 3T3-L1 adipocytes were treated with nontoxic concentrations of 12 selected flavonoids and expression of Δ4-, Δ5-, and Δ6-desaturases (DEGS1, FADS1, and FADS2, respectively) was determined. The flavonoids tested were more cytotoxic to HepG2 and 3T3-L1 than to C2C12 cells. In HepG2 cells, FADS1 was induced by quercetin and FADS2 expression was increased by daidzein, genistein, and pratensein treatment. DEGS1 was increased by apigenin, luteolin, orobol, and quercetin administration. In differentiated C2C12 cells, substances had no inducing effect or even lowered target gene expression. Pratensein induced both FADS1 and FADS2 in differentiated 3T3-L1 cells and DEGS1 was increased by treatment with apigenin, genistein, luteolin, orobol, and quercetin. In conclusion, pratensein may be an interesting test compound for further studies in vitro and in vivo on omega-3 synthesis since it induces its rate-limiting enzyme FADS2. Apigenin, luteolin, orobol, and quercetin induced DEGS1 and thereby possibly synthesis of proapoptotic CER in malignant HepG2 cells and 3T3-L1. In contrast, in benign C2C12 cells, they did not elevate mRNA steady state levels of DEGS1. That may partly explain the higher resistance of C2C12 cells against flavonoids compared to the other cell lines. By affecting tumor cells and nontumor cells differently, these flavonoids may be promising substances for further research regarding anticancer properties. © 2018 BioFactors, 44(5):485-495, 2018.
Collapse
Affiliation(s)
- Gianna Kühn
- Institute of Human Nutrition and Food Science, University of Kiel, Kiel, Germany
| | - Kathrin Pallauf
- Institute of Human Nutrition and Food Science, University of Kiel, Kiel, Germany
| | - Carsten Schulz
- Institute of Animal Breeding and Husbandry, University of Kiel, Kiel, Germany
- GMA-Gesellschaft für Marine Aquakultur mbH, Büsum, Germany
| | - Gerald Rimbach
- Institute of Human Nutrition and Food Science, University of Kiel, Kiel, Germany
| |
Collapse
|
39
|
Khadge S, Thiele GM, Sharp JG, McGuire TR, Klassen LW, Black PN, DiRusso CC, Talmadge JE. Long-Chain Omega-3 Polyunsaturated Fatty Acids Modulate Mammary Gland Composition and Inflammation. J Mammary Gland Biol Neoplasia 2018; 23:43-58. [PMID: 29574638 DOI: 10.1007/s10911-018-9391-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 03/19/2018] [Indexed: 01/07/2023] Open
Abstract
Studies in rodents have shown that dietary modifications as mammary glands (MG) develop, regulates susceptibility to mammary tumor initiation. However, the effects of dietary PUFA composition on MGs in adult life, remains poorly understood. This study investigated morphological alterations and inflammatory microenvironments in the MGs of adult mice fed isocaloric and isolipidic liquid diets with varying compositions of omega (ω)-6 and long-chain (Lc)-ω3FA that were pair-fed. Despite similar consumption levels of the diets, mice fed the ω-3 diet had significantly lower body-weight gains, and abdominal-fat and mammary fat pad (MFP) weights. Fatty acid analysis showed significantly higher levels of Lc-ω-3FAs in the MFPs of mice on the ω-3 diet, while in the MFPs from the ω-6 group, Lc-ω-3FAs were undetectable. Our study revealed that MGs from ω-3 group had a significantly lower ductal end-point density, branching density, an absence of ductal sprouts, a thinner ductal stroma, fewer proliferating epithelial cells and a lower transcription levels of estrogen receptor 1 and amphiregulin. An analysis of the MFP and abdominal-fat showed significantly smaller adipocytes in the ω-3 group, which was accompanied by lower transcription levels of leptin, IGF1, and IGF1R. Further, MFPs from the ω-3 group had significantly decreased numbers and sizes of crown-like-structures (CLS), F4/80+ macrophages and decreased expression of proinflammatory mediators including Ptgs2, IL6, CCL2, TNFα, NFκB, and IFNγ. Together, these results support dietary Lc-ω-3FA regulation of MG structure and density and adipose tissue inflammation with the potential for dietary Lc-ω-3FA to decrease the risk of mammary gland tumor formation.
Collapse
Affiliation(s)
- Saraswoti Khadge
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Geoffrey M Thiele
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
- Veteran Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - John Graham Sharp
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Timothy R McGuire
- Department of Pharmacy Practice, University of Nebraska Medical Center, Omaha, NE, USA
| | - Lynell W Klassen
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
- Veteran Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Paul N Black
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Concetta C DiRusso
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - James E Talmadge
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA.
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|