1
|
Wang Z, Dai Q, Luo H, Han X, Feng Q, Cao X. Nano-vibration exciter: Hypoxia-inducible factor 1 signaling pathway-mediated extracellular vesicles as bioactive glass substitutes for bone regeneration. Bioact Mater 2024; 40:460-473. [PMID: 39036347 PMCID: PMC11259761 DOI: 10.1016/j.bioactmat.2024.06.023] [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: 03/25/2024] [Revised: 06/11/2024] [Accepted: 06/13/2024] [Indexed: 07/23/2024] Open
Abstract
Bioactive glasses (BG) play a vital role in angiogenesis and osteogenesis through releasing functional ions. However, the rapid ion release in the early stage will cause excessive accumulation of metal ions, which in turn leads to obvious cytotoxicity, long-term inflammation, and bone repair failure. Inspired by the vibration exciter, small extracellular vesicles (sEVs) obtained by treating mesenchymal stem cells with copper-doped bioactive glass (CuBG-sEVs), is prepared as a nano-vibration exciter. The nano-vibration exciter can convert the ion signals of CuBG into biochemical factor signals through hypoxia-inducible factor 1 (HIF-1) signaling pathway and its activated autophagy, so as to better exert the osteogenic activity of BG. The results showed that CuBG extracts could significantly improve the enrichment of key miRNAs and increase the yield of CuBG-sEVs by activating HIF-1 signaling pathway and its activated autophagy. Cell experiments showed that CuBG-sEVs are favor to cell recruitment, vascularization and osteogenesis as the enrichment of key miRNAs. The animal experiments results showed that CuBG-sEVs stimulated angiogenesis mediated by CD31 and promoted bone regeneration by activating signaling pathways related to osteogenesis. These findings underscored the significant potential of sEVs as alternative strategies to better roles of BG.
Collapse
Affiliation(s)
- Zetao Wang
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, PR China
- National Engineering Research Centre for Tissue Restoration and Reconstruction, Guangzhou, 510006, PR China
| | - Qiyuan Dai
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, PR China
- National Engineering Research Centre for Tissue Restoration and Reconstruction, Guangzhou, 510006, PR China
| | - Huitong Luo
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, PR China
- National Engineering Research Centre for Tissue Restoration and Reconstruction, Guangzhou, 510006, PR China
| | - Xiyuan Han
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, PR China
- National Engineering Research Centre for Tissue Restoration and Reconstruction, Guangzhou, 510006, PR China
| | - Qi Feng
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, PR China
- National Engineering Research Centre for Tissue Restoration and Reconstruction, Guangzhou, 510006, PR China
| | - Xiaodong Cao
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, PR China
- National Engineering Research Centre for Tissue Restoration and Reconstruction, Guangzhou, 510006, PR China
- Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, 510006, PR China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, PR China
- Zhongshan Institute of Modern Industrial Technology of SCUT, Zhongshan, Guangdong, 528437, PR China
| |
Collapse
|
2
|
Li X, Liu C, Zhang R, Li Y, Ye D, Wang H, He M, Sun Y. Biosynthetic deficiency of docosahexaenoic acid causes nonalcoholic fatty liver disease and ferroptosis-mediated hepatocyte injury. J Biol Chem 2024; 300:107405. [PMID: 38788853 PMCID: PMC11231757 DOI: 10.1016/j.jbc.2024.107405] [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: 11/03/2023] [Revised: 05/07/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
Exogenous omega-3 fatty acids, particularly docosahexaenoic acid (DHA), have shown to exert beneficial effects on nonalcoholic fatty liver disease (NAFLD), which is characterized by the excessive accumulation of lipids and chronic injury in the liver. However, the effect of endogenous DHA biosynthesis on the lipid homeostasis of liver is poorly understood. In this study, we used a DHA biosynthesis-deficient zebrafish model, elovl2 mutant, to explore the effect of endogenously biosynthesized DHA on hepatic lipid homeostasis. We found the pathways of lipogenesis and lipid uptake were strongly activated, while the pathways of lipid oxidation and lipid transport were inhibited in the liver of elovl2 mutants, leading to lipid droplet accumulation in the mutant hepatocytes and NAFLD. Furthermore, the elovl2 mutant hepatocytes exhibited disrupted mitochondrial structure and function, activated endoplasmic reticulum stress, and hepatic injury. We further unveiled that the hepatic cell death and injury was mainly mediated by ferroptosis, rather than apoptosis, in elovl2 mutants. Elevating DHA content in elovl2 mutants, either by the introduction of an omega-3 desaturase (fat1) transgene or by feeding with a DHA-rich diet, could strongly alleviate NAFLD features and ferroptosis-mediated hepatic injury. Together, our study elucidates the essential role of endogenous DHA biosynthesis in maintaining hepatic lipid homeostasis and liver health, highlighting that DHA deficiency can lead to NAFLD and ferroptosis-mediated hepatic injury.
Collapse
Affiliation(s)
- Xuehui Li
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Chengjie Liu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Ru Zhang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Yi Li
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Ding Ye
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Houpeng Wang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Mudan He
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Yonghua Sun
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Hubei Hongshan Laboratory, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China; The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan, China.
| |
Collapse
|
3
|
Colombo C, Lanfranchi C, Tosetti G, Corti F, Primignani M. Management of liver disease and portal hypertension in cystic fibrosis: a review. Expert Rev Respir Med 2024; 18:269-281. [PMID: 38962827 DOI: 10.1080/17476348.2024.2365842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 06/05/2024] [Indexed: 07/05/2024]
Abstract
INTRODUCTION Cystic fibrosis (CF)-associated liver disease can significantly affect the quality of life and survival of people with CF. The hepatobiliary manifestations in CF are various, with focal/multilobular biliary cirrhosis more common in children and porto-sinusoidal vascular disease (PSVD) in young adults. Portal hypertensive complications, particularly bleeding from esophagogastric varices and hypersplenism are common, while liver failure is rarer and mainly linked to biliary disease. AREAS COVERED This review explores current therapeutic options for CF-associated liver disease, presenting ongoing studies and new insights into parthenogenesis for potential future therapies. EXPERT OPINION Monitoring for signs of portal hypertension is essential. Limited evidence supports ursodeoxycholic acid (UDCA) efficacy in halting CF liver disease progression. The effect of cystic fibrosis transmembrane conductance regulator (CFTR) modulators on liver outcomes lacks definitive data, since patients with CF-related liver disease were excluded from trials due to potential hepatotoxicity. A proposed approach involves using UDCA and modulators in early stages, along with anti-inflammatory agents, with further therapeutic strategies awaiting randomized trials. Prevention of portal hypertensive bleeding includes endoscopic sclerotherapy or ligation of esophageal varices. Nonselective beta-blockers may also prevent bleeding and could be cautiously implemented. Other non-etiological treatments require investigation.
Collapse
Affiliation(s)
- Carla Colombo
- Cystic Fibrosis Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Chiara Lanfranchi
- Cystic Fibrosis Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Giulia Tosetti
- Division of Gastroenterology and Hepatology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Fabiola Corti
- Cystic Fibrosis Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Massimo Primignani
- Division of Gastroenterology and Hepatology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
4
|
Rezaei K, Bejoy AM, Rotarescu RD, Klievik BJ, Metherel AH. Sex-dependent differences in tissue and blood n-3 PUFA levels following ALA or ALA + DHA feeding of liver-specific Elovl2-KO and control mice. Prostaglandins Leukot Essent Fatty Acids 2024; 201:102621. [PMID: 38763083 DOI: 10.1016/j.plefa.2024.102621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 05/13/2024] [Accepted: 05/13/2024] [Indexed: 05/21/2024]
Abstract
Docosahexaenoic acid (DHA, 22:6n-3) must be consumed from the diet or synthesized from polyunsaturated fatty acid (PUFA) precursors, such as α-linolenic acid (ALA, 18:3n-3). Elongase 2 (encoded by Elovl2 gene) catalyzes two elongation reactions in the PUFA biosynthesis pathway and may be important in regulating the observed sex differences in n-3 PUFA levels. Our aim was to determine how targeted knockout of liver Elovl2 affects tissue and blood n-3 PUFA levels in male and female C57BL/6J mice. Twenty-eight-day old male and female liver Elovl2-KO and control mice were placed onto one of two dietary protocols for a total of 8 weeks (4-8 mice per genotype, per diet, per sex): 1) an 8-week 2 % ALA in total fat diet or 2) a 4-week 2 % ALA diet followed by a 4-week 2 % ALA + 2 % DHA diet. Following this 8-week feeding period, 12-week-old mice were sacrificed and serum, red blood cells (RBC), liver, heart and brain were collected and fatty acid levels measured. Significant interaction effects (p < 0.05, sex x genotype) for serum, RBC, liver and heart DHA levels were identified. In serum and liver, DHA levels were significantly different (p < 0.01) between all groups with male controls > female controls > female KO > male KO in serum and female controls > male controls > female KO > male KO in liver. In RBCs and the heart, female controls = male controls > female KO > male KO (p < 0.001). The addition of DHA to diet removed the interaction effects on DHA levels in the serum, liver and heart, yielding a significant sex effect in serum, liver (female > male, p < 0.01) and brain (male > female, p < 0.05) and genotype effect in serum and heart (control > KO, p < 0.05). Ablation of liver Elovl2 results in significantly lower blood and tissue DHA in a sex-dependent manner, suggesting a role for Elovl2 on sex differences in n-3 PUFA levels.
Collapse
Affiliation(s)
- Kuorosh Rezaei
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Ashley M Bejoy
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Ruxandra D Rotarescu
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Brinley J Klievik
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Adam H Metherel
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
5
|
Siapoush S, Rezaei R, Alavifard H, Hatami B, Zali MR, Vosough M, Lorzadeh S, Łos MJ, Baghaei K, Ghavami S. Therapeutic implications of targeting autophagy and TGF-β crosstalk for the treatment of liver fibrosis. Life Sci 2023; 329:121894. [PMID: 37380126 DOI: 10.1016/j.lfs.2023.121894] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 06/19/2023] [Accepted: 06/25/2023] [Indexed: 06/30/2023]
Abstract
Liver fibrosis is characterized by the excessive deposition and accumulation of extracellular matrix components, mainly collagens, and occurs in response to a broad spectrum of triggers with different etiologies. Under stress conditions, autophagy serves as a highly conserved homeostatic system for cell survival and is importantly involved in various biological processes. Transforming growth factor-β1 (TGF-β1) has emerged as a central cytokine in hepatic stellate cell (HSC) activation and is the main mediator of liver fibrosis. A growing body of evidence from preclinical and clinical studies suggests that TGF-β1 regulates autophagy, a process that affects various essential (patho)physiological aspects related to liver fibrosis. This review comprehensively highlights recent advances in our understanding of cellular and molecular mechanisms of autophagy, its regulation by TGF-β, and the implication of autophagy in the pathogenesis of progressive liver disorders. Moreover, we evaluated crosstalk between autophagy and TGF-β1 signalling and discussed whether simultaneous inhibition of these pathways could represent a novel approach to improve the efficacy of anti-fibrotic therapy in the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Samaneh Siapoush
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ramazan Rezaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Helia Alavifard
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Behzad Hatami
- Gastroenterology and Liver Diseases Research center, Research institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research center, Research institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Shahrokh Lorzadeh
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Marek J Łos
- Biotechnology Center, Silesian University of Technology, 8 Krzywousty St., 44-100 Gliwice, Poland; Autophagy Research Center, Department of Biochemistry; Shiraz University of Medical Sciences, Shiraz, Iran; LinkoCare Life Sciences AB, Linkoping, Sweden
| | - Kaveh Baghaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Gastroenterology and Liver Diseases Research center, Research institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada; Faculty of Medicine in Zabrze, University of Technology in Katowice, 41-800 Zabrze, Poland; Research Institute of Oncology and Hematology, Cancer Care Manitoba-University of Manitoba, Winnipeg, Manitoba, Canada; Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, Manitoba, Canada.
| |
Collapse
|
6
|
Yi Q, Yang J, Wu Y, Wang Y, Cao Q, Wen W. Immune microenvironment changes of liver cirrhosis: emerging role of mesenchymal stromal cells. Front Immunol 2023; 14:1204524. [PMID: 37539053 PMCID: PMC10395751 DOI: 10.3389/fimmu.2023.1204524] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 06/21/2023] [Indexed: 08/05/2023] Open
Abstract
Cirrhosis is a progressive and diffuse liver disease characterized by liver tissue fibrosis and impaired liver function. This condition is brought about by several factors, including chronic hepatitis, hepatic steatosis, alcohol abuse, and other immunological injuries. The pathogenesis of liver cirrhosis is a complex process that involves the interaction of various immune cells and cytokines, which work together to create the hepatic homeostasis imbalance in the liver. Some studies have indicated that alterations in the immune microenvironment of liver cirrhosis are closely linked to the development and prognosis of the disease. The noteworthy function of mesenchymal stem cells and their paracrine secretion lies in their ability to promote the production of cytokines, which in turn enhance the self-repairing capabilities of tissues. The objective of this review is to provide a summary of the alterations in liver homeostasis and to discuss intercellular communication within the organ. Recent research on MSCs is yielding a blueprint for cell typing and biomarker immunoregulation. Hopefully, as MSCs researches continue to progress, novel therapeutic approaches will emerge to address cirrhosis.
Collapse
Affiliation(s)
- Qiuyun Yi
- National Center for Liver Cancer, Third Affiliated Hospital of Naval Medical University, Shanghai, China
- International Cooperation Laboratory on Signal Transduction, Third Affiliated Hospital of Naval Medical University (Second Military Medical University), Shanghai, China
| | - Jinxian Yang
- National Center for Liver Cancer, Third Affiliated Hospital of Naval Medical University, Shanghai, China
- International Cooperation Laboratory on Signal Transduction, Third Affiliated Hospital of Naval Medical University (Second Military Medical University), Shanghai, China
| | - Ying Wu
- Department of Breast and Thyroid Surgery, Changhai Hospital, Naval Military Medical University, Shanghai, China
| | - Ying Wang
- Department of Laboratory Diagnosis, Third Affiliated Hospital of Naval Medical University (Second Military Medical University), Shanghai, China
| | - Qiqi Cao
- National Center for Liver Cancer, Third Affiliated Hospital of Naval Medical University, Shanghai, China
- International Cooperation Laboratory on Signal Transduction, Third Affiliated Hospital of Naval Medical University (Second Military Medical University), Shanghai, China
| | - Wen Wen
- National Center for Liver Cancer, Third Affiliated Hospital of Naval Medical University, Shanghai, China
- Department of Laboratory Diagnosis, Third Affiliated Hospital of Naval Medical University (Second Military Medical University), Shanghai, China
| |
Collapse
|
7
|
Gao Y, Zheng B, Xu S, Zhao Z, Liu W, Wang T, Yuan M, Sun X, Tan Y, Xu Q, Wu X. Mitochondrial folate metabolism-mediated α-linolenic acid exhaustion masks liver fibrosis resolution. J Biol Chem 2023:104909. [PMID: 37307917 PMCID: PMC10344950 DOI: 10.1016/j.jbc.2023.104909] [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: 12/21/2022] [Revised: 05/02/2023] [Accepted: 06/01/2023] [Indexed: 06/14/2023] Open
Abstract
Sustainable TGF-β1 signaling drives organ fibrogenesis. However, the cellular adaptation to maintain TGF-β1 signaling remains unclear. In this study, we revealed that dietary folate restriction promoted the resolution of liver fibrosis in mice with nonalcoholic steatohepatitis (NASH). In activated hepatic stellate cells (HSCs), folate shifted toward mitochondrial metabolism to sustain TGF-β1 signaling. Mechanistically, nontargeted metabolomics screening identified that α-linolenic acid (ALA) is exhausted by mitochondrial folate metabolism in activated HSCs. Knocking down serine hydroxymethyltransferase 2 (SHMT2) increases the bioconversion of ALA to docosahexaenoic acid (DHA) which inhibits TGF-β1 signaling. Finally, blocking mitochondrial folate metabolism promoted liver fibrosis resolution in NASH mice. In conclusion, mitochondrial folate metabolism/ALA exhaustion/TGF-βR1 reproduction is a feedforward signaling to sustain profibrotic TGF-β1 signaling and targeting mitochondrial folate metabolism is a promising strategy to enforce liver fibrosis resolution.
Collapse
Affiliation(s)
- Yanjie Gao
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Bingfeng Zheng
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Shuaiqi Xu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Zhibo Zhao
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Wanyue Liu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Tingyu Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Manman Yuan
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Xueqing Sun
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Yang Tan
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210023, China
| | - Qiang Xu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210023, China.
| | - Xingxin Wu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu 210023, China.
| |
Collapse
|
8
|
Liu YC, Limbu SM, Wang JG, Wang M, Chen LQ, Qiao F, Luo Y, Zhang ML, Du ZY. Dietary docosahexaenoic acid reduces fat deposition and alleviates liver damage induced by D-galactosamine and lipopolysaccharides in Nile tilapia (Oreochromis niloticus). Comp Biochem Physiol C Toxicol Pharmacol 2023; 268:109603. [PMID: 36906246 DOI: 10.1016/j.cbpc.2023.109603] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 02/13/2023] [Accepted: 03/06/2023] [Indexed: 03/11/2023]
Abstract
Liver health is important to maintain survival and growth of fish. Currently, the role of dietary docosahexaenoic acid (DHA) in improving fish liver health is largely unknown. This study investigated the role of DHA supplementation in fat deposition and liver damage caused by D-galactosamine (D-GalN) and lipopolysaccharides (LPS) in Nile tilapia (Oreochromis niloticus). Four diets were formulated as control diet (Con), Con supplemented with 1 % DHA, 2 % DHA and 4 % DHA diets, respectively. The diets were fed to 25 Nile tilapia (2.0 ± 0.1 g, average initial weight) in triplicates for four weeks. After the four weeks, 20 fish in each treatment were randomly selected and injected with a mixture of 500 mg D-GalN and 10 μL LPS per mL to induce acute liver injury. The results showed that the Nile tilapia fed on DHA diets decreased visceral somatic index, liver lipid content and serum and liver triglyceride concentrations than those fed on the Con diet. Moreover, after D-GalN/LPS injection, the fish fed on DHA diets decreased alanine aminotransferase and aspartate transaminase activities in the serum. The results of liver qPCR and transcriptomics assays together showed that the DHA diets feeding improved liver health by downregulating the expression of the genes related to toll-like receptor 4 (TLR4) signaling pathway, inflammation and apoptosis. This study indicates that DHA supplementation in Nile tilapia alleviates the liver damage caused by D-GalN/LPS through increasing lipid catabolism, decreasing lipogenesis, TLR4 signaling pathway, inflammation, and apoptosis. Our study provides novel knowledge on the role of DHA in improving liver health in cultured aquatic animals for sustainable aquaculture.
Collapse
Affiliation(s)
- Yi-Chan Liu
- LANEH, School of Life Sciences, East China Normal University, Shanghai, PR China
| | - Samwel Mchele Limbu
- Department of Aquaculture Technology, School of Aquatic Sciences and Fisheries Technology, University of Dar es Salaam, P. O. Box 60091, Dar es Salaam, Tanzania; UDSM-ECNU Joint Research Center for Aquaculture and Fish Biology (JRCAFB), Dar es Salaam, Tanzania
| | - Jin-Gang Wang
- LANEH, School of Life Sciences, East China Normal University, Shanghai, PR China
| | - Mai Wang
- LANEH, School of Life Sciences, East China Normal University, Shanghai, PR China
| | - Li-Qiao Chen
- LANEH, School of Life Sciences, East China Normal University, Shanghai, PR China
| | - Fang Qiao
- LANEH, School of Life Sciences, East China Normal University, Shanghai, PR China
| | - Yuan Luo
- LANEH, School of Life Sciences, East China Normal University, Shanghai, PR China
| | - Mei-Ling Zhang
- LANEH, School of Life Sciences, East China Normal University, Shanghai, PR China
| | - Zhen-Yu Du
- LANEH, School of Life Sciences, East China Normal University, Shanghai, PR China; ECNU-UDSM Joint Research Center for Aquaculture and Fish Biology (JRCAFB), Shanghai, PR China.
| |
Collapse
|
9
|
Lacalle-Bergeron L, Izquierdo-Sandoval D, Fernández-Quintela A, Portillo MP, Sancho JV, Hernández F, Portolés T. LC-IMS-HRMS for identification of biomarkers in untargeted metabolomics: The effects of pterostilbene and resveratrol consumption in liver steatosis, animal model. Food Res Int 2023; 165:112376. [PMID: 36869462 DOI: 10.1016/j.foodres.2022.112376] [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: 09/14/2022] [Revised: 12/12/2022] [Accepted: 12/24/2022] [Indexed: 01/03/2023]
Abstract
Untargeted metabolomics with the combination of ion mobility separation coupled to high resolution mass spectrometry (IMS-HRMS) was applied to investigate the impact of resveratrol and pterostilbene supplementation on the metabolic fingerprint of the Wistar rats liver with induced liver steatosis. RP-LC and HILIC in both ionisation modes were employed to analyse the liver samples (n = 40) from Wistar rats fed with a high-fat and high-fructose diet, supplemented or not with resveratrol and pterostilbene. After univariate and multivariate statistical analysis, 34 metabolites were highlighted in the different diets and elucidated. Despite the structural similarity, different alterations in liver metabolism were observed by the supplementations. Resveratrol treatment was characterised by the alteration in metabolism of 17 lysophospholipids, while pterostilbene affected some vitamins and derivatives, among others. IMS has demonstrated great potential in the elucidation process thanks to the additional structural descriptor the CCS (Å2), providing more confidence in the identification.
Collapse
Affiliation(s)
- Leticia Lacalle-Bergeron
- Enviromental and Public Health Analytical Chemistry, Research Institute for Pesticides and Water (IUPA), Universitat Jaume I, Av. Sos Baynat S/N, 12071 Castellón de la Plana, Spain
| | - David Izquierdo-Sandoval
- Enviromental and Public Health Analytical Chemistry, Research Institute for Pesticides and Water (IUPA), Universitat Jaume I, Av. Sos Baynat S/N, 12071 Castellón de la Plana, Spain
| | - Alfredo Fernández-Quintela
- Nutrition and Obesity Group, Department of Nutrition and Food Science, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Lucio Lascaray Research Centre, 01006 Vitoria-Gasteiz, Spain; BIOARABA Institute of Health, 01009 Vitoria-Gasteiz, Spain; CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos III (ISCIII), 01006 Vitoria-Gasteiz, Spain
| | - María P Portillo
- Nutrition and Obesity Group, Department of Nutrition and Food Science, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Lucio Lascaray Research Centre, 01006 Vitoria-Gasteiz, Spain; BIOARABA Institute of Health, 01009 Vitoria-Gasteiz, Spain; CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos III (ISCIII), 01006 Vitoria-Gasteiz, Spain.
| | - Juan Vicente Sancho
- Enviromental and Public Health Analytical Chemistry, Research Institute for Pesticides and Water (IUPA), Universitat Jaume I, Av. Sos Baynat S/N, 12071 Castellón de la Plana, Spain
| | - Félix Hernández
- Enviromental and Public Health Analytical Chemistry, Research Institute for Pesticides and Water (IUPA), Universitat Jaume I, Av. Sos Baynat S/N, 12071 Castellón de la Plana, Spain
| | - Tania Portolés
- Enviromental and Public Health Analytical Chemistry, Research Institute for Pesticides and Water (IUPA), Universitat Jaume I, Av. Sos Baynat S/N, 12071 Castellón de la Plana, Spain.
| |
Collapse
|
10
|
Wang Y, Li J, do Vale GD, Chaudhary J, Anwar A, McDonald JG, Qin T, Zhang H, Corbin IR. Repeated trans-arterial treatments of LDL-DHA nanoparticles induce multiple pathways of tumor cell death in hepatocellular carcinoma bearing rats. Front Oncol 2022; 12:1052221. [PMID: 36505796 PMCID: PMC9730405 DOI: 10.3389/fonc.2022.1052221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 11/07/2022] [Indexed: 11/27/2022] Open
Abstract
Introduction Repeated hepatic arterial delivery of therapeutic agents to the liver by percutaneously implanted port-catheter systems has been widely used to treat unresectable liver cancer. This approach is applied to assess the therapeutic efficacy of repeated low-density lipoprotein-docosahexaenoic acid (LDL-DHA) nanoparticle treatments in a rat model of hepatocellular carcinoma. Methods N1S1 hepatoma bearing rats underwent placement of a percutaneously implanted hepatic artery port-catheter system and were allocated to untreated, control LDL-triolein (LDL-TO) or LDL-DHA nanoparticle infusions groups. Treatments were performed every three days over a nine day study period. MRI was performed at baseline and throughout the study. At the end of the study tissue samples were collected for analyses. Results and Discussion Implantation of the port catheters was successful in all rats. MRI showed that repeated infusions of LDL-DHA nanoparticles significantly impaired the growth of the rat hepatomas eventually leading to tumor regression. The tumors in the LDL-TO treated group showed delayed growth, while the untreated tumors grew steadily throughout the study. Histopathology and MRI support these findings demonstrating extensive tumor necrosis in LDL-DHA treated groups while the control groups displayed minor necrosis. Molecular and biochemical analyses also revealed that LDL-DHA treated tumors had increased levels of nuclear factor-kappa B and lipid peroxidation and depletion of glutathione peroxidase 4 relative to the control groups. Evidence of both ferroptosis and apoptosis tumor cell death was observed following LDL-DHA treatments. In conclusion repeated transarterial infusions of LDL-DHA nanoparticles provides sustained repression of tumor growth in a rat hepatoma model.
Collapse
Affiliation(s)
- Yuzhu Wang
- Department of Hepatobiliary and Pancreatic Surgery, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, Henan, China
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, United States
| | - Junjie Li
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, United States
| | - Goncalo Dias do Vale
- Center for Human Nutrition and Department of Molecular Genetics, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, United States
| | - Jaideep Chaudhary
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, United States
| | - Arnida Anwar
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, United States
| | - Jeffrey G. McDonald
- Center for Human Nutrition and Department of Molecular Genetics, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, United States
| | - Tao Qin
- Department of Hepatobiliary and Pancreatic Surgery, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, Henan, China
| | - Hongwei Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, Henan, China
| | - Ian R. Corbin
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, United States
- Internal Medicine Division of Liver and Digestive Diseases, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, United States
- Radiology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, United States
| |
Collapse
|
11
|
Ontoria-Oviedo I, Amaro-Prellezo E, Castellano D, Venegas-Venegas E, González-Santos F, Ruiz-Saurí A, Pelacho B, Prósper F, Pérez del Caz MD, Sepúlveda P. Topical Administration of a Marine Oil Rich in Pro-Resolving Lipid Mediators Accelerates Wound Healing in Diabetic db/db Mice through Angiogenesis and Macrophage Polarization. Int J Mol Sci 2022; 23:ijms23179918. [PMID: 36077316 PMCID: PMC9456080 DOI: 10.3390/ijms23179918] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/09/2022] [Accepted: 08/29/2022] [Indexed: 11/16/2022] Open
Abstract
Impaired wound healing in patients with type 2 diabetes (DM2) is characterized by chronic inflammation, which delays wound closure. Specialized pro-resolving lipid mediators (SPMs) are bioactive molecules produced from essential polyunsaturated fatty acids (PUFAs), principally omega-3 docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA). SPMs are potent regulators of inflammation and have been used to suppress chronic inflammation in peripheral artery disease, non-alcoholic fatty liver disease, and central nervous system syndromes. LIPINOVA® is a commercially available safe-grade nutritional supplement made from a fractionated marine lipid concentrate derived from anchovy and sardine oil that is rich in SPMs and EPA, as well as DHA precursors. Here, we assessed the effect of LIPINOVA® in wound dressing applications. LIPINOVA® showed biocompatibility with keratinocytes and fibroblasts, reduced the abundance of pro-inflammatory macrophages (Mφ1), and promoted in vitro wound closure. Daily application of the marine oil to open wounds made by punch biopsy in db/db mice promoted wound closure by accelerating the resolution of inflammation, inducing neoangiogenesis and Mφ1/Mφ2 macrophage polarization. In conclusion, LIPINOVA® displays pro-resolutive properties and could be exploited as a therapeutic agent for the treatment of diabetic ulcers.
Collapse
Affiliation(s)
- Imelda Ontoria-Oviedo
- Regenerative Medicine and Heart Transplantation Unit, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
| | - Elena Amaro-Prellezo
- Regenerative Medicine and Heart Transplantation Unit, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
| | - Delia Castellano
- Regenerative Medicine and Heart Transplantation Unit, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
| | | | | | - Amparo Ruiz-Saurí
- Departamento de Patología, Facultad de Medicina, Instituto de Investigación Sanitaria INCLIVA, Universitat de Valencia, 46026 Valencia, Spain
| | - Beatriz Pelacho
- Regenerative Medicine Department, Center for Applied Medical Research (CIMA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), University of Navarra, 31008 Pamplona, Spain
| | - Felipe Prósper
- Regenerative Medicine Department, Center for Applied Medical Research (CIMA), Instituto de Investigación Sanitaria de Navarra (IdiSNA), University of Navarra, 31008 Pamplona, Spain
- Departamento de Hematología y Terapia Celular, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - María Dolores Pérez del Caz
- Regenerative Medicine and Heart Transplantation Unit, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
| | - Pilar Sepúlveda
- Regenerative Medicine and Heart Transplantation Unit, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
- CIBERCV, Carlos III Institute of Health, 28029 Madrid, Spain
- Correspondence: or ; Tel.: +34-96-1246635
| |
Collapse
|
12
|
Rovira E, Moreno B, Razquin N, Hjerpe R, Gonzalez-Lopez M, Barrio R, Ruiz de los Mozos I, Ule J, Pastor F, Blazquez L, Fortes P. U1A is a positive regulator of the expression of heterologous and cellular genes involved in cell proliferation and migration. MOLECULAR THERAPY - NUCLEIC ACIDS 2022; 28:831-846. [PMID: 35664701 PMCID: PMC9136276 DOI: 10.1016/j.omtn.2022.05.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 05/07/2022] [Indexed: 11/16/2022]
Abstract
Here, we show that direct recruitment of U1A to target transcripts can increase gene expression. This is a new regulatory role, in addition to previous knowledge showing that U1A decreases the levels of U1A mRNA and other specific targets. In fact, genome-wide, U1A more often increases rather than represses gene expression and many U1A-upregulated transcripts are directly bound by U1A according to individual nucleotide resolution crosslinking and immunoprecipitation (iCLIP) studies. Interestingly, U1A-mediated positive regulation can be transferred to a heterologous system for biotechnological purposes. Finally, U1A-bound genes are enriched for those involved in cell cycle and adhesion. In agreement with this, higher U1A mRNA expression associates with lower disease-free survival and overall survival in many cancer types, and U1A mRNA levels positively correlate with those of some oncogenes involved in cell proliferation. Accordingly, U1A depletion leads to decreased expression of these genes and the migration-related gene CCN2/CTGF, which shows the strongest regulation by U1A. A decrease in U1A causes a strong drop in CCN2 expression and CTGF secretion and defects in the expression of CTGF EMT targets, cell migration, and proliferation. These results support U1A as a putative therapeutic target for cancer treatment. In addition, U1A-binding sequences should be considered in biotechnological applications.
Collapse
Affiliation(s)
- Eric Rovira
- Department of Gene Therapy and Regulation of Gene Expression, Center for Applied Medical Research (CIMA), University of Navarra (UNAV), 31008 Pamplona, Spain
| | - Beatriz Moreno
- Department of Molecular Therapy, Aptamer Unit, Center for Applied Medical Research (CIMA), University of Navarra (UNAV), 31008 Pamplona, Spain
| | - Nerea Razquin
- Department of Gene Therapy and Regulation of Gene Expression, Center for Applied Medical Research (CIMA), University of Navarra (UNAV), 31008 Pamplona, Spain
| | - Roland Hjerpe
- Department of Functional Genomics, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160 Derio, Spain
| | - Monika Gonzalez-Lopez
- Department of Functional Genomics, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160 Derio, Spain
| | - Rosa Barrio
- Department of Functional Genomics, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160 Derio, Spain
| | - Igor Ruiz de los Mozos
- Department of Neuromuscular Diseases, Institute of Neurology, UCL, WC1B5EH London, UK
- RNA Networks Lab, The Francis Crick Institute, NW11BF London, UK
| | - Jernej Ule
- Department of Neuromuscular Diseases, Institute of Neurology, UCL, WC1B5EH London, UK
- RNA Networks Lab, The Francis Crick Institute, NW11BF London, UK
| | - Fernando Pastor
- Department of Molecular Therapy, Aptamer Unit, Center for Applied Medical Research (CIMA), University of Navarra (UNAV), 31008 Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
| | - Lorea Blazquez
- Department of Neuromuscular Diseases, Institute of Neurology, UCL, WC1B5EH London, UK
- RNA Networks Lab, The Francis Crick Institute, NW11BF London, UK
- Neurosciences Area, Biodonostia Health Research Institute, 20014 San Sebastian, Spain
- Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain
- Corresponding author. Lorea Blazquez, Department of Neuromuscular Diseases, Institute of Neurology, UCL, WC1B5EH London, UK.
| | - Puri Fortes
- Department of Gene Therapy and Regulation of Gene Expression, Center for Applied Medical Research (CIMA), University of Navarra (UNAV), 31008 Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
- Liver and Digestive Diseases Networking Biomedical Research Centre (CIBERehd), Spain
- Spanish Network for Advanced Therapies (TERAV ISCIII), Spain
- Corresponding author. Puri Fortes, Neurosciences Area, Biodonostia Health Research Institute, 20014 San Sebastian, Spain.
| |
Collapse
|
13
|
Yalcin EB, Tong M, Homans C, de la Monte SM. Myriocin Treatment Reverses Alcohol-Induced Alterations in Polyunsaturated Fatty Acid-Containing Phospholipid Expression in the Liver. Nutr Metab Insights 2022; 15:11786388221082012. [PMID: 35250275 PMCID: PMC8891894 DOI: 10.1177/11786388221082012] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 01/23/2022] [Indexed: 01/30/2023] Open
Abstract
Chronic heavy alcohol exposure causes steatohepatitis manifested by abnormal intra-hepatocyte accumulation of lipid and parenchymal inflammation. Attendant alterations in polyunsaturated fatty acid (PUFA)-containing phospholipids could cause alcoholic liver disease (ALD) to progress by promoting oxidative stress, inflammation, and fibrogenesis. Previously we showed that myriocin, a serine palmitoyltransferase inhibitor, ameliorates experimental alcohol-induced steatohepatitis. However, the surprising overall therapeutic responses suggested that myriocin's targets may go beyond sphingolipids. To this end, the present study examines the effects of myriocin on hepatic composition of docosahexaenoic acid (DHA)- and arachidonic acid (AA)-containing phospholipids in an experimental model of ALD. A chronic+binge ethanol exposure model was generated by feeding Long Evans rats with ethanol-containing diets (24% caloric content) for 8 weeks and simultaneously binge gavage administering 2 g/kg ethanol on Tuesdays, Thursdays and Saturdays during Weeks 6-8. Myriocin was administered by i.p. injection on Mondays, Wednesdays, and Fridays of Weeks 3-8. Control rats were studied in parallel. Upon euthanasia, the livers were harvested to examine ethanol- and/or myriocin-modulation of hepatic lipids using matrix-assisted laser desorption/ionization imaging mass spectrometry (MALDI-IMS). Results were analyzed statistically by two-way analysis of variance and depicted with data bar plots and heatmaps. Chronic+binge ethanol exposures significantly increased hepatic expression of AA-containing phospholipids including PE(36:4) (P = .005), PE(38:4) (P = .03), and PI(38:4) (P = .04) and reduced DHA-containing phospholipids including PS(40:6) (P = .03) and PE(40:6) (P = .04) relative to control. Myriocin partially reversed ethanol's effects on hepatic PUFA expression by decreasing PE(36:4) (P = .004) and increasing PS(40:6) (P = .04) and PI(40:6) (P = .0003) relative to ethanol-exposed rats. Ethanol-mediated alterations in hepatic PUFA-containing phospholipids may contribute to hepatic oxidative and inflammatory injury by increasing AA and fibrogenesis by inhibiting DHA. The results suggest that Myriocin may help reduce or prevent long-term and progressive liver injury stemming from excessive chronic+binge ethanol consumption.
Collapse
Affiliation(s)
- Emine B Yalcin
- Liver Research Center, Division of Gastroenterology and Department of Medicine, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA,Division of Research, Providence VA Medical Center, Providence, RI, USA,Emine B Yalcin, Providence VA Medical Center, Research Service Building 35, 830 Chalkstone Ave, Providence, RI 02908, USA.
| | - Ming Tong
- Liver Research Center, Division of Gastroenterology and Department of Medicine, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA
| | - Camilla Homans
- Health and Human Biology, Brown University, Providence, RI, USA
| | - Suzanne M de la Monte
- Liver Research Center, Division of Gastroenterology and Department of Medicine, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA,Division of Research, Providence VA Medical Center, Providence, RI, USA,Department of Pathology and Laboratory Medicine, Providence VA Medical Center and the Women & Infants Hospital of Rhode Island, Providence, RI, USA,Departments of Neurology, Neurosurgery, and Pathology, Rhode Island Hospital and the Alpert Medical School of Brown University, Providence, RI, USA
| |
Collapse
|
14
|
Huang X, Sun J, Bian C, Ji S, Ji H. Docosahexaenoic acid lessens hepatic lipid accumulation and inflammation via the AMP-activated protein kinase and endoplasmic reticulum stress signaling pathways in grass carp ( Ctenopharyngodon idella). Food Funct 2022; 13:1846-1859. [PMID: 35084424 DOI: 10.1039/d1fo03214c] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The liver is the primary organ for frontline immune defense and lipid metabolism. Excessive lipid accumulation in the liver severely affects its metabolic homeostasis and causes metabolic diseases. Docosahexaenoic acid (DHA) is known for its beneficial effects on lipid metabolism and anti-inflammation, but its molecular mechanism remains unknown, especially in fish. In this study, we evaluated the protective effects of DHA on hepatic steatosis of grass carp (Ctenopharyngodon idella) in vivo and in vitro and mainly focused on the AMP-activated protein kinase (AMPK) and endoplasmic reticulum stress (ER stress) signaling pathway analysis. Grass carp were fed with purified diets supplemented with 0%, 0.5% and 1% DHA for 8 weeks in vivo. 1% DHA supplementation significantly decreased the liver triglyceride (TG), malondialdehyde (MDA), serum tumor necrosis factor α (TNFα) and nuclear factor kappa B (NFκB) contents. DHA administration suppressed ER stress and decreased the mRNA expressions related to hepatic inflammation and lipogenesis, accompanied by the activation of AMPK. Correspondingly, DHA activated the AMPK signaling pathway, and inhibited palmitic acid (PA)-evoked ER stress and lipid accumulation and inflammation of grass carp hepatocytes in vitro. In contrast, the inhibitor of AMPK (compound C, CC) abrogated the effects of DHA to improve PA-induced liver injury and ER stress. In conclusion, DHA inhibits ER stress in hepatocytes by the activation of AMPK and exerts protective effects on hepatic steatosis in terms of improving antioxidant ability, relieving hepatic inflammation and inhibiting hepatic lipogenesis. Our findings give a theoretical foundation for further elucidation of the beneficial role of DHA in vertebrates.
Collapse
Affiliation(s)
- Xiaocheng Huang
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, Yangling 712100, China.
| | - Jian Sun
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, Yangling 712100, China.
| | - Chenchen Bian
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, Yangling 712100, China.
| | - Shanghong Ji
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, Yangling 712100, China.
| | - Hong Ji
- College of Animal Science and Technology, Northwest Agriculture and Forestry University, Yangling 712100, China.
| |
Collapse
|
15
|
Fridén M, Rosqvist F, Ahlström H, Niessen HG, Schultheis C, Hockings P, Hulthe J, Gummesson A, Wanders A, Rorsman F, Risérus U, Vessby J. Hepatic Unsaturated Fatty Acids Are Linked to Lower Degree of Fibrosis in Non-alcoholic Fatty Liver Disease. Front Med (Lausanne) 2022; 8:814951. [PMID: 35083257 PMCID: PMC8784562 DOI: 10.3389/fmed.2021.814951] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 12/17/2021] [Indexed: 12/12/2022] Open
Abstract
Background: The hepatic lipidome of patients with early stages of non-alcoholic fatty liver disease (NAFLD) has been fairly well-explored. However, studies on more progressive forms of NAFLD, i.e., liver fibrosis, are limited. Materials and methods: Liver fatty acids were determined in cholesteryl esters (CE), phospholipids (PL), and triacylglycerols (TAG) by gas chromatography. Cross-sectional associations between fatty acids and biopsy-proven NAFLD fibrosis (n = 60) were assessed using multivariable logistic regression models. Stages of fibrosis were dichotomized into none-mild (F0–1) or significant fibrosis (F2–4). Models were adjusted for body-mass index (BMI), age and patatin-like phospholipase domain-containing protein 3 (PNPLA3 rs738409) (I148M) genotype. A secondary analysis examined whether associations from the primary analysis could be confirmed in the corresponding plasma lipid fractions. Results: PL behenic acid (22:0) was directly associated [OR (95% CI): 1.86 (1.00, 3.45)] whereas PL docosahexaenoic acid (22:6n-3) [OR (95% CI): 0.45 (0.23, 0.89)], TAG oleic acid (18:1n-9) [OR (95% CI): 0.52 (0.28, 0.95)] and 18:1n-9 and vaccenic acid (18:1n-7) (18:1) [OR (95% CI): 0.52 (0.28, 0.96)] were inversely associated with liver fibrosis. In plasma, TAG 18:1n-9 [OR (95% CI): 0.55 (0.31, 0.99)], TAG 18:1 [OR (95% CI): 0.54 (0.30, 0.97)] and PL 22:0 [OR (95% CI): 0.46 (0.25, 0.86)] were inversely associated with liver fibrosis. Conclusion: Higher TAG 18:1n-9 levels were linked to lower fibrosis in both liver and plasma, possibly reflecting an altered fatty acid metabolism. Whether PL 22:6n-3 has a protective role, together with a potentially adverse effect of hepatic 22:0, on liver fibrosis warrants large-scale studies.
Collapse
Affiliation(s)
- Michael Fridén
- Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, Uppsala University, Uppsala, Sweden
| | - Fredrik Rosqvist
- Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, Uppsala University, Uppsala, Sweden
| | - Håkan Ahlström
- Department of Surgical Sciences, Radiology, Uppsala University, Uppsala, Sweden.,Antaros Medical AB, BioVenture Hub, Mölndal, Sweden
| | - Heiko G Niessen
- Department of Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Christian Schultheis
- Department of Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Paul Hockings
- Antaros Medical AB, BioVenture Hub, Mölndal, Sweden.,MedTech West, Chalmers University of Technology, Gothenburg, Sweden
| | | | - Anders Gummesson
- Department of Clinical Genetics and Genomics, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Alkwin Wanders
- Department of Pathology, Aalborg University Hospital, Aalborg, Denmark
| | - Fredrik Rorsman
- Department of Medical Sciences, Gastroenterology and Hepatology, Uppsala University, Uppsala, Sweden
| | - Ulf Risérus
- Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, Uppsala University, Uppsala, Sweden
| | - Johan Vessby
- Department of Medical Sciences, Gastroenterology and Hepatology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
16
|
Rodríguez MJ, Sabaj M, Tolosa G, Herrera Vielma F, Zúñiga MJ, González DR, Zúñiga-Hernández J. Maresin-1 Prevents Liver Fibrosis by Targeting Nrf2 and NF-κB, Reducing Oxidative Stress and Inflammation. Cells 2021; 10:3406. [PMID: 34943914 PMCID: PMC8699629 DOI: 10.3390/cells10123406] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/19/2021] [Accepted: 11/22/2021] [Indexed: 12/12/2022] Open
Abstract
Liver fibrosis is a complex process characterized by the excessive accumulation of extracellular matrix (ECM) and an alteration in liver architecture, as a result of most types of chronic liver diseases such as cirrhosis, hepatocellular carcinoma (HCC) and liver failure. Maresin-1 (MaR1) is derivative of ω-3 docosahexaenoic acid (DHA), which has been shown to have pro-resolutive and anti-inflammatory effects. We tested the hypothesis that the application of MaR1 could prevent the development of fibrosis in an animal model of chronic hepatic damage. Sprague-Dawley rats were induced with liver fibrosis by injections of diethylnitrosamine (DEN) and treated with or without MaR1 for four weeks. In the MaR1-treated animals, levels of AST and ALT were normalized in comparison with DEN alone, the hepatic architecture was improved, and inflammation and necrotic areas were reduced. Cell proliferation, assessed by the mitotic activity index and the expression of Ki-67, was increased in the MaR1-treated group. MaR1 attenuated liver fibrosis and oxidative stress was induced by DEN. Plasma levels of the pro-inflammatory mediators TNF-α and IL-1β were reduced in MaR1-treated animals, whereas the levels of IL-10, an anti-inflammatory cytokine, increased. Interestingly, MaR1 inhibited the translocation of the p65 subunit of NF-κB, while increasing the activation of Nrf2, a key regulator of the antioxidant response. Finally, MaR1 treatment reduced the levels of the pro-fibrotic mediator TGF-β and its receptor, while normalizing the hepatic levels of IGF-1, a proliferative agent. Taken together, these results suggest that MaR1 improves the parameters of DEN-induced liver fibrosis, activating hepatocyte proliferation and decreasing oxidative stress and inflammation. These results open the possibility of MaR1 as a potential therapeutic agent in fibrosis and other liver pathologies.
Collapse
Affiliation(s)
- María José Rodríguez
- Departamento de Ciencias Básicas Biomédicas, Facultad de Ciencias de la Salud, Universidad de Talca, Talca 3460000, Chile; (M.J.R.); (F.H.V.); (M.J.Z.); (D.R.G.)
- Programa de Doctorado en Ciencias Mención Investigación y Desarrollo de Productos Bioactivos, Instituto de Química de los Recursos Naturales, Universidad de Talca, Talca 3460000, Chile
| | - Matías Sabaj
- Escuela de Tecnología Médica, Facultad de Ciencias de la Salud, Universidad de Talca, Talca 3460000, Chile; (M.S.); (G.T.)
| | - Gerardo Tolosa
- Escuela de Tecnología Médica, Facultad de Ciencias de la Salud, Universidad de Talca, Talca 3460000, Chile; (M.S.); (G.T.)
| | - Francisca Herrera Vielma
- Departamento de Ciencias Básicas Biomédicas, Facultad de Ciencias de la Salud, Universidad de Talca, Talca 3460000, Chile; (M.J.R.); (F.H.V.); (M.J.Z.); (D.R.G.)
- Programa de Doctorado en Ciencias Mención Investigación y Desarrollo de Productos Bioactivos, Instituto de Química de los Recursos Naturales, Universidad de Talca, Talca 3460000, Chile
| | - María José Zúñiga
- Departamento de Ciencias Básicas Biomédicas, Facultad de Ciencias de la Salud, Universidad de Talca, Talca 3460000, Chile; (M.J.R.); (F.H.V.); (M.J.Z.); (D.R.G.)
| | - Daniel R. González
- Departamento de Ciencias Básicas Biomédicas, Facultad de Ciencias de la Salud, Universidad de Talca, Talca 3460000, Chile; (M.J.R.); (F.H.V.); (M.J.Z.); (D.R.G.)
| | - Jessica Zúñiga-Hernández
- Departamento de Ciencias Básicas Biomédicas, Facultad de Ciencias de la Salud, Universidad de Talca, Talca 3460000, Chile; (M.J.R.); (F.H.V.); (M.J.Z.); (D.R.G.)
| |
Collapse
|
17
|
Kakizaki M, Yamamoto Y, Nakayama S, Kameda K, Nagashima E, Ito M, Suyama T, Matsuzaki Y, Chiba T, Sumiyoshi H, Inagaki Y, Kotani A. Human hepatocyte-derived extracellular vesicles attenuate the carbon tetrachloride-induced acute liver injury in mice. Cell Death Dis 2021; 12:1010. [PMID: 34707093 PMCID: PMC8551237 DOI: 10.1038/s41419-021-04204-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/25/2021] [Accepted: 08/12/2021] [Indexed: 11/18/2022]
Abstract
Acute liver injury (ALI) induced by chemicals or viruses can progress rapidly to acute liver failure (ALF), often resulting in death of patients without liver transplantation. Since liver transplantation is limited due to a paucity of donors, expensive surgical costs, and severe immune rejection, novel therapies are required to treat liver injury. Extracellular vesicles (EVs) are used for cellular communication, carrying RNAs, proteins, and lipids and delivering them intercellularly after being endocytosed by target cells. Recently, it was reported that EVs secreted from human hepatocytes have an ability to modulate the immune responses; however, these roles of EVs secreted from human hepatocytes were studied only with in vitro experiments. In the present study, we evidenced that EVs secreted from human hepatocytes attenuated the CCL4-induced ALI by inhibiting the recruitment of monocytes through downregulation of chemokine receptor in the bone marrow and recruitment of neutrophils through the reduction of C-X-C motif chemokine ligand 1 (CXCL1) and CXCL2 expression levels in the liver.
Collapse
Affiliation(s)
- Masatoshi Kakizaki
- grid.265061.60000 0001 1516 6626Department of Innovative Medical Science, Tokai University School of Medicine, Kanagawa, 259-1193 Japan ,Division of Hematological Malignancy, Institute of Medical Sciences, Tokai University, Kanagawa, 259-1193 Japan
| | - Yuichiro Yamamoto
- grid.265061.60000 0001 1516 6626Department of Innovative Medical Science, Tokai University School of Medicine, Kanagawa, 259-1193 Japan ,Division of Hematological Malignancy, Institute of Medical Sciences, Tokai University, Kanagawa, 259-1193 Japan
| | - Shunya Nakayama
- grid.265061.60000 0001 1516 6626Department of Innovative Medical Science, Tokai University School of Medicine, Kanagawa, 259-1193 Japan ,Division of Hematological Malignancy, Institute of Medical Sciences, Tokai University, Kanagawa, 259-1193 Japan
| | - Kazuaki Kameda
- grid.265061.60000 0001 1516 6626Department of Innovative Medical Science, Tokai University School of Medicine, Kanagawa, 259-1193 Japan ,Division of Hematological Malignancy, Institute of Medical Sciences, Tokai University, Kanagawa, 259-1193 Japan ,grid.415020.20000 0004 0467 0255Division of Hematology, Jichi Medical University Saitama Medical Center, Saitama, 330-8503 Japan
| | - Etsuko Nagashima
- grid.265061.60000 0001 1516 6626Department of Innovative Medical Science, Tokai University School of Medicine, Kanagawa, 259-1193 Japan ,Division of Hematological Malignancy, Institute of Medical Sciences, Tokai University, Kanagawa, 259-1193 Japan
| | - Masatoshi Ito
- grid.265061.60000 0001 1516 6626Support Center for Medical Research and Education, Tokai University School of Medicine, Kanagawa, 259-1193 Japan
| | - Takashi Suyama
- grid.411621.10000 0000 8661 1590Department of Life Science, Shimane University Faculty of Medicine, Izumo, Shimane Japan
| | - Yumi Matsuzaki
- grid.411621.10000 0000 8661 1590Department of Life Science, Shimane University Faculty of Medicine, Izumo, Shimane Japan
| | - Tetsuhiro Chiba
- grid.136304.30000 0004 0370 1101Department of Gastroenterology, Chiba University, Graduate School of Medicine, Inohana, Chuo-ku, Chiba, 260-8670 Japan
| | - Hideaki Sumiyoshi
- grid.265061.60000 0001 1516 6626Department of Innovative Medical Science, Tokai University School of Medicine, Kanagawa, 259-1193 Japan ,grid.265061.60000 0001 1516 6626Center for Matrix Biology and Medicine, Graduate School of Medicine, Tokai University, Kanagawa, 259-1193 Japan ,Institute of Medical Sciences, Tokai University, Kanagawa, 259-1193 Japan
| | - Yutaka Inagaki
- grid.265061.60000 0001 1516 6626Department of Innovative Medical Science, Tokai University School of Medicine, Kanagawa, 259-1193 Japan ,grid.265061.60000 0001 1516 6626Center for Matrix Biology and Medicine, Graduate School of Medicine, Tokai University, Kanagawa, 259-1193 Japan ,Institute of Medical Sciences, Tokai University, Kanagawa, 259-1193 Japan
| | - Ai Kotani
- grid.265061.60000 0001 1516 6626Department of Innovative Medical Science, Tokai University School of Medicine, Kanagawa, 259-1193 Japan ,Division of Hematological Malignancy, Institute of Medical Sciences, Tokai University, Kanagawa, 259-1193 Japan
| |
Collapse
|
18
|
Mierziak J, Kostyn K, Boba A, Czemplik M, Kulma A, Wojtasik W. Influence of the Bioactive Diet Components on the Gene Expression Regulation. Nutrients 2021; 13:3673. [PMID: 34835928 PMCID: PMC8619229 DOI: 10.3390/nu13113673] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/13/2021] [Accepted: 10/14/2021] [Indexed: 02/07/2023] Open
Abstract
Diet bioactive components, in the concept of nutrigenetics and nutrigenomics, consist of food constituents, which can transfer information from the external environment and influence gene expression in the cell and thus the function of the whole organism. It is crucial to regard food not only as the source of energy and basic nutriments, crucial for living and organism development, but also as the factor influencing health/disease, biochemical mechanisms, and activation of biochemical pathways. Bioactive components of the diet regulate gene expression through changes in the chromatin structure (including DNA methylation and histone modification), non-coding RNA, activation of transcription factors by signalling cascades, or direct ligand binding to the nuclear receptors. Analysis of interactions between diet components and human genome structure and gene activity is a modern approach that will help to better understand these relations and will allow designing dietary guidances, which can help maintain good health.
Collapse
Affiliation(s)
- Justyna Mierziak
- Faculty of Biotechnology, University of Wrocław, Przybyszewskiego 63/77, 51-148 Wroclaw, Poland; (A.B.); (M.C.); (A.K.)
| | - Kamil Kostyn
- Department of Genetics, Plant Breeding & Seed Production, Faculty of Life Sciences and Technology, Wroclaw University of Environmental and Life Sciences, pl. Grunwaldzki 24A, 50-363 Wroclaw, Poland;
| | - Aleksandra Boba
- Faculty of Biotechnology, University of Wrocław, Przybyszewskiego 63/77, 51-148 Wroclaw, Poland; (A.B.); (M.C.); (A.K.)
| | - Magdalena Czemplik
- Faculty of Biotechnology, University of Wrocław, Przybyszewskiego 63/77, 51-148 Wroclaw, Poland; (A.B.); (M.C.); (A.K.)
| | - Anna Kulma
- Faculty of Biotechnology, University of Wrocław, Przybyszewskiego 63/77, 51-148 Wroclaw, Poland; (A.B.); (M.C.); (A.K.)
| | - Wioleta Wojtasik
- Faculty of Biotechnology, University of Wrocław, Przybyszewskiego 63/77, 51-148 Wroclaw, Poland; (A.B.); (M.C.); (A.K.)
| |
Collapse
|
19
|
Liver Lipids of Patients with Hepatitis B and C and Associated Hepatocellular Carcinoma. Int J Mol Sci 2021; 22:ijms22105297. [PMID: 34069902 PMCID: PMC8157577 DOI: 10.3390/ijms22105297] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 05/04/2021] [Accepted: 05/15/2021] [Indexed: 12/11/2022] Open
Abstract
Hepatocellular carcinoma (HCC) still remains a difficult to cure malignancy. In recent years, the focus has shifted to lipid metabolism for the treatment of HCC. Very little is known about hepatitis B virus (HBV) and C virus (HCV)-related hepatic lipid disturbances in non-malignant and cancer tissues. The present study showed that triacylglycerol and cholesterol concentrations were similar in tumor adjacent HBV and HCV liver, and were not induced in the HCC tissues. Higher levels of free cholesterol, polyunsaturated phospholipids and diacylglycerol species were noted in non-tumorous HBV compared to HCV liver. Moreover, polyunsaturated phospholipids and diacylglycerols, and ceramides declined in tumors of HBV infected patients. All of these lipids remained unchanged in HCV-related HCC. In HCV tumors, polyunsaturated phosphatidylinositol levels were even induced. There were no associations of these lipid classes in non-tumor tissues with hepatic inflammation and fibrosis scores. Moreover, these lipids did not correlate with tumor grade or T-stage in HCC tissues. Lipid reprogramming of the three analysed HBV/HCV related tumors mostly resembled HBV-HCC. Indeed, lipid composition of non-tumorous HCV tissue, HCV tumors, HBV tumors and HBV/HCV tumors was highly similar. The tumor suppressor protein p53 regulates lipid metabolism. The p53 and p53S392 protein levels were induced in the tumors of HBV, HCV and double infected patients, and this was significant in HBV infection. Negative correlation of tumor p53 protein with free cholesterol indicates a role of p53 in cholesterol metabolism. In summary, the current study suggests that therapeutic strategies to target lipid metabolism in chronic viral hepatitis and associated cancers have to consider disease etiology.
Collapse
|
20
|
Kim Y. Emerging Treatment Options for Sarcopenia in Chronic Liver Disease. Life (Basel) 2021; 11:life11030250. [PMID: 33803020 PMCID: PMC8002763 DOI: 10.3390/life11030250] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/13/2021] [Accepted: 03/15/2021] [Indexed: 12/17/2022] Open
Abstract
Sarcopenia is characterized by a skeletal muscle disorder with progressive and generalized loss of muscle mass and function, and it increases the risk of adverse outcomes with considerable prevalence in patients with chronic liver disease. Sarcopenia in chronic liver disease underlies complicated and multifactorial mechanisms for pathogenesis, including alterations in protein turnover, hyperammonemia, energy disposal, hormonal changes, and chronic inflammation. The key contribution to sarcopenia in patients with chronic liver diseases can be the hyperammonemia-induced upregulation of myostatin, which causes muscle atrophy via the expression of atrophy-related genes. Several clinical studies on emerging treatment options for sarcopenia have been reported, but only a few have focused on patients with chronic liver diseases, with mostly nutritional and behavioral interventions being carried out. The inhibition of the myostatin-activin receptor signaling pathway and hormonal therapy might be the most promising therapeutic options in combination with an ammonia-lowering approach in sarcopenic patients with chronic liver diseases. This review focuses on current and emerging treatment options for sarcopenia in chronic liver diseases with underlying mechanisms to counteract this condition.
Collapse
Affiliation(s)
- Yun Kim
- Hanyang Medicine-Engineering-Bio Collaborative & Comprehensive Center for Drug Development, Hanyang University, Seoul 04763, Korea
| |
Collapse
|
21
|
Abriz AE, Rahbarghazi R, Nourazarian A, Avci ÇB, Mahboob SA, Rahnema M, Araghi A, Heidarzadeh M. Effect of docosahexaenoic acid plus insulin on atherosclerotic human endothelial cells. JOURNAL OF INFLAMMATION-LONDON 2021; 18:10. [PMID: 33602249 PMCID: PMC7890865 DOI: 10.1186/s12950-021-00277-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 02/08/2021] [Indexed: 12/25/2022]
Abstract
Background Atherosclerosis is touted as one of the most critical consequences of diabetes mellitus indicated by local inflammation of endothelial cells. The Effect of Omega 3 fatty acids, mainly docosahexaenoic acid (DHA), has been investigated in cells after exposure to high doses of lipids. The current experiment aimed to address the modulatory effects of docosahexaenoic acid and insulin in palmitic-treated human endothelial cells. Methods Human umbilical vein endothelial cells were treated with 1 mM palmitic acid, 50 μM insulin, 50 μM docosahexaenoic acid, and their combination for 48 h. Cell survival rate and apoptosis were measured using MTT and flow cytometry assays. The Griess assay detected NO levels. Protein levels of TNF-α, IL-6, and NF-κB were studied using ELISA and immunofluorescence imaging. The expression of genes participating in atherosclerosis was monitored using PCR array analysis. Results Oil Red O staining showed the inhibitory effect of DHA and insulin to reduce the intracellular accumulation of palmitic acid. Both DHA and Insulin blunted palmitic acid detrimental effects on HUVECs indicated by an increased survival rate (p < 0.05). The percent of apoptotic cells was decreased in palmitic-treated cells received insulin and DHA compared to palmitic-treated group (p < 0.05). Based on our data, DHA and Insulin diminished the production of all inflammatory cytokines, TNF-α, IL-6, and NF-κB, in palmitic-treated cells (p < 0.05). Similar to these data, NO production was also decreased in all groups treated with insulin and DHA compared to the palmitic-treated cells (p < 0.05). PCR array analysis revealed the modulatory effect of DHA and insulin on the expression of atherosclerosis-related genes pre-treated with palmitic acid compared to the control group (p < 0.05). Conclusion DHA and Insulin could alter the dynamic growth and dysfunctional activity of human endothelial cells after treatment with palmitic acid. Taken together, Omega 3 fatty acids, along with insulin, could dictate specific cell behavior in endothelial cells in vitro.
Collapse
Affiliation(s)
- Aysan Eslami Abriz
- Department of Biochemistry, Higher Education Institute of Rab-Rashid, Tabriz, Iran.,Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Nourazarian
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Golgasht St, Tabriz, 51666-16471, Iran.
| | - Çıgır Biray Avci
- Department of Medical Biology, Faculty of Medicine, Ege University, Izmir, Turkey
| | - Soltan Ali Mahboob
- Department of Biochemistry, Higher Education Institute of Rab-Rashid, Tabriz, Iran
| | - Maryam Rahnema
- Department of Biochemistry, Higher Education Institute of Rab-Rashid, Tabriz, Iran.,Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Atefeh Araghi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Amol University of Special Modern Technologies, Amol, Iran
| | - Morteza Heidarzadeh
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
22
|
Hey P, Gow P, Testro AG, Apostolov R, Chapman B, Sinclair M. Nutraceuticals for the treatment of sarcopenia in chronic liver disease. Clin Nutr ESPEN 2021; 41:13-22. [PMID: 33487256 DOI: 10.1016/j.clnesp.2020.11.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 11/30/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND AIMS Sarcopenia, defined as loss of muscle mass, strength and function, is associated with adverse clinical outcomes in patients with cirrhosis. Despite improved understanding of the multifaceted pathogenesis, there are few established therapies to treat or prevent muscle loss in this population. This narrative review examines the available literature investigating the role of nutraceuticals for the prevention or treatment of muscle wasting in chronic liver disease. METHODS A comprehensive search or Medline and PubMED databases was conducted. Reference lists were screened to identify additional articles. RESULTS A number of nutritional supplements and vitamins target the specific metabolic derangements that contribute to sarcopenia in cirrhosis including altered amino acid metabolism, hyperammonaemia and inflammation. Branched chain amino acid (BCAA) supplementation has proposed anabolic effects through dual pathways of enhanced ammonia clearance and stimulation of muscle protein synthesis. l-carnitine also has multimodal effects on muscle and shows promise as a therapy for muscle loss through anti-inflammatory, antioxidant and ammonia lowering properties. Other nutraceuticals including l-ornithine l-aspartate, omega-3 polyunsaturated fatty acids and zinc and vitamin D supplementation, may similarly have positive effects on muscle homeostasis, however further evidence to support their use in cirrhotic populations is required. CONCLUSION Nutraceuticals offer a promising and likely safe adjunct to standard care for sarcopenia in cirrhosis. While there is most evidence to support the use of BCAA and l-carnitine supplementation, further well-designed clinical trials are needed to elucidate their efficacy as a therapy for muscle loss in this population.
Collapse
Affiliation(s)
- Penelope Hey
- Liver Transplant Unit, Austin Health, 145 Studley Rd, Heidelberg, Victoria, Australia; The University of Melbourne, Parkville, Victoria, Australia.
| | - Paul Gow
- Liver Transplant Unit, Austin Health, 145 Studley Rd, Heidelberg, Victoria, Australia; The University of Melbourne, Parkville, Victoria, Australia.
| | - Adam G Testro
- Liver Transplant Unit, Austin Health, 145 Studley Rd, Heidelberg, Victoria, Australia; The University of Melbourne, Parkville, Victoria, Australia.
| | - Ross Apostolov
- Liver Transplant Unit, Austin Health, 145 Studley Rd, Heidelberg, Victoria, Australia; The University of Melbourne, Parkville, Victoria, Australia.
| | - Brooke Chapman
- The University of Melbourne, Parkville, Victoria, Australia; Department of Nutrition and Dietetics, Austin Health, 145 Studley Rd, Heidelberg, Victoria, Australia.
| | - Marie Sinclair
- Liver Transplant Unit, Austin Health, 145 Studley Rd, Heidelberg, Victoria, Australia; The University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
23
|
Quan X, Liu H, Ye D, Ding X, Su X. Forsythoside A Alleviates High Glucose-Induced Oxidative Stress and Inflammation in Podocytes by Inactivating MAPK Signaling via MMP12 Inhibition. Diabetes Metab Syndr Obes 2021; 14:1885-1895. [PMID: 33953587 PMCID: PMC8089089 DOI: 10.2147/dmso.s305092] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 03/31/2021] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Podocyte injury serves an important role during the progression of diabetic nephropathy (DN). The aim of this study was to investigate the effects of forsythoside A (FA) on high glucose (HG)-induced podocyte injury and to identify the possible mechanisms. METHODS MPC-5 podocytes were cultured under HG conditions. After exposure to different doses of FA, cell viability and apoptosis were respectively evaluated with CCK-8 assay and flow cytometry. Then, the levels of oxidative stress-related markers and inflammatory factors were examined by corresponding kits. Western blot analysis was employed to detect the expression of Nox2, Nox4, COX-2, iNOS and matrix metalloproteinases 12 (MMP12). Subsequently, MMP12 was overexpressed to assess whether the effects of FA on HG-stimulated podocyte injury were mediated by MMP12 and MAPK signaling. RESULTS Results indicated that FA dose-dependently elevated cell viability, reduced cell apoptosis in HG-induced MPC-5 cells. Additionally, FA significantly inhibited oxidative stress, which could be certified by decreased content of malondialdehyde (MDA), enhanced activities of superoxide dismutase (SOD) and catalase (CAT), and downregulated expression of Nox2 and Nox4. Moreover, notably reduced levels of tumor necrosis factor (TNF)-α, interleukin (IL)-1β and IL-6 were observed in FA-treated MPC-5 cells under HG conditions, accompanied by decreased COX-2 and iNOS expression. Remarkably, FA suppressed MMP12 expression in a dose-dependent manner, and the effects of FA on MPC-5 cells exposed to HG were partially counteracted by MMP12 overexpression. Mechanically, FA inactivated the expression of phospho-ERK (p-ERK), p-p38 and p-JNK, which was restored after MMP12 overexpression. CONCLUSION These findings demonstrate a protective mechanism of FA by inactivating MAPK signaling via MMP12 inhibition in HG-induced podocyte injury, providing a promising therapeutic candidate for the treatment of DN.
Collapse
Affiliation(s)
- Xiaohong Quan
- Experiment & Teaching Center for Basic Medicine, Chifeng University School of Basic Medical Sciences, Chifeng, Inner Mongolia, 024000, People’s Republic of China
- Correspondence: Xiaohong Quan Experiment & Teaching Center for Basic Medicine, Chifeng University School of Basic Medical Sciences, No. 1 Yingbin Road, Chifeng City, Inner Mongolia, 024000, People’s Republic of China Email
| | - Huihui Liu
- Experiment & Teaching Center for Basic Medicine, Chifeng University School of Basic Medical Sciences, Chifeng, Inner Mongolia, 024000, People’s Republic of China
| | - Dongmei Ye
- Core Facility Center for Functional Experiments, CUSBMS, Chifeng University School of Basic Medical Sciences, Chifeng, Inner Mongolia, 024000, People’s Republic of China
| | - Xinling Ding
- Department of Human Anatomy, CUSBMS, Chifeng University School of Basic Medical Sciences, Chifeng, Inner Mongolia, 024000, People’s Republic of China
| | - Xiulan Su
- Clinical Research Center for Medical Sciences, IMMU, Chifeng University School of Basic Medical Sciences, Chifeng, Inner Mongolia, 024000, People’s Republic of China
| |
Collapse
|
24
|
Xue W, Mao J, Chen Q, Ling W, Sun Y. Mogroside IIIE Alleviates High Glucose-Induced Inflammation, Oxidative Stress and Apoptosis of Podocytes by the Activation of AMPK/SIRT1 Signaling Pathway. Diabetes Metab Syndr Obes 2020; 13:3821-3830. [PMID: 33116729 PMCID: PMC7585782 DOI: 10.2147/dmso.s276184] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 09/23/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Diabetic nephropathy (DN) is the leading cause of impaired renal function. The purpose of this study was to investigate the effects of Mogroside IIIE (MG IIIE), a cucurbitane-type compound isolated from Siraitia grosvenorii, in high glucose (HG)-induced podocytes and the possible mechanisms. METHODS MPC-5 cells were cultured under normal glucose or HG conditions. After treatment with MG IIIE, cell viability was examined using a cell counting kit-8 assay. The contents of inflammatory factors and oxidative stress-related markers were determined using the corresponding kits. Additionally, apoptosis of MPC-5 cells was determined using flow cytometry assay and the levels of apoptosis-associated proteins were evaluated by Western blot analysis. Moreover, the expression of proteins in AMPK/SIRT1 signaling was tested and the compound C, an AMPK inhibitor, was used to study whether the effects of MG IIIE on HG-induced MPC-5 cells were mediated by activation of the AMPK/SIRT1 signaling pathway. RESULTS MG IIIE elevated the cell viability of HG-induced MPC-5 cells, reduced the concentrations of inflammatory cytokines and decreased the levels of oxidative stress-related markers. What's more, the apoptosis of podocytes induced by HG was inhibited after MG IIIE intervention, accompanied by the upregulated expression of Bcl-2 and downregulated expression of Bax, cleaved caspase-3 and cleaved caspase-9. It was also found that MG IIIE could activate the AMPK/SIRT1 signaling, but compound C inhibited this pathway and reversed the inhibitory effects of MG IIIE on inflammation, oxidative stress and apoptosis in HG-stimulated podocytes. CONCLUSION MG IIIE can alleviate HG-induced inflammation and oxidative stress of podocytes by the activation of AMPK-SIRT1 signaling.
Collapse
Affiliation(s)
- Wei Xue
- Department of Pharmacy, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, Guangxi530011, People’s Republic of China
| | - Juhua Mao
- Center for Drug Control, Lishui Institute for Quality Inspection and Testing, Lishui, Zhejiang323000, People’s Republic of China
| | - Qingjie Chen
- Department of Pharmacy, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, Guangxi530011, People’s Republic of China
| | - Weide Ling
- Department of Pharmacy, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, Guangxi530011, People’s Republic of China
| | - Yuqi Sun
- Department of Anesthesiology, Guangzhou 12th People’s Hospital, Guangzhou, Guangdong510620, People’s Republic of China
| |
Collapse
|
25
|
Ronda OAHO, van de Heijning BJM, de Bruin A, Thomas RE, Martini I, Koehorst M, Gerding A, Koster MH, Bloks VW, Jurdzinski A, Mulder NL, Havinga R, van der Beek EM, Reijngoud DJ, Kuipers F, Verkade HJ. Spontaneous liver disease in wild-type C57BL/6JOlaHsd mice fed semisynthetic diet. PLoS One 2020; 15:e0232069. [PMID: 32956351 PMCID: PMC7505464 DOI: 10.1371/journal.pone.0232069] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 08/16/2020] [Indexed: 11/19/2022] Open
Abstract
Mouse models are frequently used to study mechanisms of human diseases. Recently, we observed a spontaneous bimodal variation in liver weight in C57BL/6JOlaHsd mice fed a semisynthetic diet. We now characterized the spontaneous variation in liver weight and its relationship with parameters of hepatic lipid and bile acid (BA) metabolism. In male C57BL/6JOlaHsd mice fed AIN-93G from birth to postnatal day (PN)70, we measured plasma BA, lipids, Very low-density lipoprotein (VLDL)-triglyceride (TG) secretion, and hepatic mRNA expression patterns. Mice were sacrificed at PN21, PN42, PN63 and PN70. Liver weight distribution was bimodal at PN70. Mice could be subdivided into two nonoverlapping groups based on liver weight: 0.6 SD 0.1 g (approximately one-third of mice, small liver; SL), and 1.0 SD 0.1 g (normal liver; NL; p<0.05). Liver histology showed a higher steatosis grade, inflammation score, more mitotic figures and more fibrosis in the SL versus the NL group. Plasma BA concentration was 14-fold higher in SL (p<0.001). VLDL-TG secretion rate was lower in SL mice, both absolutely (-66%, p<0.001) and upon correction for liver weight (-44%, p<0.001). Mice that would later have the SL-phenotype showed lower food efficiency ratios during PN21-28, suggesting the cause of the SL phenotype is present at weaning (PN21). Our data show that approximately one-third of C57BL/6JOlaHsd mice fed semisynthetic diet develop spontaneous liver disease with aberrant histology and parameters of hepatic lipid, bile acid and lipoprotein metabolism. Study designs involving this mouse strain on semisynthetic diets need to take the SL phenotype into account. Plasma lipids may serve as markers for the identification of the SL phenotype.
Collapse
Affiliation(s)
- Onne A. H. O. Ronda
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | | | - Alain de Bruin
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Dutch Molecular Pathology Center, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Rachel E. Thomas
- Dutch Molecular Pathology Center, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Ingrid Martini
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Martijn Koehorst
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Albert Gerding
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Mirjam H. Koster
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Vincent W. Bloks
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Angelika Jurdzinski
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Niels L. Mulder
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Rick Havinga
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Eline M. van der Beek
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Danone Nutricia Research, Uppsalalaan, Utrecht, The Netherlands
| | - Dirk-Jan Reijngoud
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Folkert Kuipers
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Henkjan J. Verkade
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- * E-mail:
| |
Collapse
|
26
|
Xia DN, Tan YQ, Yang JY, Zhou G. Omega-3 polyunsaturated fatty acids: a promising approach for the management of oral lichen planus. Inflamm Res 2020; 69:989-999. [PMID: 32770320 DOI: 10.1007/s00011-020-01388-0] [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: 04/08/2020] [Revised: 07/26/2020] [Accepted: 07/29/2020] [Indexed: 10/23/2022] Open
Abstract
BACKGROUND Oral lichen planus (OLP) is a T-cell-mediated inflammatory disease with a risk of malignant transformation. Although the etiology of OLP is still uncertain, growing evidence suggests that oral microbiota, antigen-specific, and non-specific mechanisms are involved in the pathogenesis of OLP. Antigen-specific mechanisms include antigen presentation, T-cell activation, nuclear factor-kappa B signaling pathway, and cytokine secretion, while non-specific mechanisms consist of matrix metalloproteinases (MMP)-9 upregulation, psychological pressure, oxidative damage, aberrant expression of microRNAs (miRNAs), and autophagy. Till now, there is no cure for OLP, and the main purpose of OLP therapy is symptomatic control. FINDING Seafood and its derivative omega-3 polyunsaturated fatty acids (n-3 PUFAs) can suppress antigen presentation, T-cell activation, and nuclear factor-kappa B signaling pathway, modulate the overexpressed inflammatory cytokines, inhibit the expression of MMP-9, as well as regulate the expression of miRNAs and autophagy. And they are possible agents for ameliorating psychological disorder and oxidative damage. Moreover, n-3 PUFAs supplementation has a beneficial effect on preventing tumorigenesis. CONCLUSION n-3 PUFAs consumption may provide a non-toxic, inexpensive administration for OLP.
Collapse
Affiliation(s)
- Duo-Na Xia
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Luoyu Road 237, 430070, Wuhan, China
| | - Ya-Qin Tan
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Luoyu Road 237, 430070, Wuhan, China
| | - Jing-Ya Yang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Luoyu Road 237, 430070, Wuhan, China
| | - Gang Zhou
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Luoyu Road 237, 430070, Wuhan, China. .,Department of Oral Medicine, School and Hospital of Stomatology, Wuhan University, Wuhan, China.
| |
Collapse
|
27
|
Mariqueo TA, Zúñiga-Hernández J. Omega-3 derivatives, specialized pro-resolving mediators: Promising therapeutic tools for the treatment of pain in chronic liver disease. Prostaglandins Leukot Essent Fatty Acids 2020; 158:102095. [PMID: 32450460 DOI: 10.1016/j.plefa.2020.102095] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 02/02/2020] [Accepted: 03/23/2020] [Indexed: 12/12/2022]
Abstract
The main causes of liver injury are associated with inflammation and permanent damage. They can cause chronic liver disease (CLD), which is mainly related to viral hepatitis, alcohol consumption and non-alcoholic steatohepatitis, leading to fibrosis, cirrhosis and hepatocellular carcinoma. These conditions prevent the liver from working normally and make it begin to fail, which in turn may prompt a liver transplant. CLD and cirrhosis are the eleventh cause of death worldwide. At present, there are no approved pharmacological treatments to prevent, treat or resolve liver fibrosis. The prevalence of pain in the hepatic disease is elevated with ranges between 30% and 40%. Most of the pain drugs require hepatic function; therefore, the suitable control of pain is still a clinical challenge. Specialized pro-resolving mediators (SPM): lipoxins, resolvins, protectins and maresins, are potent endogenous molecules (nM concentrations) that modulate inflammatory body responses by reducing neutrophil infiltration, macrophage activity and pain sensitization. SPM have anti-inflammatory properties, stimulate tissue resolution, repair and regeneration, and exhibit anti-nociceptive actions. Furthermore, SPM were tried on different cellular, animal models and human observational data of liver injury, improving the pathogenesis of inflammation and fibrosis. In the present work, we will describe recent evidence that suggests that SPM can be used as a therapeutic option for CLD. Additionally, we will examine the role of SPM in the control of pain in pathologies associated with liver injury.
Collapse
Affiliation(s)
- T A Mariqueo
- Centro de Investigaciones Medicas, Escuela de Medicina, Universidad de Talca, Talca, Chile
| | - J Zúñiga-Hernández
- Centro de Investigaciones Medicas, Escuela de Medicina, Universidad de Talca, Talca, Chile.
| |
Collapse
|
28
|
|
29
|
Moreira RJ, Castro É, Oliveira TE, Belchior T, Peixoto AS, Chaves-Filho AB, Moreno MF, Lima JD, Yoshinaga M, Miyamoto S, Morais MRPT, Zorn TMT, Cogliati B, Iwai LK, Palmisano G, Cabral FJ, Festuccia W. Lipoatrophy-Associated Insulin Resistance and Hepatic Steatosis are Attenuated by Intake of Diet Rich in Omega 3 Fatty Acids. Mol Nutr Food Res 2020; 64:e1900833. [PMID: 31978277 DOI: 10.1002/mnfr.201900833] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 01/13/2020] [Indexed: 02/06/2023]
Abstract
SCOPE Glucose homeostasis and progression of nonalcoholic fatty liver disease (NAFLD) and hepatomegaly in severe lipoatrophic mice and their modulation by intake of a diet rich in omega 3 (n-3) fatty acids (HFO) are evaluated. METHODS AND RESULTS Severe lipoatrophic mice induced by PPAR-γ deletion exclusively in adipocytes (A-PPARγ KO) and littermate controls (A-PPARγ WT) are evaluated for glucose homeostasis and liver mass, proteomics, lipidomics, inflammation, and fibrosis. Lipoatrophic mice are heavier than controls, severely glucose intolerant, and hyperinsulinemic, and develop NAFLD characterized by increased liver glycogen, triacylglycerol, and diacylglycerol contents, mitotic index, apoptosis, inflammation, steatosis score, fibrosis, and fatty acid synthase (FAS) content and activity. Lipoatrophic mice also display liver enrichment with monounsaturated in detriment of polyunsaturated fatty acids including n-3 fatty acids, and increased content of cardiolipin, a tetracyl phospholipid exclusively found at the mitochondria inner membrane. Administration of a high-fat diet rich in n-3 fatty acids (HFO) to lipoatrophic mice enriches liver with n-3 fatty acids, reduces hepatic steatosis, FAS content and activity, apoptosis, inflammation, and improves glucose homeostasis. CONCLUSION Diet enrichment with n-3 fatty acids improves glucose homeostasis and reduces liver steatosis and inflammation without affecting hepatomegaly in severe lipoatrophic mice.
Collapse
Affiliation(s)
- Rafael J Moreira
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, 05508000, Brazil
| | - Érique Castro
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, 05508000, Brazil
| | - Tiago E Oliveira
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, 05508000, Brazil
| | - Thiago Belchior
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, 05508000, Brazil
| | - Albert S Peixoto
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, 05508000, Brazil
| | - Adriano B Chaves-Filho
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, 05508000, Brazil
| | - Mayara F Moreno
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, 05508000, Brazil
| | - Janayna D Lima
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, 05508000, Brazil
| | - Marcos Yoshinaga
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, 05508000, Brazil
| | - Sayuri Miyamoto
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, 05508000, Brazil
| | - Mychel R P T Morais
- Department of Cell Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, 05508000, Brazil
| | - Telma M T Zorn
- Department of Cell Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, 05508000, Brazil
| | - Bruno Cogliati
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, 05508010, Brazil
| | - Leo K Iwai
- Special Laboratory of Applied Toxicology, Center of Toxins, Immune-response and Cell Signaling (LETA/ CeTICS), Butantan Institute, São Paulo, 05503400, Brazil
| | - Giuseppe Palmisano
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, 05508000, Brazil
| | | | - William Festuccia
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, 05508000, Brazil
| |
Collapse
|
30
|
Hu ZH, Kong YY, Ren JJ, Huang TJ, Wang YQ, Liu LX. Kidney and lung tissue modifications after BDL-induced liver injury in mice are associated with increased expression of IGFBPrP1 and activation of the NF-κB inflammation pathway. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2020; 13:192-202. [PMID: 32211099 PMCID: PMC7061808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 01/23/2020] [Indexed: 06/10/2023]
Abstract
BACKGROUND Hepatorenal and hepatopulmonary syndrome are common clinical diseases; however, their mechanisms have not been fully elucidated. Our aim was to determine whether liver injury by bile duct ligation (BDL) causes modifications in kidney and lung tissue in mice, and to explore the possible mechanism of these changes. METHODS BDL in mice was used as a research model. Pathologic changes of liver, kidney, and lung tissue were observed by hematoxylin-eosin (H&E) staining. The expression of IGFBPrP1, NF-κB, TNF-α, and IL-6 were investigated in liver, kidney, and lung tissue by immunohistochemical staining and western blot. The correlation between IGFBPrP1 and NF-κB, TNF-α, and IL-6 protein expression in liver, kidney, and lung tissues of each group was analyzed by the Pearson method. RESULTS H&E staining showed, after BDL administration in mice, different degrees of inflammatory change in liver, kidney, and lung tissues of mice in each group. The results of immunohistochemical staining and western blot analysis showed increased expressions of IGFBPrP1, NF-κB, TNF-α, and IL-6 after BDL. Pearson correlation analysis showed that IGFBPrP1 positively correlated with the expressions of NF-κB, TNF-α, and IL-6. CONCLUSION Liver injury caused by bile duct ligation can lead to kidney and lung tissue injury in mice. The mechanism of injury may be related to the high expression of liver injury factor IGFBPrP1, transcription factor NF-κB, proinflammatory cytokine TNF-α, and IL-6 in kidney and lung tissue. Moreover, an increased expression level of IGFBPrP1 may be accompanied by the activation of the NF-κB inflammatory pathway.
Collapse
Affiliation(s)
- Zhi-Hui Hu
- Department of Gastroenterology and Hepatology, The First Hospital of Shanxi Medical UniversityTaiyuan 030001, China
| | - Yang-Yang Kong
- Department of Gastroenterology and Hepatology, The First Hospital of Shanxi Medical UniversityTaiyuan 030001, China
| | - Jun-Jie Ren
- Department of Gastroenterology and Hepatology, The First Hospital of Shanxi Medical UniversityTaiyuan 030001, China
| | - Ting-Juan Huang
- Department of Gastroenterology and Hepatology, The First Hospital of Shanxi Medical UniversityTaiyuan 030001, China
| | - Yan-Qin Wang
- Department of Gastroenterology and Hepatology, The First Hospital of Shanxi Medical UniversityTaiyuan 030001, China
| | - Li-Xin Liu
- Department of Gastroenterology and Hepatology, The First Hospital of Shanxi Medical UniversityTaiyuan 030001, China
- Experimental Center of Science and Research, The First Hospital of Shanxi Medical UniversityTaiyuan 030001, China
- Key Laboratory of Cell Physiology, Department of The Ministry of Education, Shanxi Medical UniversityTaiyuan 030001, China
| |
Collapse
|
31
|
Yoo HJ, Jung KJ, Kim M, Kim M, Kang M, Jee SH, Choi Y, Lee JH. Liver Cirrhosis Patients Who Had Normal Liver Function Before Liver Cirrhosis Development Have the Altered Metabolic Profiles Before the Disease Occurrence Compared to Healthy Controls. Front Physiol 2019; 10:1421. [PMID: 31803070 PMCID: PMC6877605 DOI: 10.3389/fphys.2019.01421] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 11/04/2019] [Indexed: 12/12/2022] Open
Abstract
Liver cirrhosis (LC) is the final usual outcome of liver damage induced by various chronic liver diseases. Because of asymptomatic nature of LC, it is usually diagnosed at late and advanced stages, and patients are easy to miss the best timing for treatment. Thus, the early detection of LC is needed. In the prospective Korean Cancer Prevention Study-II (K-II), we aimed to identify valuable biomarkers for LC using metabolomics to distinguish subjects with incident LC (LC group) from subjects free from LC (control group) during a mean 7-year follow-up period. Metabolic alterations were investigated using baseline serum specimens acquired from 94 subjects with incident LC and 180 age- and sex-matched LC-free subjects via ultra-performance liquid chromatography (UPLC)-linear-trap quardrupole (LTQ)-Orbitrap mass spectrometry (MS). As a result of the metabolic analysis, 46 metabolites were identified. Among them, 11 and 18 metabolite level showed a significant increase and decrease, respectively, in the LC group compared to the control group. Nine metabolic pathways, including glyoxylate and dicarboxylate metabolism, amino acid metabolism, fatty acid metabolism, linoleic acid metabolism, α-linolenic acid metabolism, and arachidonic acid metabolism, were significantly different between the two groups. Logistic regression demonstrated that the LC emergence was independently affected by serum levels of myristic acid, palmitic acid, linoleic acid, eicosapentaenoic acid (EPA), lysophosphatidic acid (LPA) (18:1), glycolic acid, lysophosphatidylcholine (lysoPC) (22:6), and succinylacetone (R 2 = 0.837, P < 0.001). This prospective study revealed that dysregulation of various metabolism had the clinical relevance on the LC development. Moreover, myristic acid, palmitic acid, linoleic acid, EPA, LPA (18:1), glycolic acid, lysoPC (22:6), and succinylacetone were emerged as independent variables influencing the incidence of LC. The results support that the early biomarkers found in this study may useful for predicting and remedying the risk of LC.
Collapse
Affiliation(s)
- Hye Jin Yoo
- National Leading Research Laboratory of Clinical Nutrigenetics/Nutrigenomics, Department of Food and Nutrition, College of Human Ecology, Yonsei University, Seoul, South Korea
- Research Center for Silver Science, Institute of Symbiotic Life-TECH, Yonsei University, Seoul, South Korea
| | - Keum Ji Jung
- Institute for Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, South Korea
| | - Minkyung Kim
- Research Center for Silver Science, Institute of Symbiotic Life-TECH, Yonsei University, Seoul, South Korea
| | - Minjoo Kim
- Research Center for Silver Science, Institute of Symbiotic Life-TECH, Yonsei University, Seoul, South Korea
| | - Minsik Kang
- National Leading Research Laboratory of Clinical Nutrigenetics/Nutrigenomics, Department of Food and Nutrition, College of Human Ecology, Yonsei University, Seoul, South Korea
| | - Sun Ha Jee
- Institute for Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, South Korea
| | - Yoonjeong Choi
- Institute for Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, South Korea
| | - Jong Ho Lee
- National Leading Research Laboratory of Clinical Nutrigenetics/Nutrigenomics, Department of Food and Nutrition, College of Human Ecology, Yonsei University, Seoul, South Korea
- Research Center for Silver Science, Institute of Symbiotic Life-TECH, Yonsei University, Seoul, South Korea
| |
Collapse
|
32
|
A Nutraceutical Rich in Docosahexaenoic Acid Improves Portal Hypertension in a Preclinical Model of Advanced Chronic Liver Disease. Nutrients 2019; 11:nu11102358. [PMID: 31623374 PMCID: PMC6835927 DOI: 10.3390/nu11102358] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 09/25/2019] [Accepted: 09/29/2019] [Indexed: 02/06/2023] Open
Abstract
Inflammation and oxidative stress play a key role in the pathophysiology of advanced chronic liver disease (ACLD) and portal hypertension (PH). Considering the current lack of effective treatments, we evaluated an anti-inflammatory and antioxidant nutraceutical rich in docosahexaenoic acid (DHA) as a possible therapy for ACLD. We investigated the effects of two-week DHA supplementation (500 mg/kg) on hepatic fatty acids, PH, oxidative stress, inflammation, and hepatic stellate cell (HSC) phenotype in rats with ACLD. Additionally, the effects of DHA were evaluated in murine macrophages and human HSC. In contrast to vehicle-treated animals, cirrhotic rats receiving DHA reestablished a healthy hepatic fatty acid profile, which was associated with an improvement in PH. The mechanisms underlying this hemodynamic improvement included a reduction in oxidative stress and inflammation, as well as a marked HSC deactivation, confirmed in human HSC. Experiments with cultured macrophages showed that treatment with DHA protects against pro-inflammatory insults. The present preclinical study demonstrates that a nutraceutical rich in DHA significantly improves PH in chronic liver disease mainly by suppressing inflammation and oxidative stress-driven HSC activation, encouraging its evaluation as a new treatment for PH and cirrhosis.
Collapse
|