1
|
Li F, Wu M, Wang F, Luo L, Wu Z, Huang Z, Wen Z. Unveiling the endocrine connections of NAFLD: evidence from a comprehensive mendelian randomization study. Biomed Eng Lett 2025; 15:239-248. [PMID: 39781064 PMCID: PMC11704114 DOI: 10.1007/s13534-024-00442-8] [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: 08/18/2024] [Revised: 10/17/2024] [Accepted: 10/29/2024] [Indexed: 01/03/2025] Open
Abstract
Background NAFLD is gaining recognition as a complex, multifactorial condition with suspected associations with endocrine disorders. This investigation employed MR analysis to explore the potential causality linking NAFLD to a spectrum of endocrine diseases, encompassing T1D, T2D, obesity, graves' disease, and acromegaly. Methods Our methodology leveraged a stringent IV selection process, adhering to the STROBE-MR guidelines. The MR analysis was conducted utilizing three distinct methods: IVW, WM, and MR-Egger. The IVW method was prioritized as the primary analytical approach. We conducted MR analyses to analyze the causal relationship between NAFLD and metabolic disorders. We also examined 1400 metabolites implicated in NAFLD. Metabolic pathway analysis was performed using the MetaboAnalyst database. Results The findings indicated that T2D (OR = 1.211, 95%CI: 0.836-1.585) and obesity (OR = 1.245, 95%CI: 0.816-1.674) are associated with an increased risk of NAFLD development. Further exploration into the the 1400 metabolites revealed that cys-gly and diacetylornithine are predictive of NAFLD, T2D, and obesity, whereas isovalerylcarnitine exhibited an inverse association, potentially inhibiting disease development. Metabolic pathways involving alanine, aspartate, and glutamate metabolism were identified as pivotal regulators in the pathophysiology of NAFLD, T2D, and obesity. Conclusion The present study generated innovative viewpoints on the etiology of NAFLD. Our findings underscore the significant role of T2D and obesity in NAFLD pathogenesis through metabolic pathways, presenting opportunities for targeted therapeutic strategies and warranting further investigation. Supplementary Information The online version contains supplementary material available at 10.1007/s13534-024-00442-8.
Collapse
Affiliation(s)
- Fan Li
- Department of Gastroenterology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006 China
| | - Mingjun Wu
- Nanchang Hongdu Hospital of Traditional Chinese Medicine, Nanchang, China
| | - Fenfen Wang
- Department of Gastroenterology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006 China
| | - Linfei Luo
- Department of Gastroenterology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006 China
| | - Zhengqiang Wu
- Department of Gastroenterology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006 China
| | - Zixiang Huang
- Department of Gastroenterology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006 China
| | - Zhili Wen
- Department of Gastroenterology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006 China
| |
Collapse
|
2
|
Fogal V, Michopoulos F, Jarnuczak AF, Hamza GM, Harlfinger S, Davey P, Hulme H, Atkinson SJ, Gabrowski P, Cheung T, Grondine M, Hoover C, Rose J, Bray C, Foster AJ, Askin S, Majumder MM, Fitzpatrick P, Miele E, Macdonald R, Keun HC, Coen M. Mechanistic safety assessment via multi-omic characterisation of systemic pathway perturbations following in vivo MAT2A inhibition. Arch Toxicol 2024; 98:2589-2603. [PMID: 38755480 PMCID: PMC11272821 DOI: 10.1007/s00204-024-03771-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 04/24/2024] [Indexed: 05/18/2024]
Abstract
The tumour suppressor p16/CDKN2A and the metabolic gene, methyl-thio-adenosine phosphorylase (MTAP), are frequently co-deleted in some of the most aggressive and currently untreatable cancers. Cells with MTAP deletion are vulnerable to inhibition of the metabolic enzyme, methionine-adenosyl transferase 2A (MAT2A), and the protein arginine methyl transferase (PRMT5). This synthetic lethality has paved the way for the rapid development of drugs targeting the MAT2A/PRMT5 axis. MAT2A and its liver- and pancreas-specific isoform, MAT1A, generate the universal methyl donor S-adenosylmethionine (SAM) from ATP and methionine. Given the pleiotropic role SAM plays in methylation of diverse substrates, characterising the extent of SAM depletion and downstream perturbations following MAT2A/MAT1A inhibition (MATi) is critical for safety assessment. We have assessed in vivo target engagement and the resultant systemic phenotype using multi-omic tools to characterise response to a MAT2A inhibitor (AZ'9567). We observed significant SAM depletion and extensive methionine accumulation in the plasma, liver, brain and heart of treated rats, providing the first assessment of both global SAM depletion and evidence of hepatic MAT1A target engagement. An integrative analysis of multi-omic data from liver tissue identified broad perturbations in pathways covering one-carbon metabolism, trans-sulfuration and lipid metabolism. We infer that these pathway-wide perturbations represent adaptive responses to SAM depletion and confer a risk of oxidative stress, hepatic steatosis and an associated disturbance in plasma and cellular lipid homeostasis. The alterations also explain the dramatic increase in plasma and tissue methionine, which could be used as a safety and PD biomarker going forward to the clinic.
Collapse
Affiliation(s)
- Valentina Fogal
- Oncology Safety, Safety Sciences, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Cambridge, UK
- Cancer Metabolism & Systems Toxicology Group, Division of Cancer, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Filippos Michopoulos
- Bioscience, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Andrew F Jarnuczak
- Data Sciences & Quantitative Biology, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Ghaith M Hamza
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, R&D Boston, Waltham, USA
| | | | - Paul Davey
- Chemistry, Oncology R&D AstraZeneca, Cambridge, UK
| | - Heather Hulme
- Imaging and Data Analytics, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | | | - Piotr Gabrowski
- Biological Insights Knowledge Graph, R&D IT, AstraZeneca, Barcelona, Spain
| | - Tony Cheung
- Oncology R&D, AstraZeneca, R&D Boston, Waltham, USA
| | | | - Clare Hoover
- Oncology Safety Pathology, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, R&D Boston, Waltham, USA
| | - Jonathan Rose
- Animal Science & Technologies, R&D, AstraZeneca, Cambridge, UK
| | - Chandler Bray
- Cancer Metabolism & Systems Toxicology Group, Division of Cancer, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Alison J Foster
- Regulatory Toxicology and Safety Pharmacology, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Sean Askin
- Advanced Drug Delivery, Pharmaceutical Sci, R&D, AstraZeneca, Cambridge, UK
| | - Muntasir Mamun Majumder
- Safety Sciences, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Paul Fitzpatrick
- Safety Sciences, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Eric Miele
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, R&D Boston, Waltham, USA
| | - Ruth Macdonald
- Animal Science & Technologies, R&D, AstraZeneca, Cambridge, UK
| | - Hector C Keun
- Cancer Metabolism & Systems Toxicology Group, Division of Cancer, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Muireann Coen
- Oncology Safety, Safety Sciences, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Cambridge, UK.
| |
Collapse
|
3
|
Ungvari A, Gulej R, Csik B, Mukli P, Negri S, Tarantini S, Yabluchanskiy A, Benyo Z, Csiszar A, Ungvari Z. The Role of Methionine-Rich Diet in Unhealthy Cerebrovascular and Brain Aging: Mechanisms and Implications for Cognitive Impairment. Nutrients 2023; 15:4662. [PMID: 37960316 PMCID: PMC10650229 DOI: 10.3390/nu15214662] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 10/28/2023] [Accepted: 10/31/2023] [Indexed: 11/15/2023] Open
Abstract
As aging societies in the western world face a growing prevalence of vascular cognitive impairment and Alzheimer's disease (AD), understanding their underlying causes and associated risk factors becomes increasingly critical. A salient concern in the western dietary context is the high consumption of methionine-rich foods such as red meat. The present review delves into the impact of this methionine-heavy diet and the resultant hyperhomocysteinemia on accelerated cerebrovascular and brain aging, emphasizing their potential roles in cognitive impairment. Through a comprehensive exploration of existing evidence, a link between high methionine intake and hyperhomocysteinemia and oxidative stress, mitochondrial dysfunction, inflammation, and accelerated epigenetic aging is drawn. Moreover, the microvascular determinants of cognitive deterioration, including endothelial dysfunction, reduced cerebral blood flow, microvascular rarefaction, impaired neurovascular coupling, and blood-brain barrier (BBB) disruption, are explored. The mechanisms by which excessive methionine consumption and hyperhomocysteinemia might drive cerebromicrovascular and brain aging processes are elucidated. By presenting an intricate understanding of the relationships among methionine-rich diets, hyperhomocysteinemia, cerebrovascular and brain aging, and cognitive impairment, avenues for future research and potential therapeutic interventions are suggested.
Collapse
Affiliation(s)
- Anna Ungvari
- Department of Public Health, Semmelweis University, 1089 Budapest, Hungary
| | - Rafal Gulej
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (R.G.); (B.C.); (P.M.); (S.N.); (S.T.); (A.Y.); (A.C.); (Z.U.)
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Boglarka Csik
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (R.G.); (B.C.); (P.M.); (S.N.); (S.T.); (A.Y.); (A.C.); (Z.U.)
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- International Training Program in Geroscience, Department of Public Health, Doctoral School of Basic and Translational Medicine, Semmelweis University, 1089 Budapest, Hungary
| | - Peter Mukli
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (R.G.); (B.C.); (P.M.); (S.N.); (S.T.); (A.Y.); (A.C.); (Z.U.)
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- International Training Program in Geroscience, Department of Public Health, Doctoral School of Basic and Translational Medicine, Semmelweis University, 1089 Budapest, Hungary
| | - Sharon Negri
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (R.G.); (B.C.); (P.M.); (S.N.); (S.T.); (A.Y.); (A.C.); (Z.U.)
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Stefano Tarantini
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (R.G.); (B.C.); (P.M.); (S.N.); (S.T.); (A.Y.); (A.C.); (Z.U.)
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- International Training Program in Geroscience, Department of Public Health, Doctoral School of Basic and Translational Medicine, Semmelweis University, 1089 Budapest, Hungary
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK 73104, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (R.G.); (B.C.); (P.M.); (S.N.); (S.T.); (A.Y.); (A.C.); (Z.U.)
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- International Training Program in Geroscience, Department of Public Health, Doctoral School of Basic and Translational Medicine, Semmelweis University, 1089 Budapest, Hungary
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK 73104, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Zoltan Benyo
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary;
- Cerebrovascular and Neurocognitive Disorders Research Group, Eötvös Loránd Research Network, Semmelweis University, 1094 Budapest, Hungary
| | - Anna Csiszar
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (R.G.); (B.C.); (P.M.); (S.N.); (S.T.); (A.Y.); (A.C.); (Z.U.)
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK 73104, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- International Training Program in Geroscience, Department of Translational Medicine, Doctoral School of Basic and Translational Medicine, Semmelweis University, 1089 Budapest, Hungary
| | - Zoltan Ungvari
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (R.G.); (B.C.); (P.M.); (S.N.); (S.T.); (A.Y.); (A.C.); (Z.U.)
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- International Training Program in Geroscience, Department of Public Health, Doctoral School of Basic and Translational Medicine, Semmelweis University, 1089 Budapest, Hungary
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK 73104, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
4
|
Zheng L, Liu Y, Zhang Y, Xu B, Sagada G, Wang Z, Chen C, Lang X, Zhang J, Shao Q. Comparative study on the effects of crystalline L-methionine and methionine hydroxy analogue calcium supplementations in the diet of juvenile Pacific white shrimp ( Litopenaeus vannamei). Front Physiol 2023; 14:1067354. [PMID: 36793420 PMCID: PMC9923173 DOI: 10.3389/fphys.2023.1067354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 01/17/2023] [Indexed: 01/31/2023] Open
Abstract
An 8-week feeding trial was conducted to evaluate the effects of L-methionine and methionine hydroxy analogue calcium (MHA-Ca) supplements in low-fishmeal diet on growth performance, hepatopancreas morphology, protein metabolism, anti-oxidative capacity, and immunity of Pacific white shrimp (Litopena eus vannamei). Four isonitrogenous and isoenergetic diets were designed: PC (203.3 g/kg fishmeal), NC (100 g/kg fishmeal), MET (100 g/kg fishmeal +3 g/kg L-methionine) and MHA-Ca (100 g/kg fishmeal +3 g/kg MHA-Ca). White shrimp (initial body weight 0.23 ± 0.00 g, 50 shrimp per tank) were allocated to 12 tanks and divided among 4 treatments in triplicates. In response to L-methionine and MHA-Ca supplementations, the shrimp exhibited higher weight gain rate (WGR), specific growth rate (SGR), condition factor (CF), and lower hepatosomatic index (HSI) compared to those fed the NC diet (p < 0.05). The WGR and SGR of shrimp fed L-methionine and MHA-Ca showed no difference with those in the PC diet (p > 0.05). Both of L-methionine and MHA-Ca supplementary diets significantly decreased the malondialdehyde (MDA) levels of shrimp when compared with the NC diet (p < 0.05). L-methionine supplementation improved the lysozyme (LZM) activity and total antioxidant capacity (T-AOC) of shrimp, while the MHA-Ca addition elevated the reduced glutathione (GSH) levels in comparison with those fed the NC diet (p < 0.05). Hypertrophied blister cells in hepatocytes were observed in shrimp fed the NC diet, and alleviated with L-methionine and MHA-Ca supplementations. Shrimp fed the MET and MHA-Ca diets had higher mRNA expression levels of target of rapamycin (tor) than those fed the NC diet (p < 0.05). Compared to the NC group, dietary MHA-Ca supplementation upregulated the expression level of cysteine dioxygenase (cdo) (p < 0.05), while L-methionine supplementation had no significant impact (p > 0.05). The expression levels of superoxide dismutase (sod) and glutathione peroxidase (gpx) were significantly upregulated by L-methionine supplemented diet in comparison with those in the NC group (p < 0.05). Overall, the addition of both L-methionine and MHA-Ca elevated the growth performance, facilitated protein synthesis, and ameliorated hepatopancreatic damage induced by plant-protein enriched diet in L. vannamei. L-methionine and MHA-Ca supplements enhanced anti-oxidants differently.
Collapse
Affiliation(s)
- Lu Zheng
- Aqua-feed and Nutrition Laboratory, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Yuechong Liu
- Aqua-feed and Nutrition Laboratory, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Yanmei Zhang
- Aqua-feed and Nutrition Laboratory, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Bingying Xu
- Aqua-feed and Nutrition Laboratory, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Gladstone Sagada
- Aqua-feed and Nutrition Laboratory, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Zhixuan Wang
- Shandong NHU Amino Acid Co., Ltd, Weifang, China
| | - Cong Chen
- Shandong NHU Amino Acid Co., Ltd, Weifang, China
| | | | - Jiaonan Zhang
- Fujian Province Key Laboratory of Special Aquatic Formula Feed, Fujian Tianma Science and Technology Co., Ltd, Fuqing, China
| | - Qingjun Shao
- Aqua-feed and Nutrition Laboratory, College of Animal Sciences, Zhejiang University, Hangzhou, China,*Correspondence: Qingjun Shao,
| |
Collapse
|
5
|
Liang H, Ji K, Ge X, Zhu J, Ren M, Mi H. Methionine played a positive role in improving the intestinal digestion capacity, anti-inflammatory reaction and oxidation resistance of grass carp, Ctenopharyngodon idella, fry. FISH & SHELLFISH IMMUNOLOGY 2022; 128:389-397. [PMID: 35940539 DOI: 10.1016/j.fsi.2022.07.066] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 07/20/2022] [Accepted: 07/20/2022] [Indexed: 06/15/2023]
Abstract
A study was carried out to appraisal the function of methionine on intestinal digestion and the health of grass carp (Ctenopharyngodon idella) fry (initial weight 0.36 ± 0.01 g). The fry were fed graded dietary methionine levels (0.33%-1.20% dry matter) in 18 recirculatory tanks (180 L). After an 8-week breeding experiment, the results revealed that 0.71%-1.20% dietary methionine levels markedly upregulated the mRNA levels of intestinal digestion including trypsin, amylase, chymotrypsin and AKP, and 0.71%-0.87% dietary methionine level significantly increased intestinal trypsin activities compared with the 0.33% dietary methionine level. For inflammation, 0.71%-1.20% dietary methionine levels downregulated the mRNA levels of NF-κBp65, IL-1β, IL-6, IL-8, IL-15 and IL-17D, whereas upregulated the mRNA levels of anti-inflammatory cytokines, including IL-4/13B, IL-10 and IL-11. In terms of antioxidants, although dietary methionine levels had no significant effect on the expression of most core genes of the Nrf2/ARE signaling pathway, such as Nrf2, Keap 1, GPx4, CAT, Cu/Zn-SOD. Furthermore, dietary methionine levels had no significant effect on the expression of p38MAPK, IL-12p35, TGF-β2 and IL-4/13A. 0.71%-1.20% dietary methionine levels still increased the mRNA levels of GPx1α, GSTR and GSTP1. Furthermore, higher intestinal catalase activity and glutathione contents were also observed in fry fed 0.71%-1.20% diets. In summary, 0.71%-1.20% dietary methionine levels played a positive role in improving the intestinal digestion capacity of digestion, anti-inflammatory reaction and oxidation resistance of grass carp fry. This study provided a theoretical basis for improving the survival rate and growth of grass carp fry.
Collapse
Affiliation(s)
- Hualiang Liang
- Key Laboratory of Integrated Rice-Fish Farming Ecology, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, 214081, China
| | - Ke Ji
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, 214081, China
| | - Xianping Ge
- Key Laboratory of Integrated Rice-Fish Farming Ecology, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, 214081, China; Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, 214081, China
| | - Jian Zhu
- Key Laboratory of Integrated Rice-Fish Farming Ecology, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, 214081, China; Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, 214081, China
| | - Mingchun Ren
- Key Laboratory of Integrated Rice-Fish Farming Ecology, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, 214081, China; Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, 214081, China.
| | - Haifeng Mi
- Tongwei Co, Ltd, Healthy Aquaculture Key Laboratory of Sichuan Province, Chengdu, 610093, China.
| |
Collapse
|
6
|
Effects of dietary supplementation with tropical microfungi Mucor circinelloides and Trichoderma harzianum powder on growth performance, bacterial communities, and the biomass nutritional profile of white shrimp grow-out culture (Litopenaeus vannamei). AQUACULTURE AND FISHERIES 2022. [DOI: 10.1016/j.aaf.2022.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
7
|
Alshawsh MA, Alsalahi A, Alshehade SA, Saghir SAM, Ahmeda AF, Al Zarzour RH, Mahmoud AM. A Comparison of the Gene Expression Profiles of Non-Alcoholic Fatty Liver Disease between Animal Models of a High-Fat Diet and Methionine-Choline-Deficient Diet. Molecules 2022; 27:molecules27030858. [PMID: 35164140 PMCID: PMC8839835 DOI: 10.3390/molecules27030858] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 12/12/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) embraces several forms of liver disorders involving fat disposition in hepatocytes ranging from simple steatosis to the severe stage, namely, non-alcoholic steatohepatitis (NASH). Recently, several experimental in vivo animal models for NAFLD/NASH have been established. However, no reproducible experimental animal model displays the full spectrum of pathophysiological, histological, molecular, and clinical features associated with human NAFLD/NASH progression. Although methionine-choline-deficient (MCD) diet and high-fat diet (HFD) models can mimic histological and metabolic abnormalities of human disease, respectively, the molecular signaling pathways are extremely important for understanding the pathogenesis of the disease. This review aimed to assess the differences in gene expression patterns and NAFLD/NASH progression pathways among the most common dietary animal models, i.e., HFD- and MCD diet-fed animals. Studies showed that the HFD and MCD diet could induce either up- or downregulation of the expression of genes and proteins that are involved in lipid metabolism, inflammation, oxidative stress, and fibrogenesis pathways. Interestingly, the MCD diet model could spontaneously develop liver fibrosis within two to four weeks and has significant effects on the expression of genes that encode proteins and enzymes involved in the liver fibrogenesis pathway. However, such effects in the HFD model were found to occur after 24 weeks with insulin resistance but appear to cause less severe fibrosis. In conclusion, assessing the abnormal gene expression patterns caused by different diet types provides valuable information regarding the molecular mechanisms of NAFLD/NASH and predicts the clinical progression of the disease. However, expression profiling studies concerning genetic variants involved in the development and progression of NAFLD/NASH should be conducted.
Collapse
Affiliation(s)
- Mohammed Abdullah Alshawsh
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia;
- Correspondence:
| | - Abdulsamad Alsalahi
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia;
| | - Salah Abdalrazak Alshehade
- Discipline of Pharmacology, School of Pharmaceutical Sciences, Universiti Sains Malaysia (USM), Gelugor 11800, Malaysia; (S.A.A.); (R.H.A.Z.)
| | - Sultan Ayesh Mohammed Saghir
- Department of Medical Analysis, Princess Aisha Bint Al-Hussein College of Nursing and Medical Sciences, Al-Hussein Bin Talal University, Ma’an 71111, Jordan;
| | - Ahmad Faheem Ahmeda
- Department of Basic Medical Sciences, College of Medicine, Ajman University, Ajman P.O. Box 346, United Arab Emirates;
- Center of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman P.O. Box 346, United Arab Emirates
| | - Raghdaa Hamdan Al Zarzour
- Discipline of Pharmacology, School of Pharmaceutical Sciences, Universiti Sains Malaysia (USM), Gelugor 11800, Malaysia; (S.A.A.); (R.H.A.Z.)
| | - Ayman Moawad Mahmoud
- Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, Beni-Suef 62514, Egypt;
| |
Collapse
|
8
|
Hadchouel A, Drummond D, Pontoizeau C, Aoust L, Hurtado Nedelec MM, El Benna J, Gachelin E, Perisson C, Vigier C, Schiff M, Lacaille F, Molina TJ, Berteloot L, Renolleau S, Ottolenghi C, Tréluyer JM, de Blic J, Delacourt C. Methionine supplementation for multi-organ dysfunction in MetRS-related pulmonary alveolar proteinosis. Eur Respir J 2021; 59:13993003.01554-2021. [PMID: 34503986 DOI: 10.1183/13993003.01554-2021] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 08/17/2021] [Indexed: 11/09/2022]
Abstract
INTRODUCTION Pulmonary alveolar proteinosis related to mutations in the methionine tRNA synthetase (MARS1) gene is a severe, early-onset disease that results in death before the age of 2 years in one-third of patients. It is associated with a liver disease, growth failure and systemic inflammation. As methionine supplementation in yeast models restored normal enzymatic activity of the synthetase, we studied the tolerance, safety and efficacy of daily oral methionine supplementation in patients with severe and early disease. METHODS Four patients received methionine supplementation and were followed for respiratory, hepatic, growth, and inflammation-related outcomes. Their course was compared to those of historical controls. Reactive oxygen species (ROS) production by patient monocytes before and after methionine supplementation was also studied. RESULTS Methionine supplementation was associated with respiratory improvement, clearance of the extracellular lipoproteinaceous material, and discontinuation of whole-lung lavage in all patients. The three patients who required oxygen or non-invasive ventilation could be weaned off within 60 days. Liver dysfunction, inflammation, and growth delay also improved or resolved. At a cellular level, methionine supplementation normalised the production of reactive oxygen species by peripheral monocytes. CONCLUSION Methionine supplementation was associated with important improvements in children with pulmonary alveolar proteinosis related to mutations in the MARS1 gene. This study paves the way for similar strategies for other tRNA synthetase deficiencies.
Collapse
Affiliation(s)
- Alice Hadchouel
- AP-HP, Service de Pneumologie Pédiatrique, Hôpital Universitaire Necker-Enfants Malades, Centre de Référence pour les Maladies Respiratoires Rares de l'Enfant, Paris, France .,Faculté de Médecine, Université de Paris, Paris, France
| | - David Drummond
- AP-HP, Service de Pneumologie Pédiatrique, Hôpital Universitaire Necker-Enfants Malades, Centre de Référence pour les Maladies Respiratoires Rares de l'Enfant, Paris, France.,Faculté de Médecine, Université de Paris, Paris, France
| | - Clément Pontoizeau
- Faculté de Médecine, Université de Paris, Paris, France.,AP-HP, UF de Métabolomique, Hôpital Universitaire Necker-Enfants Malades, Paris, France
| | - Laura Aoust
- AP-HP, Service de Pneumologie Pédiatrique, Hôpital Universitaire Necker-Enfants Malades, Centre de Référence pour les Maladies Respiratoires Rares de l'Enfant, Paris, France.,Faculté de Médecine, Université de Paris, Paris, France
| | - Maria-Margarita Hurtado Nedelec
- INSERM-U1149, Faculté de Médecine, Centre de Recherche sur l'Inflammation (CRI), CNRS-ERL8252, Laboratoire d'Excellence Inflamex, Université Paris Diderot-Sorbonne Paris Cité, , Paris, France.,AP-HP, UF Dysfonctionnements Immunitaires, Centre Hospitalier Universitaire Xavier Bichat, Paris, France
| | - Jamel El Benna
- INSERM-U1149, Faculté de Médecine, Centre de Recherche sur l'Inflammation (CRI), CNRS-ERL8252, Laboratoire d'Excellence Inflamex, Université Paris Diderot-Sorbonne Paris Cité, , Paris, France
| | - Elsa Gachelin
- Service de Pédiatrie, CHU Reunion site Félix Guyon, Saint Denis, France
| | | | | | - Manuel Schiff
- AP-HP, Service de Maladies Héréditaires du Métabolisme, Hôpital Necker-Enfants Malades, Centre de Référence Maladies Héréditaires du Métabolisme, Paris, France.,Institut Imagine, Inserm UMRS 1163, Paris, France
| | - Florence Lacaille
- AP-HP, Service de Gastroentérologie-Hépatologie-Nutrition Pédiatrique, Hôpital Necker-Enfants Malades, Paris, France
| | - Thierry Jo Molina
- Institut Imagine, Inserm UMRS 1163, Paris, France.,AP-HP, Service de Pathologie, Hôpital Universitaire Necker-Enfants Malades, Paris, France
| | - Laureline Berteloot
- Institut Imagine, Inserm UMRS 1163, Paris, France.,AP-HP, Service d'Imagerie Pédiatrique, Hôpital Universitaire Necker-Enfants Malades, Paris, France
| | - Sylvain Renolleau
- Faculté de Médecine, Université de Paris, Paris, France.,AP-HP, Service de Réanimation médico-chirurgicale pédiatrique, Hôpital Universitaire Necker-Enfants Malades, Paris, France
| | - Chris Ottolenghi
- Faculté de Médecine, Université de Paris, Paris, France.,AP-HP, UF de Métabolomique, Hôpital Universitaire Necker-Enfants Malades, Paris, France
| | - Jean-Marc Tréluyer
- Faculté de Médecine, Université de Paris, Paris, France.,Groupe Hospitalier APHP Centre Université de Paris Recherche Clinique et Pharmacologie Necker Cochin, Paris, France
| | - Jacques de Blic
- AP-HP, Service de Pneumologie Pédiatrique, Hôpital Universitaire Necker-Enfants Malades, Centre de Référence pour les Maladies Respiratoires Rares de l'Enfant, Paris, France.,Faculté de Médecine, Université de Paris, Paris, France.,Authors contributed equally to this article
| | - Christophe Delacourt
- AP-HP, Service de Pneumologie Pédiatrique, Hôpital Universitaire Necker-Enfants Malades, Centre de Référence pour les Maladies Respiratoires Rares de l'Enfant, Paris, France.,Faculté de Médecine, Université de Paris, Paris, France.,Authors contributed equally to this article
| |
Collapse
|
9
|
Tillmann S, Awwad HM, MacPherson CW, Happ DF, Treccani G, Geisel J, Tompkins TA, Ueland PM, Wegener G, Obeid R. The Kynurenine Pathway Is Upregulated by Methyl-deficient Diet and Changes Are Averted by Probiotics. Mol Nutr Food Res 2021; 65:e2100078. [PMID: 33686786 DOI: 10.1002/mnfr.202100078] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/17/2021] [Indexed: 12/16/2022]
Abstract
SCOPE Probiotics exert immunomodulatory effects and may influence tryptophan metabolism in the host. Deficiency of nutrients related to C1 metabolism might stimulate inflammation by enhancing the kynurenine pathway. This study used Sprague Dawley rats to investigate whether a methyl-deficient diet (MDD) may influence tryptophan/kynurenine pathways and cytokines and whether probiotics can mitigate these effects. METHODS AND RESULTS Rats are fed a control or MDD diet. Animals on the MDD diet received vehicle, probiotics (L. helveticus R0052 and B. longum R0175), choline, or probiotics + choline for 10 weeks (n = 10 per group). Concentrations of plasma kynurenine metabolites and the methylation and inflammatory markers in plasma and liver are measured. RESULTS MDD animals (vs controls) show upregulation of plasma kynurenine, kynurenic acid, xanthurenic acid, 3-hydroxyxanthranilic acid, quinolinic acid, nicotinic acid, and nicotinamide (all p < 0.05). In the MDD rats, the probiotics (vs vehicle) cause lower anthranilic acid and a trend towards lower kynurenic acid and picolinic acid. Compared to probiotics alone, probiotics + choline is associated with a reduced enrichment of the bacterial strains in cecum. The interventions have no effect on inflammatory markers. CONCLUSIONS Probiotics counterbalance the effect of MDD diet and downregulate downstream metabolites of the kynurenine pathway.
Collapse
Affiliation(s)
- Sandra Tillmann
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus C, DK-8000, Denmark
| | - Hussain M Awwad
- Department of Clinical Chemistry and Laboratory Medicine, Saarland University Hospital, Building 57, Homburg/Saar, D-66421, Germany
| | - Chad W MacPherson
- Rosell Institute for Microbiome and Probiotics, Montreal, Quebec, Canada
| | - Denise F Happ
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus C, DK-8000, Denmark
| | - Giulia Treccani
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus C, DK-8000, Denmark
| | - Juergen Geisel
- Department of Clinical Chemistry and Laboratory Medicine, Saarland University Hospital, Building 57, Homburg/Saar, D-66421, Germany
| | - Thomas A Tompkins
- Rosell Institute for Microbiome and Probiotics, Montreal, Quebec, Canada
| | - Per Magne Ueland
- Department of Clinical Science, University of Bergen, New Lab Building, 9th floor, Bergen, Hordaland, 5021, Norway
| | - Gregers Wegener
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus C, DK-8000, Denmark
| | - Rima Obeid
- Department of Clinical Chemistry and Laboratory Medicine, Saarland University Hospital, Building 57, Homburg/Saar, D-66421, Germany.,Aarhus Institute of Advanced Studies, Aarhus University, Aarhus C, DK-8000, Denmark
| |
Collapse
|
10
|
Soret PA, Magusto J, Housset C, Gautheron J. In Vitro and In Vivo Models of Non-Alcoholic Fatty Liver Disease: A Critical Appraisal. J Clin Med 2020; 10:jcm10010036. [PMID: 33374435 PMCID: PMC7794936 DOI: 10.3390/jcm10010036] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/22/2020] [Accepted: 12/23/2020] [Indexed: 02/07/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD), including non-alcoholic fatty liver (NAFL) and non-alcoholic steatohepatitis (NASH), represents the hepatic manifestation of obesity and metabolic syndrome. Due to the spread of the obesity epidemic, NAFLD is becoming the most common chronic liver disease and one of the principal indications for liver transplantation. However, no pharmacological treatment is currently approved to prevent the outbreak of NASH, which leads to fibrosis and cirrhosis. Preclinical research is required to improve our knowledge of NAFLD physiopathology and to identify new therapeutic targets. In the present review, we summarize advances in NAFLD preclinical models from cellular models, including new bioengineered platforms, to in vivo models, with a particular focus on genetic and dietary mouse models. We aim to discuss the advantages and limits of these different models.
Collapse
Affiliation(s)
- Pierre-Antoine Soret
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, Inserm, 75012 Paris, France; (P.-A.S.); (J.M.); (C.H.)
- Assistance Publique-Hôpitaux de Paris (AP-HP), Hepatology Department, Reference Center for Inflammatory Biliary Diseases and Autoimmune Hepatitis, Saint-Antoine Hospital, 75012 Paris, France
| | - Julie Magusto
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, Inserm, 75012 Paris, France; (P.-A.S.); (J.M.); (C.H.)
- Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne Université, Inserm, AP-HP, 75013 Paris, France
| | - Chantal Housset
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, Inserm, 75012 Paris, France; (P.-A.S.); (J.M.); (C.H.)
- Assistance Publique-Hôpitaux de Paris (AP-HP), Hepatology Department, Reference Center for Inflammatory Biliary Diseases and Autoimmune Hepatitis, Saint-Antoine Hospital, 75012 Paris, France
- Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne Université, Inserm, AP-HP, 75013 Paris, France
| | - Jérémie Gautheron
- Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, Inserm, 75012 Paris, France; (P.-A.S.); (J.M.); (C.H.)
- Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne Université, Inserm, AP-HP, 75013 Paris, France
- Correspondence:
| |
Collapse
|
11
|
Peng C, Stewart AG, Woodman OL, Ritchie RH, Qin CX. Non-Alcoholic Steatohepatitis: A Review of Its Mechanism, Models and Medical Treatments. Front Pharmacol 2020; 11:603926. [PMID: 33343375 PMCID: PMC7745178 DOI: 10.3389/fphar.2020.603926] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 10/19/2020] [Indexed: 12/11/2022] Open
Abstract
Non-alcoholic steatohepatitis (NASH) develops from non-alcoholic fatty liver disease (NAFLD). Currently, around 25% of the population is estimated to have NAFLD, and 25% of NAFLD patients are estimated to have NASH. NASH is typically characterized by liver steatosis inflammation, and fibrosis driven by metabolic disruptions such as obesity, diabetes, and dyslipidemia. NASH patients with significant fibrosis have increased risk of developing cirrhosis and liver failure. Currently, NASH is the second leading cause for liver transplant in the United States. More importantly, the risk of developing hepatocellular carcinoma from NASH has also been highlighted in recent studies. Patients may have NAFLD for years before progressing into NASH. Although the pathogenesis of NASH is not completely understood, the current “multiple-hits” hypothesis suggests that in addition to fat accumulation, elevated oxidative and ER stress may also drive liver inflammation and fibrosis. The development of clinically relevant animal models and pharmacological treatments for NASH have been hampered by the limited understanding of the disease mechanism and a lack of sensitive, non-invasive diagnostic tools. Currently, most pre-clinical animal models are divided into three main groups which includes: genetic models, diet-induced, and toxin + diet-induced animal models. Although dietary models mimic the natural course of NASH in humans, the models often only induce mild liver injury. Many genetic and toxin + diet-induced models rapidly induce the development of metabolic disruption and serious liver injury, but not without their own shortcomings. This review provides an overview of the “multiple-hits” hypothesis and an evaluation of the currently existing animal models of NASH. This review also provides an update on the available interventions for managing NASH as well as pharmacological agents that are currently undergoing clinical trials for the treatment of NASH.
Collapse
Affiliation(s)
- Cheng Peng
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Melbourne, VIC, Australia.,Baker Heart & Diabetes Institute, Melbourne, VIC, Australia.,Department of Pharmacology and Therapeutics, University of Melbourne, Melbourne, VIC, Australia
| | - Alastair G Stewart
- Department of Pharmacology and Therapeutics, University of Melbourne, Melbourne, VIC, Australia.,Australian Research Council, Centre for Personalised Therapeutics Technologies, Lancaster, CBR, Australia
| | - Owen L Woodman
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Melbourne, VIC, Australia
| | - Rebecca H Ritchie
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Melbourne, VIC, Australia.,Baker Heart & Diabetes Institute, Melbourne, VIC, Australia.,Department of Pharmacology and Therapeutics, University of Melbourne, Melbourne, VIC, Australia
| | - Cheng Xue Qin
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Melbourne, VIC, Australia.,Baker Heart & Diabetes Institute, Melbourne, VIC, Australia.,Department of Pharmacology and Therapeutics, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
12
|
LaBranche TP, Kopec AK, Mantena SR, Hollingshead BD, Harrington AW, Stewart ZS, Zhan Y, Hayes KD, Whiteley LO, Burdick AD, Davis JW. Zucker Lean Rats With Hepatic Steatosis Recapitulate Asymptomatic Metabolic Syndrome and Exhibit Greater Sensitivity to Drug-Induced Liver Injury Compared With Standard Nonclinical Sprague-Dawley Rat Model. Toxicol Pathol 2020; 48:994-1007. [PMID: 33252024 DOI: 10.1177/0192623320968716] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Fatty liver disease is a potential risk factor for drug-induced liver injury (DILI). Despite advances in nonclinical in vitro and in vivo models to assess liver injury during drug development, the pharmaceutical industry is still plagued by idiosyncratic DILI. Here, we tested the hypothesis that certain features of asymptomatic metabolic syndrome (namely hepatic steatosis) increase the risk for DILI in certain phenotypes of the human population. Comparison of the Zucker Lean (ZL) and Zucker Fatty rats fed a high fat diet (HFD) revealed that HFD-fed ZL rats developed mild hepatic steatosis with compensatory hyperinsulinemia without increases in liver enzymes. We then challenged steatotic HFD-fed ZL rats and Sprague-Dawley (SD) rats fed normal chow, a nonclinical model widely used in the pharmaceutical industry, with acetaminophen overdose to induce liver injury. Observations in HFD-fed ZL rats included increased liver injury enzymes and greater incidence and severity of hepatic necrosis compared with similarly treated SD rats. The HFD-fed ZL rats also had disproportionately higher hepatic drug accumulation, which was linked with abnormal hepatocellular efflux transporter distribution. Here, we identify ZL rats with HFD-induced hepatic steatosis as a more sensitive nonclinical in vivo test system for modeling DILI compared with SD rats fed normal chow.
Collapse
Affiliation(s)
- Timothy P LaBranche
- 376392Pfizer Inc, Cambridge, MA, USA.,Blueprint Medicines, Cambridge, MA, USA.,*Timothy P. LaBranche and Anna K. Kopec contributed equally
| | - Anna K Kopec
- 2253Pfizer Inc, Groton, CT, USA.,*Timothy P. LaBranche and Anna K. Kopec contributed equally
| | | | | | - Andrew W Harrington
- 2253Pfizer Inc, Chesterfield, MO, USA.,Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St Louis, MO, USA
| | - Zachary S Stewart
- 2253Pfizer Inc, Andover, MA, USA.,Hooke Laboratories, Lawrence, MA, USA
| | | | - Kyle D Hayes
- 2253Pfizer Inc, Andover, MA, USA.,Mallinckrodt Pharmaceuticals, Hazelwood, MO, USA
| | | | | | - John W Davis
- 376392Pfizer Inc, Cambridge, MA, USA.,Dyne Therapeutics, Waltham, MA, USA
| |
Collapse
|
13
|
Miyachi Y, Yagi S, Hirata M, Iwamura S, Yao S, Shirai H, Okumura S, Iida T, Ito T, Uozumi R, Kaido T, Uemoto S. Etiology of Liver Steatosis Influences the Severity of Ischemia/Reperfusion Injury and Survival After Liver Transplantation in Rats. Liver Transpl 2020; 26:1504-1515. [PMID: 32511857 DOI: 10.1002/lt.25814] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 04/27/2020] [Accepted: 05/21/2020] [Indexed: 01/13/2023]
Abstract
Liver steatosis is a leading cause of graft disposal in liver transplantation, though the degree of steatosis is often the single factor determining acceptability of the graft. We investigated how the cause of liver steatosis affects graft function in rat orthotopic liver transplantation (OLT). OLT was performed using 2 types of steatotic liver grafts: the fasting and hyperalimentation (FHA) model and the methionine- and choline-deficient diet models. The FHA and 4-week feeding of a methionine- and choline-deficient diet (MCDD4wk) groups showed similar liver triglyceride levels without signs of steatohepatitis. Therefore, the 2 groups were compared in the following experiment. With 6-hour cold storage, the 7-day survival rate after OLT was far worse in the FHA than in the MCDD4wk group (0% versus 100%, P = 0.002). With 1-hour cold storage, the FHA group showed higher aspartate aminotransferase and alanine aminotransferase levels and histological injury scores in zones 1 and 2 at 24 hours after reperfusion than the normal liver and MCDD4wk groups. Intrahepatic microcirculation and tissue adenosine triphosphate levels were significantly lower in the FHA group after reperfusion. Hepatocyte necrosis, sinusoidal endothelial cell injury, and abnormal swelling of the mitochondria were also found in the FHA group after reperfusion. Tissue malondialdehyde levels were higher in the MCDD4wk group before and after reperfusion. However, the grafts up-regulated several antioxidant enzymes soon after reperfusion. Even though the degree of steatosis was equivalent, the 2 liver steatosis models possessed quite unique basal characteristics and showed completely different responses against ischemia/reperfusion injury and survival after transplantation. Our results demonstrate that the degree of fat accumulation is not a single determinant for the usability of steatotic liver grafts.
Collapse
Affiliation(s)
- Yosuke Miyachi
- Division of Hepato-Biliary-Pancreatic and Transplant Surgery, Department of Surgery, Kyoto University, Kyoto, Japan
| | - Shintaro Yagi
- Division of Hepato-Biliary-Pancreatic and Transplant Surgery, Department of Surgery, Kyoto University, Kyoto, Japan
| | - Masaaki Hirata
- Division of Hepato-Biliary-Pancreatic and Transplant Surgery, Department of Surgery, Kyoto University, Kyoto, Japan
| | - Sena Iwamura
- Division of Hepato-Biliary-Pancreatic and Transplant Surgery, Department of Surgery, Kyoto University, Kyoto, Japan
| | - Siyuan Yao
- Division of Hepato-Biliary-Pancreatic and Transplant Surgery, Department of Surgery, Kyoto University, Kyoto, Japan
| | - Hisaya Shirai
- Division of Hepato-Biliary-Pancreatic and Transplant Surgery, Department of Surgery, Kyoto University, Kyoto, Japan
| | - Shinya Okumura
- Division of Hepato-Biliary-Pancreatic and Transplant Surgery, Department of Surgery, Kyoto University, Kyoto, Japan
| | - Taku Iida
- Division of Hepato-Biliary-Pancreatic and Transplant Surgery, Department of Surgery, Kyoto University, Kyoto, Japan
| | - Takashi Ito
- Division of Hepato-Biliary-Pancreatic and Transplant Surgery, Department of Surgery, Kyoto University, Kyoto, Japan
| | - Ryuji Uozumi
- Biomedical Statistics and Bioinformatics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Toshimi Kaido
- Division of Hepato-Biliary-Pancreatic and Transplant Surgery, Department of Surgery, Kyoto University, Kyoto, Japan
| | - Shinji Uemoto
- Division of Hepato-Biliary-Pancreatic and Transplant Surgery, Department of Surgery, Kyoto University, Kyoto, Japan
| |
Collapse
|
14
|
Liu S, Wang X, Zhang R, Song M, Zhang N, Li W, Wang Y, Xu Y, Zhang L. Amino acid, fatty acid, and carbohydrate metabolomic profiles with ginsenoside-induced insecticidal efficacy against Ostrinia furnacalis (Guenee). J Ginseng Res 2020; 44:544-551. [PMID: 32617034 PMCID: PMC7322809 DOI: 10.1016/j.jgr.2019.04.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 03/14/2019] [Accepted: 04/23/2019] [Indexed: 01/22/2023] Open
Abstract
Background Previous studies have shown the insecticidal efficacy of ginsenosides. In the present study, we aimed to investigate the metabolic mechanism related to the inhibitory effect of panaxadiol saponins (PDSs) against the Asian corn borer Ostrinia furnacalis (Guenee). Methods Third instar larvae of O. furnacalis were fed normal diets with different concentrations of PDSs for 4 days. The consumption index, relative growth rate, approximate digestibility, and conversion of ingested and digested food were recorded. A targeted gas chromatography–mass spectrometry assay was performed to detect the profiles of amino acids, fatty acids, and carbohydrates in larvae of O. furnacalis. In addition, the activity of detoxification-related enzymes was determined. Results and Conclusions PDSs decreased the consumption index, relative growth rate, approximate digestibility, and conversion of ingested and digested food in the 3rd instar larvae of O. furnacalis in a dose-dependent manner. PDSs decreased 15 free amino acids, 16 free fatty acids, and 5 carbohydrates and increased the levels of palmitoleic acid, palmitic acid, and 9-octadecenoic acid in the 3rd instar larvae. The activity of detoxification-related enzymes, such as acetylcholinesterase, glutathione S-transferase, cytochrome P450, carboxylesterase, trehalase, acid phosphatase, and alkaline phosphatase, was reduced in a dose-dependent manner in the 3rd instar larvae exposed to PDSs. These data confirmed the inhibitory effect of PDSs against growth, food utilization, and detoxification in the 3rd instar larvae of O. furnacalis and the potential for using PDSs as an efficient tool for insect pest management for O. furnacalis larvae. Panaxadiol saponins (PDSs) reduced larval growth and food consumption and utilization in Ostrinia furnacalis. PDSs reduced the levels of free amino acids, fatty acids, and sugar in larvae. PDSs inhibited the activity of acetylcholinesterase, glutathione S-transferase, cytochrome P450, carboxylesterase, trehalase, acid phosphatase, and alkaline phosphatase. All the inhibitory effects of PDSs against O. furnacalis larvae were dose dependent.
Collapse
Affiliation(s)
- Shuangli Liu
- National & Local Joint Engineering Research Center for Ginseng Breeding and Application (Jilin), Jilin Agricultural University, China
| | - Xiaohui Wang
- Research Center of Agricultural Environment and Resources, Jilin Academy of Agricultural Sciences, China
| | - Rui Zhang
- National & Local Joint Engineering Research Center for Ginseng Breeding and Application (Jilin), Jilin Agricultural University, China
| | - Mingjie Song
- National & Local Joint Engineering Research Center for Ginseng Breeding and Application (Jilin), Jilin Agricultural University, China
| | - Nanqi Zhang
- National & Local Joint Engineering Research Center for Ginseng Breeding and Application (Jilin), Jilin Agricultural University, China
| | - Wanying Li
- National & Local Joint Engineering Research Center for Ginseng Breeding and Application (Jilin), Jilin Agricultural University, China
| | - Yingping Wang
- National & Local Joint Engineering Research Center for Ginseng Breeding and Application (Jilin), Jilin Agricultural University, China
| | - Yonghua Xu
- National & Local Joint Engineering Research Center for Ginseng Breeding and Application (Jilin), Jilin Agricultural University, China
- Corresponding author. 2888 Xincheng Street, Changchun City, Jilin Province, China.
| | - Lianxue Zhang
- National & Local Joint Engineering Research Center for Ginseng Breeding and Application (Jilin), Jilin Agricultural University, China
- Corresponding author. 2888 Xincheng Street, Changchun City, Jilin Province, China.
| |
Collapse
|
15
|
Rubio C, Puerto M, García-Rodríquez JJ, Lu VB, García-Martínez I, Alén R, Sanmartín-Salinas P, Toledo-Lobo MV, Saiz J, Ruperez J, Barbas C, Menchén L, Gribble FM, Reimann F, Guijarro LG, Carrascosa JM, Valverde ÁM. Impact of global PTP1B deficiency on the gut barrier permeability during NASH in mice. Mol Metab 2020; 35:100954. [PMID: 32244182 PMCID: PMC7082558 DOI: 10.1016/j.molmet.2020.01.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 01/23/2020] [Accepted: 01/28/2020] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE Non-alcoholic steatohepatitis (NASH) is characterized by a robust pro-inflammatory component at both hepatic and systemic levels together with a disease-specific gut microbiome signature. Protein tyrosine phosphatase 1 B (PTP1B) plays distinct roles in non-immune and immune cells, in the latter inhibiting pro-inflammatory signaling cascades. In this study, we have explored the role of PTP1B in the composition of gut microbiota and gut barrier dynamics in methionine and choline-deficient (MCD) diet-induced NASH in mice. METHODS Gut features and barrier permeability were characterized in wild-type (PTP1B WT) and PTP1B-deficient knockout (PTP1B KO) mice fed a chow or methionine/choline-deficient (MCD) diet for 4 weeks. The impact of inflammation was studied in intestinal epithelial and enteroendocrine cells. The secretion of GLP-1 was evaluated in primary colonic cultures and plasma of mice. RESULTS We found that a shift in the gut microbiota shape, disruption of gut barrier function, higher levels of serum bile acids, and decreased circulating glucagon-like peptide (GLP)-1 are features during NASH. Surprisingly, despite the pro-inflammatory phenotype of global PTP1B-deficient mice, they were partly protected against the alterations in gut microbiota composition during NASH and presented better gut barrier integrity and less permeability under this pathological condition. These effects concurred with higher colonic mucosal inflammation, decreased serum bile acids, and protection against the decrease in circulating GLP-1 levels during NASH compared with their WT counterparts together with increased expression of GLP-2-sensitive genes in the gut. At the molecular level, stimulation of enteroendocrine STC-1 cells with a pro-inflammatory conditioned medium (CM) from lipopolysaccharide (LPS)-stimulated macrophages triggered pro-inflammatory signaling cascades that were further exacerbated by a PTP1B inhibitor. Likewise, the pro-inflammatory CM induced GLP-1 secretion in primary colonic cultures, an effect augmented by PTP1B inhibition. CONCLUSION Altogether our results have unraveled a potential role of PTP1B in the gut-liver axis during NASH, likely mediated by increased sensitivity to GLPs, with potential therapeutic value.
Collapse
Affiliation(s)
- Carmen Rubio
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Madrid, Spain; Centro de Biología Molecular Severo Ochoa (CBMSO, CSIC-UAM), Madrid, Spain
| | - Marta Puerto
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; CIBER de Enfermedades Hepáticas y Digestivas (CIBERHED), ISCIII, Madrid, Spain
| | - Juan J García-Rodríquez
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Universidad Complutense, Madrid, Spain
| | - Van B Lu
- Wellcome Trust MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Irma García-Martínez
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Madrid, Spain
| | - Rosa Alén
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Madrid, Spain
| | | | - M Val Toledo-Lobo
- Departamento de Biología de Sistemas, Universidad de Alcalá de Henares, Madrid, Spain
| | - Jorge Saiz
- CEMBIO, Universidad San Pablo-CEU, Madrid, Spain
| | | | - Coral Barbas
- CEMBIO, Universidad San Pablo-CEU, Madrid, Spain
| | - Luis Menchén
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; CIBER de Enfermedades Hepáticas y Digestivas (CIBERHED), ISCIII, Madrid, Spain; Departamento de Medicina, Facultad de Medicina, Universidad Complutense de Madrid, Spain
| | - Fiona M Gribble
- Wellcome Trust MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Frank Reimann
- Wellcome Trust MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Luis G Guijarro
- Departamento de Biología de Sistemas, Universidad de Alcalá de Henares, Madrid, Spain
| | - Jose M Carrascosa
- Centro de Biología Molecular Severo Ochoa (CBMSO, CSIC-UAM), Madrid, Spain.
| | - Ángela M Valverde
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Madrid, Spain.
| |
Collapse
|
16
|
Ward NP, DeNicola GM. Sulfur metabolism and its contribution to malignancy. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2019; 347:39-103. [PMID: 31451216 DOI: 10.1016/bs.ircmb.2019.05.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Metabolic dysregulation is an appreciated hallmark of cancer and a target for therapeutic intervention. Cellular metabolism involves a series of oxidation/reduction (redox) reactions that yield the energy and biomass required for tumor growth. Cells require diverse molecular species with constituent sulfur atoms to facilitate these processes. For humans, this sulfur is derived from the dietary consumption of the proteinogenic amino acids cysteine and methionine, as only lower organisms (e.g., bacteria, fungi, and plants) can synthesize them de novo. In addition to providing the sulfur required to sustain redox chemistry, the metabolism of these sulfur-containing amino acids yield intermediate metabolites that constitute the cellular antioxidant system, mediate inter- and intracellular signaling, and facilitate the epigenetic regulation of gene expression, all of which contribute to tumorigenesis.
Collapse
Affiliation(s)
- Nathan P Ward
- Department of Cancer Physiology, Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| | - Gina M DeNicola
- Department of Cancer Physiology, Moffitt Cancer Center and Research Institute, Tampa, FL, United States.
| |
Collapse
|
17
|
Heo G, Ko KS. Long-Term Feeding of Soy Protein Attenuates Choline Deficient-Induced Adverse Effects in Wild Type Mice and Prohibitin 1 Deficient Mice Response More Sensitively. Prev Nutr Food Sci 2019; 24:32-40. [PMID: 31008094 PMCID: PMC6456240 DOI: 10.3746/pnf.2019.24.1.32] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 01/31/2019] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease, however the exact cause of NAFLD remains unknown. Methionine, an essential amino acid, is the first limiting amino acid of soy protein, and its deficiency is suggested to cause hepatocyte damage and NAFLD. The objective of this study is to examine the changes in NAFLD susceptibility with soy protein consumption and deterioration due to prohibitin 1 (PHB1) deficiency, an important protein in hepatic mitochondrial function. In this study, liver-specific phb1 +/- mice and wild-type mice were fed a normal diet, choline-deficient diet (CDD), or soy protein diet without choline (SPD) for 16 weeks. Using hematoxylin and eosin staining, we showed that SPD attenuates symptoms of hepatocyte damage and lipid accumulation induced by CDD in mouse liver. The liver damage in mice fed the SPD was alleviated by decreasing lipogenic markers and by increasing anti-inflammatory markers. Furthermore, mRNA expression of genes involved in hepatic methionine metabolism was significantly lower in liver-specific phb1 +/- mice fed with a SPD compared with wild-type mice fed with a SPD. These data suggest a CDD can cause non-alcohol related liver damage, which can be attenuated by a SPD in wild-type mice. These phenomena were not observed in liver-specific phb1 +/- mice. It may therefore be concluded that SPD attenuates CDD-induced liver damage in wild-type mice, and that PHB1 deficiency blocks the beneficial effects of SPD against CDD-induced liver damage.
Collapse
Affiliation(s)
- Gieun Heo
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul 03760, Korea
| | - Kwang Suk Ko
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul 03760, Korea
| |
Collapse
|
18
|
Mafra D, Esgalhado M, Borges NA, Cardozo LFMF, Stockler-Pinto MB, Craven H, Buchanan SJ, Lindholm B, Stenvinkel P, Shiels PG. Methyl Donor Nutrients in Chronic Kidney Disease: Impact on the Epigenetic Landscape. J Nutr 2019; 149:372-380. [PMID: 30796783 DOI: 10.1093/jn/nxy289] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 08/23/2018] [Accepted: 10/26/2018] [Indexed: 12/28/2022] Open
Abstract
Epigenetic alterations, such as those linked to DNA methylation, may potentially provide molecular explanations for complications associated with altered gene expression in illnesses, such as chronic kidney disease (CKD). Although both DNA hypo- and hypermethylation have been observed in the uremic milieu, this remains only a single aspect of the epigenetic landscape and, thus, of any biochemical dysregulation associated with CKD. Nevertheless, the role of uremia-promoting alterations on the epigenetic landscape regulating gene expression is still a novel and scarcely studied field. Although few studies have actually reported alterations of DNA methylation via methyl donor nutrient intake, emerging evidence indicates that nutritional modification of the microbiome can affect one-carbon metabolism and the capacity to methylate the genome in CKD. In this review, we discuss the nutritional modifications that may affect one-carbon metabolism and the possible impact of methyl donor nutrients on the microbiome, CKD, and its phenotype.
Collapse
Affiliation(s)
- Denise Mafra
- Post Graduation Program in Medical Sciences.,Post Graduation Program in Cardiovascular Sciences
| | | | - Natalia A Borges
- Post Graduation Program in Cardiovascular Sciences.,Post Graduation Program in Nutrition Sciences, Federal Fluminense University (UFF), Niterói-Rio de Janeiro (RJ), Brazil
| | | | - Milena B Stockler-Pinto
- Post Graduation Program in Cardiovascular Sciences.,Post Graduation Program in Nutrition Sciences, Federal Fluminense University (UFF), Niterói-Rio de Janeiro (RJ), Brazil
| | - Hannah Craven
- Wolfson Wohl Translational Research Centre, Institute of Cancer Sciences, University of Glasgow, Bearsden, Glasgow, UK
| | - Sarah J Buchanan
- Wolfson Wohl Translational Research Centre, Institute of Cancer Sciences, University of Glasgow, Bearsden, Glasgow, UK
| | - Bengt Lindholm
- Division of Renal Medicine and Baxter Novum, Department of Clinical Science, Technology and Intervention, Karolinska Institutet, Stockholm, Sweden
| | - Peter Stenvinkel
- Division of Renal Medicine and Baxter Novum, Department of Clinical Science, Technology and Intervention, Karolinska Institutet, Stockholm, Sweden
| | - Paul G Shiels
- Wolfson Wohl Translational Research Centre, Institute of Cancer Sciences, University of Glasgow, Bearsden, Glasgow, UK
| |
Collapse
|
19
|
Wu B, Li L, Ruan T, Peng X. Effect of methionine deficiency on duodenal and jejunal IgA + B cell count and immunoglobulin level of broilers. IRANIAN JOURNAL OF VETERINARY RESEARCH 2018; 19:165-171. [PMID: 30349561 PMCID: PMC6184025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 01/28/2018] [Accepted: 02/17/2018] [Indexed: 06/08/2023]
Abstract
Dietary methionine (met) is reported to enhance antibody production and boost cell-mediated immunity in chickens. Methionine deficiency has been shown to affect the development of the lymphoid organs and the generation of antibodies in chickens. This study is designed to investigate the effects of met deficiency on IgA+ B cells and immunoglobulins (sIgA, IgA, IgG and IgM) for a 6 week period in the duodenum and jejunum of Cobb broiler chicken using immunohistochemistry and enzyme-linked immunosorbent assay (ELISA) techniques. The results of the study showed that the IgA+ B cell count reduced significantly in the met deficiency group compared to the control group (P<0.05 or P<0.01). The contents of sIgA, IgA, IgG and IgM in the met deficiency group were significantly decreased (P<0.05 or P<0.01), especially at 28 and 42 days of age. It can be concluded that met deficiency exerts significant effects on the humoral immune system of intestinal mucosa. This study has provided valuable experimental insight which could be useful for future studies on the function of met in the intestine of humans and other animals.
Collapse
Affiliation(s)
- B. Wu
- Department of Animal Pathology, College of Life Science, China West Normal University, Sichuan, China
- Key Laboratory of Southwest China Wildlife Resources Conservation, Ministry of Education, Sichuan, China
| | - L. Li
- BSc in Animal Pathology, Department of Animal Pathology, College of Life Science, China West Normal University, Sichuan, China
| | - T. Ruan
- BSc in Animal Pathology, Department of Animal Pathology, College of Life Science, China West Normal University, Sichuan, China
| | - X. Peng
- Department of Animal Pathology, College of Life Science, China West Normal University, Sichuan, China
- Key Laboratory of Southwest China Wildlife Resources Conservation, Ministry of Education, Sichuan, China
| |
Collapse
|
20
|
Oxidation Resistance of the Sulfur Amino Acids: Methionine and Cysteine. BIOMED RESEARCH INTERNATIONAL 2017; 2017:9584932. [PMID: 29445748 PMCID: PMC5763110 DOI: 10.1155/2017/9584932] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 11/20/2017] [Indexed: 02/08/2023]
Abstract
Sulfur amino acids are a kind of amino acids which contain sulfhydryl, and they play a crucial role in protein structure, metabolism, immunity, and oxidation. Our review demonstrates the oxidation resistance effect of methionine and cysteine, two of the most representative sulfur amino acids, and their metabolites. Methionine and cysteine are extremely sensitive to almost all forms of reactive oxygen species, which makes them antioxidative. Moreover, methionine and cysteine are precursors of S-adenosylmethionine, hydrogen sulfide, taurine, and glutathione. These products are reported to alleviate oxidant stress induced by various oxidants and protect the tissue from the damage. However, the deficiency and excess of methionine and cysteine in diet affect the normal growth of animals; thereby a new study about defining adequate levels of methionine and cysteine intake is important.
Collapse
|
21
|
Samala N, Tersey SA, Chalasani N, Anderson RM, Mirmira RG. Molecular mechanisms of nonalcoholic fatty liver disease: Potential role for 12-lipoxygenase. J Diabetes Complications 2017; 31:1630-1637. [PMID: 28886991 PMCID: PMC5643240 DOI: 10.1016/j.jdiacomp.2017.07.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 07/25/2017] [Accepted: 07/26/2017] [Indexed: 02/07/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a spectrum of pathologies associated with fat accumulation in the liver. NAFLD is the most common cause of liver disease in the United States, affecting up to a third of the general population. It is commonly associated with features of metabolic syndrome, particularly insulin resistance. NAFLD shares the basic pathogenic mechanisms with obesity and insulin resistance, such as mitochondrial, oxidative and endoplasmic reticulum stress. Lipoxygenases catalyze the conversion of poly-unsaturated fatty acids in the plasma membrane-mainly arachidonic acid and linoleic acid-to produce oxidized pro-inflammatory lipid intermediates. 12-Lipoxygenase (12-LOX) has been studied extensively in setting of inflammation and insulin resistance. As insulin resistance is closely associated with development of NAFLD, the role of 12-LOX in pathogenesis of NAFLD has received increasing attention in recent years. In this review we discuss the role of 12-LOX in NAFLD pathogenesis and its potential role in emerging new therapeutics.
Collapse
Affiliation(s)
- Niharika Samala
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Sarah A Tersey
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Naga Chalasani
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Ryan M Anderson
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Raghavendra G Mirmira
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
22
|
Dual role of protein tyrosine phosphatase 1B in the progression and reversion of non-alcoholic steatohepatitis. Mol Metab 2017; 7:132-146. [PMID: 29126873 PMCID: PMC5784331 DOI: 10.1016/j.molmet.2017.10.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 10/19/2017] [Accepted: 10/22/2017] [Indexed: 12/13/2022] Open
Abstract
Objectives Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease in Western countries. Protein tyrosine phosphatase 1B (PTP1B), a negative modulator of insulin and cytokine signaling, is a therapeutic target for type 2 diabetes and obesity. We investigated the impact of PTP1B deficiency during NAFLD, particularly in non-alcoholic steatohepatitis (NASH). Methods NASH features were evaluated in livers from wild-type (PTP1BWT) and PTP1B-deficient (PTP1BKO) mice fed methionine/choline-deficient diet (MCD) for 8 weeks. A recovery model was established by replacing MCD to chow diet (CHD) for 2–7 days. Non-parenchymal liver cells (NPCs) were analyzed by flow cytometry. Oval cells markers were measured in human and mouse livers with NASH, and in oval cells from PTP1BWT and PTP1BKO mice. Results PTP1BWT mice fed MCD for 8 weeks exhibited NASH, NPCs infiltration, and elevated Fgf21, Il6 and Il1b mRNAs. These parameters decreased after switching to CHD. PTP1B deficiency accelerated MCD-induced NASH. Conversely, after switching to CHD, PTP1BKO mice rapidly reverted NASH compared to PTP1BWT mice in parallel to the normalization of serum triglycerides (TG) levels. Among NPCs, a drop in cytotoxic natural killer T (NKT) subpopulation was detected in PTP1BKO livers during recovery, and in these conditions M2 macrophage markers were up-regulated. Oval cells markers (EpCAM and cytokeratin 19) significantly increased during NASH only in PTP1B-deficient livers. HGF-mediated signaling and proliferative capacity were enhanced in PTP1BKO oval cells. In NASH patients, oval cells markers were also elevated. Conclusions PTP1B elicits a dual role in NASH progression and reversion. Additionally, our results support a new role for PTP1B in oval cell proliferation during NAFLD. PTP1B deficiency accelerates MCD-induced NASH. The liver inflammatory responses during NASH are enhanced in PTP1B-deficient mice. PTP1B deficiency accelerates the reversion of NASH in a recovery dietary model. In a DCC model PTP1BKO livers increased oval cells markers and proliferative capacity. PTP1B deficiency enhances HGF-mediated signaling and proliferation of oval cells.
Collapse
|
23
|
Wankhade UD, Zhong Y, Kang P, Alfaro M, Chintapalli SV, Thakali KM, Shankar K. Enhanced offspring predisposition to steatohepatitis with maternal high-fat diet is associated with epigenetic and microbiome alterations. PLoS One 2017; 12:e0175675. [PMID: 28414763 PMCID: PMC5393586 DOI: 10.1371/journal.pone.0175675] [Citation(s) in RCA: 138] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 03/29/2017] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE Non-alcoholic fatty liver disease (NAFLD) is an important co-morbidity associated with obesity and a precursor to steatohepatitis. However, the contributions of gestational and early life influences on development of NAFLD and NASH remain poorly appreciated. METHODS Two independent studies were performed to examine whether maternal over-nutrition via exposure to high fat diet (HFD) leads to exacerbated hepatic responses to post-natal HFD and methionine choline deficient (MCD) diets in the offspring. Offspring of both control diet- and HFD-fed dams were weaned onto control and HFD, creating four groups. RESULTS When compared to their control diet-fed littermates, offspring of HF-dams weaned onto HFD gained greater body weight; had increased relative liver weight and showed hepatic steatosis and inflammation. Similarly, this group revealed significantly greater immune response and pro-fibrogenic gene expression via RNA-seq. In parallel, 7-8 week old offspring were challenged with either control or MCD diets for 3 weeks. Responses to MCD diets were also exacerbated due to maternal HFD as seen by gene expression of classical pro-fibrogenic genes. Quantitative genome-scale DNA methylation analysis of over 1 million CpGs showed persistent epigenetic changes in key genes in tissue development and metabolism (Fgf21, Ppargc1β) with maternal HFD and in cell adhesion and communication (VWF, Ephb2) in the combination of maternal HFD and offspring MCD diets. Maternal HFD also influenced gut microbiome profiles in offspring leading to a decrease in α-diversity. Linear regression analysis revealed association between serum ALT levels and Coprococcus, Coriobacteriacae, Helicobacterioceae and Allobaculum. CONCLUSION Our findings indicate that maternal HFD detrimentally alters epigenetic and gut microbiome pathways to favor development of fatty liver disease and its progressive sequelae.
Collapse
Affiliation(s)
- Umesh D. Wankhade
- Arkansas Children’s Nutrition Center, Little Rock, Arkansas, United States of America
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Ying Zhong
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Ping Kang
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Maria Alfaro
- Molecular Genetic Pathology Laboratory, Arkansas Children’s Hospital, Little Rock, Arkansas, United States of America
| | - Sree V. Chintapalli
- Arkansas Children’s Nutrition Center, Little Rock, Arkansas, United States of America
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Keshari M. Thakali
- Arkansas Children’s Nutrition Center, Little Rock, Arkansas, United States of America
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Kartik Shankar
- Arkansas Children’s Nutrition Center, Little Rock, Arkansas, United States of America
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| |
Collapse
|
24
|
|
25
|
Croce AC, Ferrigno A, Di Pasqua LG, Berardo C, Piccolini VM, Bertone V, Bottiroli G, Vairetti M. Autofluorescence discrimination of metabolic fingerprint in nutritional and genetic fatty liver models. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2016; 164:13-20. [DOI: 10.1016/j.jphotobiol.2016.09.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 09/09/2016] [Accepted: 09/10/2016] [Indexed: 12/21/2022]
|
26
|
Abstract
Metabolic liver injury is one of the fastest growing health problems worldwide. Alcoholic and non-alcoholic fatty livers have been shown to be associated with progression to end-stage liver diseases, as well as to liver cancers, in humans. More importantly, there are no validated therapies for these disorders, therefore intensive research is required in this area. This review of standard operation procedures focuses on the experimental models of fatty liver disease in the mouse. Firstly, use of these experimental models might improve understanding of underlying mechanisms, and secondly this might help to test potential therapeutic options. This article includes, besides a short historic background, an insight into the pathobiochemical mechanisms and detailed experimental procedures as well as the practical implementation of these models.
Collapse
Affiliation(s)
- P Ramadori
- Department of Internal Medicine III, RWTH University Hospital Aachen, Aachen, Germany
| | - R Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, RWTH Aachen University, Aachen, Germany
| | - J Trebicka
- Department of Internal Medicine I, University of Bonn, Bonn, Germany
| | - K Streetz
- Department of Internal Medicine III, RWTH University Hospital Aachen, Aachen, Germany
| |
Collapse
|
27
|
Machado MV, Michelotti GA, Xie G, de Almeida TP, Boursier J, Bohnic B, Guy CD, Diehl AM. Mouse models of diet-induced nonalcoholic steatohepatitis reproduce the heterogeneity of the human disease. PLoS One 2015; 10:e0127991. [PMID: 26017539 PMCID: PMC4446215 DOI: 10.1371/journal.pone.0127991] [Citation(s) in RCA: 241] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 04/22/2015] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND AIMS Non-alcoholic steatohepatitis (NASH), the potentially progressive form of nonalcoholic fatty liver disease (NAFLD), is the pandemic liver disease of our time. Although there are several animal models of NASH, consensus regarding the optimal model is lacking. We aimed to compare features of NASH in the two most widely-used mouse models: methionine-choline deficient (MCD) diet and Western diet. METHODS Mice were fed standard chow, MCD diet for 8 weeks, or Western diet (45% energy from fat, predominantly saturated fat, with 0.2% cholesterol, plus drinking water supplemented with fructose and glucose) for 16 weeks. Liver pathology and metabolic profile were compared. RESULTS The metabolic profile associated with human NASH was better mimicked by Western diet. Although hepatic steatosis (i.e., triglyceride accumulation) was also more severe, liver non-esterified fatty acid content was lower than in the MCD diet group. NASH was also less severe and less reproducible in the Western diet model, as evidenced by less liver cell death/apoptosis, inflammation, ductular reaction, and fibrosis. Various mechanisms implicated in human NASH pathogenesis/progression were also less robust in the Western diet model, including oxidative stress, ER stress, autophagy deregulation, and hedgehog pathway activation. CONCLUSION Feeding mice a Western diet models metabolic perturbations that are common in humans with mild NASH, whereas administration of a MCD diet better models the pathobiological mechanisms that cause human NAFLD to progress to advanced NASH.
Collapse
Affiliation(s)
- Mariana Verdelho Machado
- Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, NC 27710, United States of America
- Gastroenterology Department, Hospital de Santa Maria, CHLN, Lisbon, Portugal
| | - Gregory Alexander Michelotti
- Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, NC 27710, United States of America
| | - Guanhua Xie
- Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, NC 27710, United States of America
| | - Thiago Pereira de Almeida
- Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, NC 27710, United States of America
| | - Jerome Boursier
- Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, NC 27710, United States of America
| | - Brittany Bohnic
- Division of Endocrinology, Duke University Medical Center, Durham, NC 27710, United States of America
| | - Cynthia D. Guy
- Division of Pathology, Duke University Medical Center, Durham, NC 27710, United States of America
| | - Anna Mae Diehl
- Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, NC 27710, United States of America
| |
Collapse
|
28
|
Das S, Alhasson F, Dattaroy D, Pourhoseini S, Seth RK, Nagarkatti M, Nagarkatti PS, Michelotti GA, Diehl AM, Kalyanaraman B, Chatterjee S. NADPH Oxidase-Derived Peroxynitrite Drives Inflammation in Mice and Human Nonalcoholic Steatohepatitis via TLR4-Lipid Raft Recruitment. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:1944-57. [PMID: 25989356 DOI: 10.1016/j.ajpath.2015.03.024] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 03/04/2015] [Accepted: 03/26/2015] [Indexed: 12/12/2022]
Abstract
The molecular events that link NADPH oxidase activation and the induction of Toll-like receptor (TLR)-4 recruitment into hepatic lipid rafts in nonalcoholic steatohepatitis (NASH) are unclear. We hypothesized that in liver, NADPH oxidase activation is key in TLR4 recruitment into lipid rafts, which in turn up-regulates NF-κB translocation to the nucleus and subsequent DNA binding, leading to NASH progression. Results from confocal microscopy showed that liver from murine and human NASH had NADPH oxidase activation, which led to the formation of highly reactive peroxynitrite, as shown by 3-nitrotyrosine formation in diseased liver. Expression and recruitment of TLR4 into the lipid rafts were significantly greater in rodent and human NASH. The described phenomenon was NADPH oxidase, p47phox, and peroxynitrite dependent, as liver from p47phox-deficient mice and from mice treated with a peroxynitrite decomposition catalyst [iron(III) tetrakis(p-sulfonatophenyl)porphyrin] or a peroxynitrite scavenger (phenylboronic acid) had markedly less Tlr4 recruitment into lipid rafts. Mechanistically, peroxynitrite-induced TLR4 recruitment was linked to increased IL-1β, sinusoidal injury, and Kupffer cell activation while blocking peroxynitrite-attenuated NASH symptoms. The results strongly suggest that NADPH oxidase-mediated peroxynitrite drove TLR4 recruitment into hepatic lipid rafts and inflammation, whereas the in vivo use of the peroxynitrite scavenger phenylboronic acid, a novel synthetic molecule having high reactivity with peroxynitrite, attenuates inflammatory pathogenesis in NASH.
Collapse
Affiliation(s)
- Suvarthi Das
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, University of South Carolina, Columbia, South Carolina
| | - Firas Alhasson
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, University of South Carolina, Columbia, South Carolina
| | - Diptadip Dattaroy
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, University of South Carolina, Columbia, South Carolina
| | - Sahar Pourhoseini
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, University of South Carolina, Columbia, South Carolina
| | - Ratanesh Kumar Seth
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, University of South Carolina, Columbia, South Carolina
| | - Mitzi Nagarkatti
- Department of Pathology, Microbiology and Immunology, University of South Carolina, Columbia, South Carolina
| | - Prakash S Nagarkatti
- Department of Pathology, Microbiology and Immunology, University of South Carolina, Columbia, South Carolina
| | - Gregory A Michelotti
- Division of Gastroenterology, Department of Medicine, Duke University, Durham, North Carolina
| | - Anna Mae Diehl
- Division of Gastroenterology, Department of Medicine, Duke University, Durham, North Carolina
| | - Balaraman Kalyanaraman
- Department of Biophysics, Free Radical Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Saurabh Chatterjee
- Environmental Health and Disease Laboratory, Department of Environmental Health Sciences, University of South Carolina, Columbia, South Carolina.
| |
Collapse
|
29
|
Polyzos SA, Kountouras J, Tsoukas MA. Circulating homocysteine in nonalcoholic fatty liver disease. Eur J Intern Med 2015; 26:152-3. [PMID: 25676807 DOI: 10.1016/j.ejim.2015.01.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 01/27/2015] [Indexed: 01/28/2023]
Affiliation(s)
- Stergios A Polyzos
- Department of Medicine, Second Medical Clinic, Aristotle University of Thessaloniki, Ippokration Hospital, Thessaloniki, Greece; Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| | - Jannis Kountouras
- Department of Medicine, Second Medical Clinic, Aristotle University of Thessaloniki, Ippokration Hospital, Thessaloniki, Greece
| | - Michael A Tsoukas
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
30
|
Pastore A, Alisi A, di Giovamberardino G, Crudele A, Ceccarelli S, Panera N, Dionisi-Vici C, Nobili V. Plasma levels of homocysteine and cysteine increased in pediatric NAFLD and strongly correlated with severity of liver damage. Int J Mol Sci 2014; 15:21202-14. [PMID: 25407526 PMCID: PMC4264220 DOI: 10.3390/ijms151121202] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 11/10/2014] [Accepted: 11/10/2014] [Indexed: 12/30/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a spectrum of metabolic abnormalities ranging from simple triglyceride accumulation in the hepatocytes to hepatic steatosis with inflammation, ballooning and fibrosis. It has been demonstrated that the pathogenesis of NAFLD involves increased oxidative stress, with consumption of the major cellular antioxidant, glutathione (GSH). Liver has a fundamental role in sulfur compound metabolism, although the data reported on plasma thiols status in NAFLD are conflicting. We recruited 63 NAFLD patients, and we analyzed all plasma thiols, such as homocysteine (Hcy), cysteine (Cys), cysteinylglycine (CysGly) and GSH, by high-performance liquid chromatography (HPLC) with fluorescence detection. Hcy, Cys and CysGly plasma levels increased in NAFLD patients (p < 0.0001); whereas GSH levels were decreased in NAFLD patients when compared to controls (p < 0.0001). On the contrary, patients with steatohepatitis exhibited lower levels of Hcy and Cys than subjects without. Furthermore, a positive correlation was found between Hcy and Cys and the presence of fibrosis in children with NAFLD. Taken together, these data demonstrated a defective hepatic sulfur metabolism in children with NAFLD, and that high levels of Hcy and Cys probably correlates with a pattern of more severe histological liver damage, due to mechanisms that require further studies.
Collapse
Affiliation(s)
- Anna Pastore
- Metabolomics and Proteomics Unit, "Bambino Gesù" Children's Hospital, IRCCS, Rome 00165, Italy.
| | - Anna Alisi
- Liver Research Unit, "Bambino Gesù" Children's Hospital, IRCCS, Rome 00165, Italy.
| | | | - Annalisa Crudele
- Liver Research Unit, "Bambino Gesù" Children's Hospital, IRCCS, Rome 00165, Italy.
| | - Sara Ceccarelli
- Liver Research Unit, "Bambino Gesù" Children's Hospital, IRCCS, Rome 00165, Italy.
| | - Nadia Panera
- Liver Research Unit, "Bambino Gesù" Children's Hospital, IRCCS, Rome 00165, Italy.
| | - Carlo Dionisi-Vici
- Metabolomics and Proteomics Unit, "Bambino Gesù" Children's Hospital, IRCCS, Rome 00165, Italy.
| | - Valerio Nobili
- Liver Research Unit, "Bambino Gesù" Children's Hospital, IRCCS, Rome 00165, Italy.
| |
Collapse
|
31
|
Methionine production—a critical review. Appl Microbiol Biotechnol 2014; 98:9893-914. [DOI: 10.1007/s00253-014-6156-y] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Revised: 10/09/2014] [Accepted: 10/12/2014] [Indexed: 12/31/2022]
|
32
|
Tanaka H, Fukahori S, Baba S, Ueno T, Sivakumar R, Yagi M, Asagiri K, Ishii S, Tanaka Y. Branched-Chain Amino Acid-Rich Supplements Containing Microelements Have Antioxidant Effects on Nonalcoholic Steatohepatitis in Mice. JPEN J Parenter Enteral Nutr 2014; 40:519-28. [PMID: 25316683 DOI: 10.1177/0148607114555160] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 09/19/2014] [Indexed: 12/15/2022]
Abstract
BACKGROUND The aim of the present study was to elucidate whether the administration of antioxidant-rich nutrients, including branched-chain amino acids (BCAAs), microelements, and vitamins, both alone and in combination, has a positive impact on liver function in a nonalcoholic steatohepatitis (NASH) mouse model and identify the mechanisms underlying these effects. METHODS Seven-week-old male KKAy mice fed a methionine- and choline-deficient diet (MCD) for 4 weeks were divided into 7 groups and fed the following planned diets for another 4 weeks: group A (normal diet), group B (MCD; control), group C (MCD with rich microelements), group D (MCD with rich BCAAs), group E (MCD with rich microelements and BCAAs), and group F (MCD with rich microelements, BCAAs, and vitamins). We then conducted biochemical assays, histological analyses, immunohistochemistry for 8-hydroxy-2'-deoxyguanosine (8-OHdG) and 4-hydroxy-2'-nonenal (4-HNE), and Western blotting for insulin glucose signaling, lipid metabolism, and endoplasmic reticulum (ER) stress-related signaling in liver specimens obtained from mice in each group. RESULTS The morphometric grades of all NASH-related findings and the mean degree of 8-OHdG immunolocalization in groups D-F were significantly lower than those observed in group B. The expression levels of insulin receptor β subunit (IRβ) and p-elF in groups E and F and those of phosphatidyl-inositol 3 kinase (PI3K85), p-AcelCoA, and PERK in group F were similar to those noted in group A. CONCLUSIONS The administration of a combination of antioxidant-rich nutrients, including BCAAs and microelements, is likely to suppress the progression of NASH by reducing oxidative stress, primarily via the downregulation of the ER stress pathway.
Collapse
Affiliation(s)
- Hiroaki Tanaka
- Department of Pediatric Surgery, Kurume University School of Medicine, Kurume, Japan
| | - Suguru Fukahori
- Department of Pediatric Surgery, Kurume University School of Medicine, Kurume, Japan
| | - Shinji Baba
- Asakura Medical Association Hospital, Fukuoka, Japan
| | - Takato Ueno
- Asakura Medical Association Hospital, Fukuoka, Japan Research Center for Innovative Cancer Therapy, Kurume University School of Medicine, Kurume, Japan
| | - Ramadoss Sivakumar
- Research Center for Innovative Cancer Therapy, Kurume University School of Medicine, Kurume, Japan
| | - Minoru Yagi
- Department of Pediatric Surgery, Kurume University School of Medicine, Kurume, Japan
| | - Kimio Asagiri
- Department of Pediatric Surgery, Kurume University School of Medicine, Kurume, Japan
| | - Shinji Ishii
- Department of Pediatric Surgery, Kurume University School of Medicine, Kurume, Japan
| | - Yoshiaki Tanaka
- Department of Pediatric Surgery, Kurume University School of Medicine, Kurume, Japan Division of Medical Safety Management, Kurume University School of Medicine, Kurume, Japan
| |
Collapse
|
33
|
Kumagai H, Yokoyama K, Katsuyama K, Hara S, Yamamoto H, Yamagata T, Taniguchi N, Hirota N, Itoh K. A new method for measuring the speed of sound in rat liver ex vivo using an ultrasound system: correlation of sound speed with fat deposition. ULTRASOUND IN MEDICINE & BIOLOGY 2014; 40:2499-2507. [PMID: 25130448 DOI: 10.1016/j.ultrasmedbio.2014.03.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2013] [Revised: 02/04/2014] [Accepted: 03/15/2014] [Indexed: 06/03/2023]
Abstract
The speed of sound correlates well with the fat content of the liver. Therefore, non-invasive quantification of sound speed in the liver might be of diagnostic value. Here we describe a new non-invasive method that would be clinically applicable for measurement of sound speed in the liver. Sprague-Dawley rats were divided into two groups: a control group and a fatty liver group prepared by keeping the rats on a choline-deficient diet for 6 wk. The livers were subjected to pathologic and biochemical analysis; the speed of sound through the liver tissue was measured using our proposed method and a pulser-receiver as standard. Our results indicated that use of the proposed method makes it feasible to diagnose fatty liver with good accuracy on the basis of sound speed. This approach would have considerable potential for non-invasive diagnosis of fatty liver and would be a valuable adjunct to conventional liver diagnostic procedures.
Collapse
Affiliation(s)
- Hideki Kumagai
- Department of Pediatrics, Jichi Medical University, Tochigi, Japan.
| | - Koji Yokoyama
- Department of Pediatrics, Jichi Medical University, Tochigi, Japan
| | - Kimito Katsuyama
- Imaging Technology Center, R&D Management Headquarters, Fujifilm Corporation, Tokyo, Japan
| | - Shoji Hara
- Imaging Technology Center, R&D Management Headquarters, Fujifilm Corporation, Tokyo, Japan
| | - Hiroaki Yamamoto
- Imaging Technology Center, R&D Management Headquarters, Fujifilm Corporation, Tokyo, Japan
| | | | - Nobuyuki Taniguchi
- Department of Clinical Laboratory Medicine, Jichi Medical University, Tochigi, Japan
| | - Norio Hirota
- Hirota Surgical Pathology Institute, Tochigi, Japan
| | | |
Collapse
|
34
|
Herbal medicines for the treatment of nonalcoholic steatohepatitis: current scenario and future prospects. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2014; 2014:648308. [PMID: 24987431 PMCID: PMC4060323 DOI: 10.1155/2014/648308] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 04/30/2014] [Indexed: 12/11/2022]
Abstract
Nonalcoholic steatohepatitis (NASH) is a multifactorial disease and has close correlations with other metabolic disorders. This makes its treatment difficult using a single pharmacological drug. Use of plant extract/decoction or polyherbal formulation to treat various liver diseases is very well mentioned in various traditional systems of medicine (Ayurveda, Japanese or traditional Chinese Medicine, and Kampo medicine). Medicinal herbs are known for their multifaceted implications and thus can form an effective treatment schedule against NASH. Till date, several plant extracts, polyherbal formulations, and phytochemicals have been evaluated for their possible therapeutic potential in preventing onset and progression of NASH in experimental models, but clinical studies using the same are sparse. Herbal extracts with antioxidants, antidiabetic, and antihyperlipidemic properties have been shown to ameliorate symptoms of NASH. This review article is a meticulous compilation of our current knowledge on the role of natural products in alleviating NASH and possible lacunae in research that needs to be addressed.
Collapse
|
35
|
Morphological and functional characterization of non-alcoholic fatty liver disease induced by a methionine-choline-deficient diet in C57BL/6 mice. DISEASE MARKERS 2013; 35:213-9. [PMID: 24167369 PMCID: PMC3776369 DOI: 10.1155/2013/527548] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2013] [Accepted: 08/23/2013] [Indexed: 01/13/2023]
Abstract
Background. The receptor-binding cancer antigen expressed on SiSo cells (RCAS1) is a human tumor-associated antigen that has been considered to play a crucial role in tumor progression by enabling cancer cells to evade immune surveillance. The present study aimed to evaluate the clinical significance of the RCAS1 expression in gastric adenocarcinoma. Material and Methods. RCAS1 protein expression was assessed immunohistochemically on 54 gastric adenocarcinoma tissue samples and was analyzed in relation to clinicopathological parameters, tumor proliferative capacity, and patients' survival. Results. Enhanced RCAS1 expression levels were significantly associated with advanced histopathological stage and presence of organ metastasis (P = 0.0084 and P = 0.0327). Gastric cancer patients with elevated RCAS1 expression levels showed significantly shorter survival times compared to those with low RCAS1 expression (log-rank test, P = 0.0168). In multivariate analysis, histopathological stage and grade of differentiation as well as the RCAS1 expression were identified as independent prognostic factors (Cox regression analysis, P = 0.0204, P = 0.0035, and P = 0.0081). Conclusions. Our data support the evidence that RCAS1 upregulation may contribute to gastric malignant progression, representing a useful biomarker to predict the biological behaviour and prognosis in gastric neoplasia.
Collapse
|
36
|
Itagaki H, Shimizu K, Morikawa S, Ogawa K, Ezaki T. Morphological and functional characterization of non-alcoholic fatty liver disease induced by a methionine-choline-deficient diet in C57BL/6 mice. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2013; 6:2683-2696. [PMID: 24294355 PMCID: PMC3843249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 09/23/2013] [Indexed: 06/02/2023]
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD), including non-alcoholic steatohepatitis (NASH), appears to be increasingly common worldwide. Its histopathology and the effects of nutrition on liver function have not been fully determined. AIM To elucidate the cellular mechanisms of NAFLD induced by a methionine-choline-deficient (MCD) diet in mice. Particular focus was placed on the role of phagocytic cells. METHODS Male C57BL/6 mice were fed an MCD diet for 30 weeks. A recovery model was also established wherein a normal control diet was provided for 2 weeks after a period of 8, 16, or 30 weeks. RESULTS Mice fed the MCD diet for ≥ 2 weeks exhibited severe steatohepatitis with elevated serum aspartate aminotransferase (AST) and alanine aminotransferase (ALT) levels. Steatohepatitis was accompanied by the infiltration of CD68-positive macrophages (Kupffer cells). The severity of steatohepatitis increased in the first 16 weeks but was seen to lessen by week 30. Fibrosis began to develop at 10 weeks and continued thereafter. Steatohepatitis and elevated serum hepatic enzyme concentrations returned to normal levels after switching the diet back to the control within the first 16 weeks, but fibrosis and CD68-positive macrophages remained. CONCLUSIONS The histopathological changes and irreversible fibrosis seen in this model were caused by prolonged feeding of an MCD diet. These results were accompanied by changes in the activity of CD68-positive cells with temporary elevation of CCL-2, MMP-13, and MMP-9 levels, all of which may trigger early steatohepatitis and late fibrosis through phagocytosis-associated MMP induction.
Collapse
Affiliation(s)
- Hiroko Itagaki
- Department of Anatomy and Developmental Biology, Graduate School of Medicine, Tokyo Women’s Medical UniversityTokyo, Japan
| | - Kazuhiko Shimizu
- Department of Anatomy and Developmental Biology, Graduate School of Medicine, Tokyo Women’s Medical UniversityTokyo, Japan
| | - Shunichi Morikawa
- Department of Anatomy and Developmental Biology, Graduate School of Medicine, Tokyo Women’s Medical UniversityTokyo, Japan
| | - Kenji Ogawa
- Department of Surgery, Tokyo Women’s Medical University Medical Center EastTokyo, Japan
| | - Taichi Ezaki
- Department of Anatomy and Developmental Biology, Graduate School of Medicine, Tokyo Women’s Medical UniversityTokyo, Japan
| |
Collapse
|
37
|
Meleshkevitch EA, Voronov DA, Miller MM, Penneda M, Fox JM, Metzler R, Boudko DY. A novel eukaryotic Na+ methionine selective symporter is essential for mosquito development. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2013; 43:755-767. [PMID: 23748165 PMCID: PMC3746589 DOI: 10.1016/j.ibmb.2013.05.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Revised: 05/16/2013] [Accepted: 05/21/2013] [Indexed: 05/30/2023]
Abstract
AeNAT5 (NCBI, ABZ81822), an orphan member of the insect-specific Nutrient Amino acid Transporter subfamily of SoLute Carrier family 6 (NAT-SLC6) and the first representative of a novel eukaryotic methionine-selective transport system (M), was cloned from cDNA of the vector mosquito, Aedes aegypti. It has orphan orthologs throughout several mosquito genomes, but not in Drosophila or outside Diptera. It shows the highest apparent affinity to L-Met (K(0.5) = 0.021 mM) and its metabolites Homocysteine and Cysteine (K(0.5) = 0.89 and 2.16 mM), but weakly interact with other substrates. It has a Na(+) - coupled mechanism (K(0.5) Na(+) ∼ 46 mM) with 1AA:1Na(+) stoichiometry that maintains ∼60% activity in Cl(-) - free media. In situ hybridization showed accumof AeNAT5 transcript in the absorptive and secretory epithelia, as well as in specific peripheral neurons and the central ganglia of mosquito larvae. The labeling pattern is distinct from that of the previously characterized AeNAT1. RNAi of AeNAT5 increases larval mortality during ecdysis and dramatically suppresses adult emergence. Our results showed that in addition to previously characterized broad spectra and aromatic amino acid selective transport systems, the mosquito NAT-SLC6 subfamily evolved a unique mechanism for selective absorption of sulfur-containing substrates. We demonstrated specific patterns of alimentary and neuronal transcription of AeNAT5 in mosquito larvae that is collateral with the indispensable function of this transporter in mosquito development.
Collapse
Affiliation(s)
- Ella A. Meleshkevitch
- Department of Physiology and Biophysics of the Rosalind Franklin University, Chicago Medical School, North Chicago, IL 60064, USA
- Whitney Laboratory for Marine Biosciences, University of Florida, St. Augustine, FL32080, USA
| | - Dmitri A. Voronov
- Institute for Information Transmission Problems, Moscow, 127994, Russia
- A.N. Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow, 119991, Russia
- Whitney Laboratory for Marine Biosciences, University of Florida, St. Augustine, FL32080, USA
| | - Melissa M. Miller
- Whitney Laboratory for Marine Biosciences, University of Florida, St. Augustine, FL32080, USA
| | - Maria Penneda
- Whitney Laboratory for Marine Biosciences, University of Florida, St. Augustine, FL32080, USA
| | - Jeffrey M. Fox
- Department of Physiology and Biophysics of the Rosalind Franklin University, Chicago Medical School, North Chicago, IL 60064, USA
| | - Ryan Metzler
- Department of Physiology and Biophysics of the Rosalind Franklin University, Chicago Medical School, North Chicago, IL 60064, USA
| | - Dmitri Y. Boudko
- Department of Physiology and Biophysics of the Rosalind Franklin University, Chicago Medical School, North Chicago, IL 60064, USA
- Whitney Laboratory for Marine Biosciences, University of Florida, St. Augustine, FL32080, USA
| |
Collapse
|
38
|
Tang HY, Xiao QG, Xu HB, Zhang Y. Optimization of Reaction Parameters for the Synthesis of Chromium Methionine Complex Using Response Surface Methodology. Org Process Res Dev 2013. [DOI: 10.1021/op3002905] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Hai-yan Tang
- National Engineering Laboratory
for Hydrometallurgical Cleaner Production Technology, Institute of Process Engineering, Beijing 100190, P.R.
China
- Key Laboratory of
Green Process
and Engineering, Chinese Academy of Sciences, Beijing 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing 100190, P.R. China
| | - Qing-gui Xiao
- National Engineering Laboratory
for Hydrometallurgical Cleaner Production Technology, Institute of Process Engineering, Beijing 100190, P.R.
China
- Key Laboratory of
Green Process
and Engineering, Chinese Academy of Sciences, Beijing 100190, P.R. China
| | - Hong-bin Xu
- National Engineering Laboratory
for Hydrometallurgical Cleaner Production Technology, Institute of Process Engineering, Beijing 100190, P.R.
China
- Key Laboratory of
Green Process
and Engineering, Chinese Academy of Sciences, Beijing 100190, P.R. China
| | - Yi Zhang
- National Engineering Laboratory
for Hydrometallurgical Cleaner Production Technology, Institute of Process Engineering, Beijing 100190, P.R.
China
- Key Laboratory of
Green Process
and Engineering, Chinese Academy of Sciences, Beijing 100190, P.R. China
| |
Collapse
|
39
|
Non-alcoholic steatohepatitis: an overview including treatments with herbals as alternative therapeutics. J Appl Biomed 2012. [DOI: 10.2478/v10136-012-0008-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
40
|
Dever JT, Elfarra AA. The biochemical and toxicological significance of hypermethionemia: new insights and clinical relevance. Expert Opin Drug Metab Toxicol 2010; 6:1333-46. [PMID: 20874374 DOI: 10.1517/17425255.2010.522177] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
IMPORTANCE OF THE FIELD Disrupted l-methionine (Met) metabolism can lead to hepatic, neurological and cardiovascular dysfunction in humans. Aberrant methyl group flux likely contributes to the development of these pathologies, but when patients also become hypermethionemic, additional toxicological mechanisms may be relevant. AREAS COVERED IN THIS REVIEW Following a discussion of the causes of hypermethionemia in humans, evidence for the toxicological roles and clinical significance of the Met transmethylation (TM), transamination (TA) and sulfoxidation (SO) pathways will be presented. WHAT THE READER WILL GAIN Recent data from freshly isolated mouse hepatocytes (FIMHs) confirmed previous in vivo results in rodents that Met TM is a detoxification pathway while Met TA leads to toxicity. Gender-related differences in Met accumulation and metabolism in FIMHs correlated with gender differences in toxicity. Data obtained from FIMHs also implicated Met SO in Met metabolism and toxicity. Currently, little is known about the mechanisms and biological significance of Met sulfoxidation in humans. TAKE HOME MESSAGE In hypermethionemic patients, clinical and dietary interventions should focus on increasing Met TM and decreasing Met TA and SO. Novel biomarkers of hypermethionemia in humans that correlate with pathological end points are needed to better understand the impact of the condition.
Collapse
Affiliation(s)
- Joseph T Dever
- University of Wisconsin-Madison, Department of Nutritional Sciences, Madison, WI 53706, USA
| | | |
Collapse
|
41
|
Caballero F, Fernández A, Matías N, Martínez L, Fucho R, Elena M, Caballeria J, Morales A, Fernández-Checa JC, García-Ruiz C. Specific contribution of methionine and choline in nutritional nonalcoholic steatohepatitis: impact on mitochondrial S-adenosyl-L-methionine and glutathione. J Biol Chem 2010; 285:18528-36. [PMID: 20395294 DOI: 10.1074/jbc.m109.099333] [Citation(s) in RCA: 182] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The pathogenesis and treatment of nonalcoholic steatohepatitis (NASH) are not well established. Feeding a diet deficient in both methionine and choline (MCD) is one of the most common models of NASH, which is characterized by steatosis, mitochondrial dysfunction, hepatocellular injury, oxidative stress, inflammation, and fibrosis. However, the individual contribution of the lack of methionine and choline in liver steatosis, advanced pathology and impact on mitochondrial S-adenosyl-L-methionine (SAM) and glutathione (GSH), known regulators of disease progression, has not been specifically addressed. Here, we examined the regulation of mitochondrial SAM and GSH and signs of disease in mice fed a MCD, methionine-deficient (MD), or choline-deficient (CD) diet. The MD diet reproduced most of the deleterious effects of MCD feeding, including weight loss, hepatocellular injury, oxidative stress, inflammation, and fibrosis, whereas CD feeding was mainly responsible for steatosis, characterized by triglycerides and free fatty acids accumulation. These findings were preceded by MCD- or MD-mediated SAM and GSH depletion in mitochondria due to decreased mitochondrial membrane fluidity associated with a lower phosphatidylcholine/phosphatidylethanolamine ratio. MCD and MD but not CD feeding resulted in increased ceramide levels by acid sphingomyelinase. Moreover, GSH ethyl ester or SAM therapy restored mitochondrial GSH and ameliorated hepatocellular injury in mice fed a MCD or MD diet. Thus, the depletion of SAM and GSH in mitochondria is an early event in the MCD model of NASH, which is determined by the lack of methionine. Moreover, therapy using permeable GSH prodrugs may be of relevance in NASH.
Collapse
Affiliation(s)
- Francisco Caballero
- Liver Unit, Centro de Investigaciones Biomédicas Esther Koplowitz, Instituto Malalties Digestivas i Metaboliques, Hospital Clínic i Provincial, 08036 Barcelona, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Powell CL, Bradford BU, Craig CP, Tsuchiya M, Uehara T, O'Connell TM, Pogribny IP, Melnyk S, Koop DR, Bleyle L, Threadgill DW, Rusyn I. Mechanism for prevention of alcohol-induced liver injury by dietary methyl donors. Toxicol Sci 2010; 115:131-9. [PMID: 20118189 DOI: 10.1093/toxsci/kfq031] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Alcohol-induced liver injury (ALI) has been associated with, among other molecular changes, abnormal hepatic methionine metabolism, resulting in decreased levels of S-adenosylmethionine (SAM). Dietary methyl donor supplements such as SAM and betaine mitigate ALI in animal models; however, the mechanisms of protection remain elusive. It has been suggested that methyl donors may act via attenuation of alcohol-induced oxidative stress. We hypothesized that the protective action of methyl donors is mediated by an effect on the oxidative metabolism of alcohol in the liver. Male C57BL/6J mice were administered a control high-fat diet or diet enriched in methyl donors with or without alcohol for 4 weeks using the enteral alcohol feeding model. As expected, attenuation of ALI and an increase in reduced glutathione:oxidized glutathione ratio were achieved with methyl donor supplementation. Interestingly, methyl donors led to a 35% increase in blood alcohol elimination rate, and while there was no effect on alcohol metabolism in the stomach, a profound effect on liver alcohol metabolism was observed. The catalase-dependent pathway of alcohol metabolism was induced, yet the increase in CYP2E1 activity by alcohol was blunted, which may be mitigating production of oxidants. Additional factors contributing to the protective effects of methyl donors in ALI were increased activity of low- and high-K(m) aldehyde dehydrogenases leading to lower hepatic acetaldehyde, maintenance of the efficient mitochondrial energy metabolism, and promotion of peroxisomal beta-oxidation. Profound changes in alcohol metabolism represent additional important mechanism of the protective effect of methyl donors in ALI.
Collapse
Affiliation(s)
- Christine L Powell
- Department of Environmental Sciences and Engineering, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Oz HS, Ebersole JL. GREEN TEA POLYPHENOLS MEDIATED APOPTOSIS IN INTESTINAL EPITHELIAL CELLS BY A FADD-DEPENDENT PATHWAY. ACTA ACUST UNITED AC 2010; 1:105-113. [PMID: 21687829 DOI: 10.4236/jct.2010.13018] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Colorectal cancer is the most common malignant complication in patients with chronic inflammatory bowel disease (IBD). In addition, these patients are at risk for developing painful complications during chemotherapy due to cytotoxic effects of drugs currently in use. Past studies have suggested a protective effect of tea consumption on gastrointestinal (GI) malignancies. Green tea polyphenols (GrTP) inhibited carcinogen-induced GI tumors in rodents and induced apoptosis in various carcinoma cell lines. We hypothesized that GrTP and its polyphenolic compounds regulate apoptosis in the intestinal epithelia. In this study, the effects of GrTP and its polyphenolics on apoptosis was evaluated in intestinal epithelial, IEC-6, cells grown to 85% confluency. GrTP (400-800 mg/ml) induced DNA fragmentation in a dose dependent fashion. Higher concentrations (>800 mg/ml) induced a mixed apoptosis and cytolysis. Epithelial cells exposed to GrTP and a major polyphenol, EGCG, but not EGC or EC, increased caspase activities in a time and dose dependent manner. The caspase inhibitors rescued cells from GrTP and EGCG-induced cell death. Concomitantly, GrTP resulted in activation of fatty acid synthase (Fas)-associated protein with death domain (FADD) and recruitment to Fas/CD95 domain 30 minutes following treatment. While GrTP also blocked NF-κB activation, an NFκ-B inhibitor (MG132) only promoted cytolysis. In conclusion, these data demonstrated GrTP and EGCG induced apoptosis in intestinal epithelia mediated by caspase-8 through a FADD dependent pathway. Future investigation may warrant preventive as well as therapeutic strategies for GrTP in GI malignancy.
Collapse
Affiliation(s)
- Helieh S Oz
- Center for Oral Health Research, College of Dentistry, University of Kentucky Medical Center, Lexington, KY
| | | |
Collapse
|
44
|
Ahsan MK, Okuyama H, Hoshino Y, Oka SI, Masutani H, Yodoi J, Nakamura H. Thioredoxin-binding protein-2 deficiency enhances methionine-choline deficient diet-induced hepatic steatosis but inhibits steatohepatitis in mice. Antioxid Redox Signal 2009; 11:2573-84. [PMID: 19764881 DOI: 10.1089/ars.2009.2385] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
In nonalcoholic fatty liver disease, oxidative stress is believed to play a crucial role as a second-hit for the progression of simple steatosis to steatohepatitis. Thioredoxin (TRX) is a potent antioxidant molecule that exerts anti-apoptotic and anti-inflammatory functions. TRX-binding protein-2 (TBP-2) is an endogenous negative regulator of TRX. Deficiency of TBP-2 in mice causes hyperlipidemia, hepatic steatosis, hypoglycemia, and bleeding tendency, resembling Reye syndrome in a fasting/glucose-deficient state. The aim of this study was to investigate the role of TBP-2 in the development of nonalcoholic steatohepatitis (NASH). TBP-2-deficient (TBP-2(-/-)) and wild-type (WT) mice were fed either a normal or methionine-choline-deficient (MCD) diet for up to 10 weeks. Compared with WT mice, TBP-2(-/-) mice showed severe simple steatosis rather than steatohepatitis. However, oxidative stress determined by lipid peroxidation and DNA damage, neutrophil infiltration, and hepatic fibrosis were attenuated in TBP-2(-/-) mice. PCR analysis showed the expressions of fibrosis-inducing and inflammatory cytokine-related genes were less in TBP-2(-/-) mice. Moreover, leptin, SREBP1c, PPARgamma, and adipogenesis-lipogenesis-related genes were upregulated in TBP-2(-/-) mice. These results strongly suggested that TBP-2 might be involved in pathogenesis of NASH in WT mice, and inhibitors of TBP-2 could be useful in the prevention or treatment of NASH.
Collapse
Affiliation(s)
- Md Kaimul Ahsan
- Thioredoxin Project, Department of Experimental Therapeutics, Translational Research Center, Kyoto University Hospital, Kyoto, Japan
| | | | | | | | | | | | | |
Collapse
|
45
|
Tessari P, Coracina A, Cosma A, Tiengo A. Hepatic lipid metabolism and non-alcoholic fatty liver disease. Nutr Metab Cardiovasc Dis 2009; 19:291-302. [PMID: 19359149 DOI: 10.1016/j.numecd.2008.12.015] [Citation(s) in RCA: 228] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2008] [Revised: 12/10/2008] [Accepted: 12/29/2008] [Indexed: 12/16/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is an increasingly recognized pathology with a high prevalence and a possible evolution to its inflammatory counterpart (non-alcoholic steatohepatitis, or NASH). The pathophysiology of NAFLD and NASH has many links with the metabolic syndrome, sharing a causative factor in insulin resistance. According to a two-hit hypothesis, increased intrahepatic triglyceride accumulation (due to increased synthesis, decreased export, or both) is followed by a second step (or "hit"), which may lead to NASH. The latter likely involves oxidative stress, cytochrome P450 activation, lipid peroxidation, increased inflammatory cytokine production, activation of hepatic stellate cells and apoptosis. However, both "hits" may be caused by the same factors. The aim of this article is to overview the biochemical steps of fat regulation in the liver and the alterations occurring in the pathogenesis of NAFLD and NASH.
Collapse
Affiliation(s)
- P Tessari
- Department of Clinical and Experimental Medicine, Chair of Metabolism, University of Padua, Italy.
| | | | | | | |
Collapse
|
46
|
The transition from fatty liver to NASH associates with SAMe depletion in db/db mice fed a methionine choline-deficient diet. Dig Dis Sci 2008; 53:2761-74. [PMID: 18299981 PMCID: PMC3991247 DOI: 10.1007/s10620-007-0193-7] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2007] [Accepted: 12/21/2007] [Indexed: 12/13/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is highly prevalent in the Western population. By mechanisms that are not completely understood, this disease may progress to nonalcoholic steatohepatitis (NASH), fibrosis, cirrhosis, and hepatocellular carcinoma (HCC). db/db mice spontaneously develop hepatic steatosis, which progresses to NASH when these mice are fed a methionine choline-deficient (MCD) diet. The goal of our studies was to identify lipid and methionine metabolism pathways affected by MCD feeding to determine potential causal events leading to the development of NASH from benign steatosis. db/db mice fed the MCD diet for 2 weeks exhibited signs of incipient NASH development such as upregulated cytokines and chemokines. At this time point, MCD diet feeding caused S-adenosylmethionine (SAMe) depletion in db/db mice, while wild-type mice on the same diet retained hepatic SAMe levels. SAMe depletion exerts pleiotropic effects upon liver homeostasis and is commonly associated with a variety of liver insults such as thioacetamide, CCL4, and alcohol treatment; thus, SAMe depletion may serve as the second hit in NASH development. It is possible that differences in hepatic lipid and/or methionine metabolism between wild-type and db/db mice underlay the differential maintenance of SAMe levels during methionine and choline restriction. Indeed, db/db mice exhibited inhibited lipid oxidation pathways, which may be a priming factor for NASH development, and db/db mice fed the MCD diet had differential methionine adenosyltransferase (MAT) expression. The occurrence of SAMe depletion at this early, benign stage of NASH development in db/db mice with fatty liver suggests that SAMe supplementation may be (A) targeted to individuals susceptible to NASH (i.e., NAFLD patients) and (B) preventative of NASH before substantial liver injury has occurred.
Collapse
|
47
|
Oz HS, Ebersole JL. Application of prodrugs to inflammatory diseases of the gut. Molecules 2008; 13:452-74. [PMID: 18305431 PMCID: PMC6244946 DOI: 10.3390/molecules13020452] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2008] [Revised: 02/20/2008] [Accepted: 02/21/2008] [Indexed: 12/11/2022] Open
Abstract
Oral delivery is the most common and preferred route of drug administration although the digestive tract exhibits several obstacles to drug delivery including motility and intraluminal pH profiles. The gut milieu represents the largest mucosal surface exposed to microorganisms with 1010-12 colony forming bacteria/g of colonic content. Approximately, one third of fecal dry matter is made of bacteria/ bacterial components. Indeed, the normal gut microbiota is responsible for healthy digestion of dietary fibers (polysaccharides) and fermentation of short chain fatty acids such as acetate and butyrate that provide carbon sources (fuel) for these bacteria. Inflammatory bowel disease (IBD) results in breakage of the mucosal barrier, an altered microbiota and dysregulated gut immunity. Prodrugs that are chemically constructed to target colonic release or are degraded specifically by colonic bacteria, can be useful in the treatment of IBD. This review describes the progress in digestive tract prodrug design and delivery in light of gut metabolic activities.
Collapse
Affiliation(s)
- Helieh S Oz
- Center for Oral Health Research, College of Dentistry and Department of Internal Medicine, University of Kentucky, Medical Center, 800 Rose Street, Lexington, KY 40536, USA.
| | | |
Collapse
|