1
|
Ashaari S, Jamialahmadi T, Davies NM, Almahmeed W, Sahebkar A. Di (2-ethyl hexyl) phthalate and its metabolite-induced metabolic syndrome: a review of molecular mechanisms. Drug Chem Toxicol 2025; 48:325-343. [PMID: 39322993 DOI: 10.1080/01480545.2024.2405830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 09/06/2024] [Accepted: 09/13/2024] [Indexed: 09/27/2024]
Abstract
OBJECTIVES Metabolic disorders, as multifactorial disorders, are induced by genetic susceptibility and exposure to environmental chemicals. Di (2-ethyl hexyl) phthalate (DEHP), a ubiquitous plasticizer, is well known as an endocrine-disrupting chemical in living organisms. In recent decades, researchers have focused on the potential of DEHP and its main metabolite (Mono (2-ethylhexyl) phthalate) (MEHP) to induce metabolic disorders. In the present review, we aimed to summarize studies regarding DEHP and MEHP-induced Metabolic syndrome (MetS) as well as address the involved mechanisms. METHODS A search has been carried out in Google Scholar, PubMed, Scopus, and Web of Science databases using appropriate keywords including 'Metabolic syndrome' or 'Metabolic disorder' or 'Obesity' or 'Hyperglycemia' or 'Hyperlipidemia' or 'Hypertension' or 'Non-alcoholic fatty liver disease' and 'DEHP' or 'Di (2-ethyl hexyl) phthalate' or 'Bis(2-ethylhexyl) phthalate' or 'MEHP' or 'Mono (2-ethylhexyl) phthalate'. Studies were chosen based on inclusion and exclusion criteria. Inclusion criteria are in vitro, in vivo, epidemiological studies, and English-written studies. Exclusion criteria are lack of access to the full text of studies, editorial articles, review articles, and conference articles. RESULTS Animal studies indicate that DEHP and MEHP disrupt insulin hemostasis, increase glucose content, and induce hyperlipidemia and hypertension as well as obesity, which could lead to type 2 diabetes mellitus (T2DM) and cardiovascular disease (CVD). DEHP and its metabolite induce such effects directly through influence on nuclear receptors such as peroxisome proliferator-activated receptors (PPARs) or indirectly through reactive oxygen species (ROS) production. Both events led to the disruption of several molecular signaling pathways and subsequently metabolic syndrome (MetS). Furthermore, epidemiological studies showed that there was a correlation between DEHP metabolites levels and obesity, hyperglycemia, and hypertension. CONCLUSIONS According to studies, DEHP and its main metabolite have the potential to induce MetS by involving various molecular mechanisms. Epidemiological studies concerning the association of DEHP and MetS in humans are not sufficient. Therefore, more studies are needed in this regard.
Collapse
Affiliation(s)
- Sorour Ashaari
- Vice Chancellery for Research and Technology, Mazandaran University of Medical Sciences, Sari, Iran
| | - Tannaz Jamialahmadi
- Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Neal M Davies
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Wael Almahmeed
- Heart and Vascular Institute, Cleveland Clinic Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
2
|
Wei W, Wang LF, Tao JJ, Zhang WK, Chen SY, Song Q, Zhang JS. The comprehensive regulatory network in seed oil biosynthesis. JOURNAL OF INTEGRATIVE PLANT BIOLOGY 2025; 67:649-668. [PMID: 39821491 DOI: 10.1111/jipb.13834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 12/03/2024] [Indexed: 01/19/2025]
Abstract
Plant oils play a crucial role in human nutrition, industrial applications and biofuel production. While the enzymes involved in fatty acid (FA) biosynthesis are well-studied, the regulatory networks governing these processes remain largely unexplored. This review explores the intricate regulatory networks modulating seed oil biosynthesis, focusing on key pathways and factors. Seed oil content is determined by the efficiency of de novo FA synthesis as well as influenced by sugar transport, lipid metabolism, FA synthesis inhibitors and fine-tuning mechanisms. At the center of this regulatory network is WRINKLED1 (WRI1), which plays a conserved role in promoting seed oil content across various plant species. WRI1 interacts with multiple proteins, and its expression level is regulated by upstream regulators, including members of the LAFL network. Beyond the LAFL network, we also discuss a potential nuclear factor-Y (NF-Y) regulatory network in soybean with an emphasis on NF-YA and NF-YB and their associated proteins. This NF-Y network represents a promising avenue for future efforts aimed at enhancing oil accumulation and improving stress tolerance in soybean. Additionally, the application of omics-based approaches is of great significance. Advances in omics technologies have greatly facilitated the identification of gene resources, opening new opportunities for genetic improvement. Importantly, several transcription factors involved in oil biosynthesis also participate in stress responses, highlighting a potential link between the two processes. This comprehensive review elucidates the complex mechanisms underlying the regulation of oil biosynthesis, offering insights into potential biotechnological strategies for improving oil production and stress tolerance in oil crops.
Collapse
Affiliation(s)
- Wei Wei
- Key Laboratory of Seed Innovation, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Long-Fei Wang
- State Key Laboratory of Crop Genetics & Germplasm Enhancement and Utilization, Jiangsu Collaborative Innovation Center for Modern Crop Production, Nanjing Agricultural University, No. 1 Weigang, Nanjing, 210095, China
| | - Jian-Jun Tao
- Key Laboratory of Seed Innovation, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Wan-Ke Zhang
- Key Laboratory of Seed Innovation, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Shou-Yi Chen
- Key Laboratory of Seed Innovation, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Qingxin Song
- State Key Laboratory of Crop Genetics & Germplasm Enhancement and Utilization, Jiangsu Collaborative Innovation Center for Modern Crop Production, Nanjing Agricultural University, No. 1 Weigang, Nanjing, 210095, China
| | - Jin-Song Zhang
- Key Laboratory of Seed Innovation, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
3
|
Tezcan D, Eryavuz Onmaz D, Körez MK, Limon M, Gülcemal S, Yılmaz S, Sivrikaya A. The role of fatty acids in patients with Behçet's disease and their association with thrombosis. Lipids 2024; 59:123-133. [PMID: 38742533 DOI: 10.1002/lipd.12398] [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: 12/27/2023] [Revised: 04/19/2024] [Accepted: 05/02/2024] [Indexed: 05/16/2024]
Abstract
Behçet's disease (BD) is a systemic disease with unknown etiopathogenesis and varying disease presentations. Fatty acids (FA) are essential biological compounds that are involved in complex metabolic pathways. They may contribute to inflammation and endothelial dysfunction by participating in many signaling pathways. Increased FAs levels are associated with an increased risk for various diseases. This study aimed to determine the relationship between FA, BD, and thrombotic complications. A total of 97 patients were recruited from the rheumatology department of a single center as a case-control study. The participants were divided into three groups: 36 patients with BD with thrombosis (Group 1), 24 patients with BD without thrombosis (Group 2), and 37 age- and sex-matched controls (Group 3). The analysis of 37 different FA with carbon numbers in the range of (4:0) and (24:1) in the samples were analyzed and compared between groups. Myristic acid (MA), methyl eicosatrienoate, and stearic acid (STA) levels were found to be significantly higher in BD with thrombosis than in BD without thrombosis, and palmitic acid (PA) levels were significantly higher in BD with thrombosis than in healthy individuals. MA was found to be a significant marker for differentiating between thrombotic BD. PA and STA are important markers for detecting thrombotic BD. In BD, lipotoxicity created by FA, such as PA, STA, and MA, plays a role as an inducer of inflammation and thrombosis through various mechanisms.
Collapse
Affiliation(s)
- Dilek Tezcan
- Department of Internal Medicine, Division of Rheumatology, Gülhane Faculty of Medicine, University of Health Sciences Turkey, Ankara, Turkey
| | - Duygu Eryavuz Onmaz
- Division of Biochemistry, Selcuk University Faculty of Medicine, Konya, Turkey
| | - Muslu Kazım Körez
- Division of Biostatistics, Selcuk University Faculty of Medicine, Konya, Turkey
| | - Muhammet Limon
- Division of Rheumatology, Selcuk University Faculty of Medicine, Konya, Turkey
| | - Semral Gülcemal
- Division of Rheumatology, Selcuk University Faculty of Medicine, Konya, Turkey
| | - Sema Yılmaz
- Division of Rheumatology, Selcuk University Faculty of Medicine, Konya, Turkey
| | - Abdullah Sivrikaya
- Division of Biochemistry, Selcuk University Faculty of Medicine, Konya, Turkey
| |
Collapse
|
4
|
Huang J, Ye J, Gao Y, Wang Y, Zhao Q, Lou T, Lai W. Identification of proteins related to SIS3 by iTRAQ and PRM-based comparative proteomic analysis in cisplatin-induced acute kidney injury. PeerJ 2024; 12:e17485. [PMID: 38854800 PMCID: PMC11160430 DOI: 10.7717/peerj.17485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 05/08/2024] [Indexed: 06/11/2024] Open
Abstract
Background Cisplatin is a commonly used nephrotoxic drug and can cause acute kidney injury (AKI). In the present study, isobaric tags for relative and absolute quantification (iTRAQ) and parallel reaction monitoring (PRM)-based comparative proteomics were used to analyze differentially expressed proteins (DEPs) to determine the key molecular mechanism in mice with cisplatin-induced AKI in the presence or absence of SIS3, a specific p-smad3 inhibitor, intervention. Methods The cisplatin-induced AKI mouse model was established and treated with SIS3. We used iTRAQ to search for DEPs, PRM to verify key DEPs and combined Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) for bioinformatics analysis. We then assessed lipid deposition, malondialdehyde (MDA) and reactive oxygen species (ROS) and detected the expression of SREBF1, SCD1, CPT1A, PPARα and NDRG1 in vitro. Results Proteomic analysis showed that the identified DEPs were mainly enriched in energy metabolism pathways, especially in lipid metabolism. When SIS3 was applied to inhibit the phosphorylation of Smad3, the expression of NDRG1 and fatty acid oxidation key proteins CPT1A and PPARα increased, the expression of lipid synthesis related proteins SREBF1 and SCD1 decreased and the production of lipid droplets, MDA and ROS decreased. Conclusion SIS3 alleviates oxidative stress, reduces lipid accumulation and promotes fatty acid oxidation through NDRG1 in cisplatin-induced AKI. Our study provides a new candidate protein for elucidating the molecular mechanisms of fatty acid metabolism disorders in cisplatin-induced acute kidney injury.
Collapse
Affiliation(s)
- Jiayan Huang
- Department of Nephrology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of Nephrology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Jian Ye
- Department of Nephrology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yi Gao
- Department of Critical Care Medicine/ICU (Intensive Care Unit), The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yu Wang
- Department of Nephrology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Qing Zhao
- Department of Nephrology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Tanqi Lou
- Department of Nephrology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Weiyan Lai
- Department of Nephrology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
5
|
Tsap MI, Yatsenko AS, Hegermann J, Beckmann B, Tsikas D, Shcherbata HR. Unraveling the link between neuropathy target esterase NTE/SWS, lysosomal storage diseases, inflammation, abnormal fatty acid metabolism, and leaky brain barrier. eLife 2024; 13:e98020. [PMID: 38660940 PMCID: PMC11090517 DOI: 10.7554/elife.98020] [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: 03/21/2024] [Accepted: 04/12/2024] [Indexed: 04/26/2024] Open
Abstract
Mutations in Drosophila Swiss cheese (SWS) gene or its vertebrate orthologue neuropathy target esterase (NTE) lead to progressive neuronal degeneration in flies and humans. Despite its enzymatic function as a phospholipase is well established, the molecular mechanism responsible for maintaining nervous system integrity remains unclear. In this study, we found that NTE/SWS is present in surface glia that forms the blood-brain barrier (BBB) and that NTE/SWS is important to maintain its structure and permeability. Importantly, BBB glia-specific expression of Drosophila NTE/SWS or human NTE in the sws mutant background fully rescues surface glial organization and partially restores BBB integrity, suggesting a conserved function of NTE/SWS. Interestingly, sws mutant glia showed abnormal organization of plasma membrane domains and tight junction rafts accompanied by the accumulation of lipid droplets, lysosomes, and multilamellar bodies. Since the observed cellular phenotypes closely resemble the characteristics described in a group of metabolic disorders known as lysosomal storage diseases (LSDs), our data established a novel connection between NTE/SWS and these conditions. We found that mutants with defective BBB exhibit elevated levels of fatty acids, which are precursors of eicosanoids and are involved in the inflammatory response. Also, as a consequence of a permeable BBB, several innate immunity factors are upregulated in an age-dependent manner, while BBB glia-specific expression of NTE/SWS normalizes inflammatory response. Treatment with anti-inflammatory agents prevents the abnormal architecture of the BBB, suggesting that inflammation contributes to the maintenance of a healthy brain barrier. Considering the link between a malfunctioning BBB and various neurodegenerative diseases, gaining a deeper understanding of the molecular mechanisms causing inflammation due to a defective BBB could help to promote the use of anti-inflammatory therapies for age-related neurodegeneration.
Collapse
Affiliation(s)
- Mariana I Tsap
- Institute of Cell Biochemistry, Hannover Medical School, Hannover, Germany
| | - Andriy S Yatsenko
- Institute of Cell Biochemistry, Hannover Medical School, Hannover, Germany
| | - Jan Hegermann
- Institute of Functional and Applied Anatomy, Research Core Unit Electron Microscopy, Hannover Medical School, Hannover, Germany
| | - Bibiana Beckmann
- Institute of Toxicology, Hannover Medical School, Hannover, Germany
| | - Dimitrios Tsikas
- Institute of Toxicology, Hannover Medical School, Hannover, Germany
| | - Halyna R Shcherbata
- Institute of Cell Biochemistry, Hannover Medical School, Hannover, Germany
- Mount Desert Island Biological Laboratory, Bar Harbor, United States
| |
Collapse
|
6
|
Da Porto A, Donnini D, Vanin F, Romanin A, Antonello M, Toritto P, Varisco E, Brosolo G, Catena C, Sechi LA, Soardo G. Effects of Monacolin K in Nondiabetic Patients with NAFLD: A Pilot Study. Nutrients 2023; 15:nu15081887. [PMID: 37111106 PMCID: PMC10144054 DOI: 10.3390/nu15081887] [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: 02/24/2023] [Revised: 04/01/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a common chronic liver condition with significant risk of progression to steatohepatitis and cirrhosis. Therapeutic strategies in NAFLD include lifestyle changes mainly related to dietary interventions and use of drugs or nutritional components that could improve plasma lipid profiles and insulin sensitivity and decrease the local inflammatory response. In this study, we tested the effects of monacolin K, an inhibitor of HMCoA reductase. In a prospective, uncontrolled, open study, we treated 24 patients with NAFLD and mild hypercholesterolemia with 10 mg/day of monacolin K. At baseline and after 26 weeks, we measured in plasma liver tests, lipids, malondialdehyde, and oxidized glutathione, and assessed biochemical steatosis scores, liver elastography, and body composition with bioimpedance analysis. Monacolin K significantly reduced plasma alanine aminotransferase, cholesterol, triglycerides and the homeostatic model assessment (HOMA) index that indicated improved insulin sensitivity. No significant changes were found in body fat mass and visceral fat, nor in liver elastography, while the fatty liver index (FLI) was significantly decreased. Plasma levels of both malondialdehyde and oxidized glutathione were markedly reduced by monacolin K treatment, suggesting a reduction in oxidative stress and lipid peroxidation. In summary, this pilot study suggests possible benefits of monacolin K use in NAFLD patients that could be linked to a reduction in oxidative stress. This hypothesis should be further investigated in future studies.
Collapse
Affiliation(s)
- Andrea Da Porto
- Clinica Medica, Department of Medicine, University of Udine, 33100 Udine, Italy
- Diabetes and Metabolism Unit, Department of Medicine, University of Udine, 33100 Udine, Italy
| | - Debora Donnini
- Clinica Medica, Department of Medicine, University of Udine, 33100 Udine, Italy
- Liver Unit, Department of Medicine, University of Udine, 33100 Udine, Italy
| | - Fabio Vanin
- Clinica Medica, Department of Medicine, University of Udine, 33100 Udine, Italy
- Liver Unit, Department of Medicine, University of Udine, 33100 Udine, Italy
| | - Arianna Romanin
- Clinica Medica, Department of Medicine, University of Udine, 33100 Udine, Italy
- Liver Unit, Department of Medicine, University of Udine, 33100 Udine, Italy
| | - Martina Antonello
- Clinica Medica, Department of Medicine, University of Udine, 33100 Udine, Italy
- Diabetes and Metabolism Unit, Department of Medicine, University of Udine, 33100 Udine, Italy
| | - Paolo Toritto
- Clinica Medica, Department of Medicine, University of Udine, 33100 Udine, Italy
- Liver Unit, Department of Medicine, University of Udine, 33100 Udine, Italy
| | - Eleonora Varisco
- Clinica Medica, Department of Medicine, University of Udine, 33100 Udine, Italy
- Liver Unit, Department of Medicine, University of Udine, 33100 Udine, Italy
| | - Gabriele Brosolo
- Clinica Medica, Department of Medicine, University of Udine, 33100 Udine, Italy
| | - Cristiana Catena
- Clinica Medica, Department of Medicine, University of Udine, 33100 Udine, Italy
| | - Leonardo A Sechi
- Clinica Medica, Department of Medicine, University of Udine, 33100 Udine, Italy
| | - Giorgio Soardo
- Clinica Medica, Department of Medicine, University of Udine, 33100 Udine, Italy
- Liver Unit, Department of Medicine, University of Udine, 33100 Udine, Italy
- Italian Liver Foundation, Area Science Park, Basovizza, 34149 Trieste, Italy
| |
Collapse
|
7
|
Bale BF, Doneen AL, Leimgruber PP, Vigerust DJ. The critical issue linking lipids and inflammation: Clinical utility of stopping oxidative stress. Front Cardiovasc Med 2022; 9:1042729. [PMID: 36439997 PMCID: PMC9682196 DOI: 10.3389/fcvm.2022.1042729] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 10/24/2022] [Indexed: 07/30/2023] Open
Abstract
The formation of an atheroma begins when lipoproteins become trapped in the intima. Entrapped lipoproteins become oxidized and activate the innate immune system. This immunity represents the primary association between lipids and inflammation. When the trapping continues, the link between lipids and inflammation becomes chronic and detrimental, resulting in atherosclerosis. When entrapment ceases, the association between lipids and inflammation is temporary and healthy, and the atherogenic process halts. Therefore, the link between lipids and inflammation depends upon lipoprotein retention in the intima. The entrapment is due to electrostatic forces uniting apolipoprotein B to polysaccharide chains on intimal proteoglycans. The genetic transformation of contractile smooth muscle cells in the media into migratory secretory smooth muscle cells produces the intimal proteoglycans. The protein, platelet-derived growth factor produced by activated platelets, is the primary stimulus for this genetic change. Oxidative stress is the main stimulus to activate platelets. Therefore, minimizing oxidative stress would significantly reduce the retention of lipoproteins. Less entrapment decreases the association between lipids and inflammation. More importantly, it would halt atherogenesis. This review will analyze oxidative stress as the critical link between lipids, inflammation, and the pathogenesis of atherosclerosis. Through this perspective, we will discuss stopping oxidative stress to disrupt a harmful association between lipids and inflammation. Numerous therapeutic options will be discussed to mitigate oxidative stress. This paper will add a new meaning to the Morse code distress signal SOS-stopping oxidative stress.
Collapse
Affiliation(s)
- Bradley Field Bale
- Department of Medical Education and Clinical Sciences, Washington State University College of Medicine, Spokane, WA, United States
| | - Amy Lynn Doneen
- Department of Medical Education and Clinical Sciences, Washington State University College of Medicine, Spokane, WA, United States
| | - Pierre P. Leimgruber
- Department of Medical Education and Clinical Sciences, Washington State University College of Medicine, Spokane, WA, United States
- Department of Medical Education and Clinical Sciences, University of Washington School of Medicine, Seattle, WA, United States
| | - David John Vigerust
- Department of Neurological Surgery, Vanderbilt University School of Medicine, Nashville, TN, United States
| |
Collapse
|
8
|
Catena C, Brosolo G, Da Porto A, Donnini D, Bulfone L, Vacca A, Soardo G, Sechi LA. Association of non-alcoholic fatty liver disease with left ventricular changes in treatment-naive patients with uncomplicated hypertension. Front Cardiovasc Med 2022; 9:1030968. [PMID: 36312275 PMCID: PMC9606246 DOI: 10.3389/fcvm.2022.1030968] [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: 08/29/2022] [Accepted: 09/27/2022] [Indexed: 11/13/2022] Open
Abstract
Background and aims Cardiac structural and functional changes have been demonstrated in patients with non-alcoholic fatty liver disease (NAFLD). Because of the frequent association of NAFLD with hypertension, we aimed to examine the relationship of liver steatosis with left ventricular (LV) changes in patients with hypertension. Materials and methods In a cross-sectional study, we included 360 untreated, essential hypertensive patients who were free of major cardiovascular and renal complications. Liver steatosis was assessed by three different biochemical scores (NAFLD Liver Fat Score, LFS; Fatty Liver Index, FLI; Hepatic Steatosis Index, HSI). Echocardiography was performed with standard B-mode and tissue-Doppler imaging. Results LV hypertrophy was present in 19.4% and LV diastolic dysfunction in 49.2% of patients who had significantly higher body mass index (BMI), blood pressure (BP), and homeostatic model assessment (HOMA) index and higher frequency of the metabolic syndrome and liver steatosis that was defined by presence of 2 or more positive scores. LV mass index increased progressively across patients who had none, 1, or 2 or more liver steatosis scores, with associated progressive worsening of LV diastolic function. LV mass index was significantly and positively correlated with age, BMI, BP, HOMA-index, LFS, and HSI. Logistic regression analysis showed that age, BP, and liver steatosis scores independently predicted LV hypertrophy and diastolic dysfunction. Liver steatosis independently predicted LV dysfunction but not LV hypertrophy even after inclusion in analysis of the HOMA-index. Conclusion NAFLD is associated with LV hypertrophy and diastolic dysfunction in untreated patients with hypertension. In hypertension, NAFLD could contribute to LV diastolic dysfunction with mechanisms unrelated to insulin resistance.
Collapse
Affiliation(s)
- Cristiana Catena
- Internal Medicine and European Hypertension Excellence Center, Department of Medicine, University of Udine, Udine, Italy
| | - Gabriele Brosolo
- Internal Medicine and European Hypertension Excellence Center, Department of Medicine, University of Udine, Udine, Italy
| | - Andrea Da Porto
- Diabetes and Metabolism Unit, Department of Medicine, University of Udine, Udine, Italy
| | - Debora Donnini
- Liver Unit, Department of Medicine, University of Udine, Udine, Italy
| | - Luca Bulfone
- Internal Medicine and European Hypertension Excellence Center, Department of Medicine, University of Udine, Udine, Italy
| | - Antonio Vacca
- Internal Medicine and European Hypertension Excellence Center, Department of Medicine, University of Udine, Udine, Italy
| | - Giorgio Soardo
- Liver Unit, Department of Medicine, University of Udine, Udine, Italy
| | - Leonardo A. Sechi
- Internal Medicine and European Hypertension Excellence Center, Department of Medicine, University of Udine, Udine, Italy,Diabetes and Metabolism Unit, Department of Medicine, University of Udine, Udine, Italy,Liver Unit, Department of Medicine, University of Udine, Udine, Italy,*Correspondence: Leonardo A. Sechi,
| |
Collapse
|
9
|
Association of Body Mass Index with Hearing Loss in Korean Adult Population. J Pers Med 2022; 12:jpm12050786. [PMID: 35629208 PMCID: PMC9147342 DOI: 10.3390/jpm12050786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/12/2022] [Accepted: 05/12/2022] [Indexed: 11/21/2022] Open
Abstract
This study aimed to explore the relationship between body mass index (BMI) and hearing loss. We analyzed data from the Korean National Health Insurance Service Health Screening Cohort 2009−2019 (291,471 patients with hearing loss and 6,088,979 control participants). Both patient groups were subsequently divided into four groups according to BMI: <18.5 (underweight), 18.5−24.9 (normal), 25−29.9 (obese I), and ≥30 (obese II). To evaluate the relationship between BMI and hearing loss, multivariate logistic regression analysis was used, adjusting for age, sex, smoking, alcohol consumption, blood pressure, triglycerides, total cholesterol, low-density lipoprotein, proteinuria, serum creatinine, aspartate aminotransferase, alanine aminotransferase, and fasting glucose levels. The adjusted odds ratio (OR) of the underweight group for hearing loss was 1.21 (95% CI = 1.19−1.24) compared to the normal BMI group, whereas the adjusted ORs of obese I and obese II groups for hearing loss were 0.95 and 0.87, respectively. Being underweight was generally associated with an increased prevalence of hearing loss in the Korean adult population.
Collapse
|
10
|
Yu W, Ma Y, Shrivastava SK, Srivastava RK, Shankar S. Chronic alcohol exposure induces hepatocyte damage by inducing oxidative stress, SATB2 and stem cell‐like characteristics, and activating lipogenesis. J Cell Mol Med 2022; 26:2119-2131. [PMID: 35152538 PMCID: PMC8980954 DOI: 10.1111/jcmm.17235] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/25/2022] [Accepted: 01/31/2022] [Indexed: 12/12/2022] Open
Abstract
Alcohol is a risk factor for hepatocellular carcinoma (HCC). However, the molecular mechanism by which chronic alcohol consumption contributes to HCC is not well understood. The purpose of the study was to demonstrate the effects of chronic ethanol exposure on the damage of human normal hepatocytes. Our data showed that chronic exposure of hepatocytes with ethanol induced changes similar to transformed hepatocytes that is, exhibited colonies and anchorage‐independent growth. These damaged hepatocytes contained high levels of reactive oxygen species (ROS) and showed induction of the SATB2 gene. Furthermore, damaged hepatocytes gained the phenotypes of CSCs which expressed stem cell markers (CD133, CD44, CD90, EpCAM, AFP and LGR5), and pluripotency maintaining factors (Sox‐2, POU5F1/Oct4 and KLF‐4). Ethanol exposure also induced Nanog, a pluripotency maintaining transcription factor that functions in concert with Oct4 and SOX‐2. Furthermore, ethanol induced expression of EMT‐related transcription factors (Snail, Slug and Zeb1), N‐Cadherin, and inhibited E‐cadherin expression in damaged hepatocytes. Ethanol enhanced recruitment of SATB2 to promoters of Bcl‐2, Nanog, c‐Myc, Klf4 and Oct4. Ethanol also induced activation of the Wnt/TCF‐LEF1 pathway and its targets (Bcl‐2, Cyclin D1, AXIN2 and Myc). Finally, ethanol induced hepatocellular steatosis, SREBP1 transcription, and modulated the expression of SREBP1c, ACAC, ACLY, FASN, IL‐1β, IL‐6, TNF‐α, GPC3, FLNB and p53. These data suggest that chronic alcohol consumption may contribute towards the development of HCC by damaging normal hepatocytes with the generation of inflammatory environment, induction of SATB2, stem cell‐like characteristics, and cellular steatosis.
Collapse
Affiliation(s)
- Wei Yu
- Kansas City VA Medical Center Kansas City Missouri USA
| | - Yiming Ma
- Kansas City VA Medical Center Kansas City Missouri USA
| | - Sushant K. Shrivastava
- Department of Pharmaceutics Indian Institute of Technology Banaras Hindu University Varanasi U.P. India
| | - Rakesh K. Srivastava
- Kansas City VA Medical Center Kansas City Missouri USA
- Department of Genetics Louisiana State University Health Sciences Center New Orleans Louisina USA
- Stanley S. Scott Cancer Center Department of Genetics Louisiana State University Health Sciences Center New Orleans Louisina USA
- A.B. Freeman School of Business Tulane University New Orleans Louisina USA
| | - Sharmila Shankar
- Kansas City VA Medical Center Kansas City Missouri USA
- John W. Deming Department of Medicine Tulane University School of Medicine New Orleans Louisina USA
- Southeast Louisiana Veterans Health Care System New Orleans Louisina USA
| |
Collapse
|
11
|
Deng Z, Gao S, An Y, Huang Y, Liu H, Zhu W, Lu W, He M, Xie W, Yu D, Li Y. Effects of earthworm extract on the lipid profile and fatty liver induced by a high-fat diet in guinea pigs. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:292. [PMID: 33708919 PMCID: PMC7944315 DOI: 10.21037/atm-20-5362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background Non-alcoholic fatty liver disease (NAFLD), characterized by the accumulation of excess fat in the liver in people who consume little or no alcohol, is becoming increasingly common around the world, especially in developed countries. Extracts from earthworms have been used as alternative therapies for a variety of diseases but not in NAFLD. Therefore, the aim of this study was to investigate the effect of earthworm extract (EE) on diet-induced fatty liver disease in guinea pigs. Methods EE was extracted, and the effect of EE on the lipid levels and liver damage in guinea pigs fed a high-fat diet (HFD) was assessed. Thirty male guinea pigs at 3 weeks of age were allocated equally to five groups, namely, chow diet, HFD, and HFD with different dosages (0.3, 1.4 and 6.8 µg per kg bodyweight per day) of EE for 4 weeks, and their body weight was monitored throughout the experiment. Liver tissues were examined for gross morphology and histology. Serum levels of total cholesterol (TC), triglycerides (TG), low-density lipoprotein cholesterol (LDL-C), alanine transaminase (ALT) and aspartate aminotransferase (AST) were determined using an autoanalyser. Results HFD induced NAFLD in guinea pigs. HFD-fed guinea pigs that received EE treatment showed milder increases in the serum levels of TC, TG and LDL-C, as well as in the body weight growth rate, compared to the HFD group without EE supplementation. EE intervention reduced the number of lipid-containing hepatocytes, hepatocellular ballooning and sinusoidal distortion in the liver in HFD-fed animals. ALT in serum was significantly elevated by HFD. No statistically significant difference in ALT levels was found between the chow diet group and the HFD group with EE treatment. Conclusions This study demonstrates that the administration of EE suppressed the induction of serum TC, TG and LDL-C in response to HFD. EE also reduced liver damage in HFD-fed guinea pigs. These findings suggest that EE has alleviating effects on dyslipidaemia and liver damage associated with NAFLD.
Collapse
Affiliation(s)
- Zhenhan Deng
- Department of Sports Medicine, Key Laboratory of Tissue Engineering of Shenzhen, Shenzhen Second People's Hospital/the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China.,Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China.,School of Medicine, Shenzhen University, Shenzhen, China
| | - Shanshan Gao
- Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Yunfei An
- Department of Animal Science, College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Yong Huang
- Department of Sports Medicine, Key Laboratory of Tissue Engineering of Shenzhen, Shenzhen Second People's Hospital/the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Haifeng Liu
- Department of Sports Medicine, Key Laboratory of Tissue Engineering of Shenzhen, Shenzhen Second People's Hospital/the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Weimin Zhu
- Department of Sports Medicine, Key Laboratory of Tissue Engineering of Shenzhen, Shenzhen Second People's Hospital/the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Wei Lu
- Department of Sports Medicine, Key Laboratory of Tissue Engineering of Shenzhen, Shenzhen Second People's Hospital/the First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Miao He
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
| | - Wenqing Xie
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
| | - Dengjie Yu
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
| | - Yusheng Li
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
12
|
Yang M, Chen Z, Xiang S, Xia F, Tang W, Yao X, Zhou B. Hugan Qingzhi medication ameliorates free fatty acid-induced L02 hepatocyte endoplasmic reticulum stress by regulating the activation of PKC-δ. BMC Complement Med Ther 2020; 20:377. [PMID: 33308192 PMCID: PMC7730760 DOI: 10.1186/s12906-020-03164-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 11/23/2020] [Indexed: 12/16/2022] Open
Abstract
Background Previous studies have found that Hugan Qingzhi tablet (HQT) has significant lipid-lowering and antioxidant effects on non-alcoholic fatty liver disease (NAFLD). Moreover, the results of proteomic analysis confirmed that various proteins in endoplasmic reticulum stress (ERS) pathway were activated and recovered by HQT. However, its mechanism remains confused. The purpose of this study was to explore the effects of HQT-medicated serum on hepatic ERS and its relevant mechanisms. Methods L02 cells were induced by Free Fatty Acid (FFA) for 24 h to establish a model of hepatic ERS and pretreated with the drug-medicated rat serum for 24 h. Accumulation of intracellular lipid was evaluated using Oil Red O staining and Triglyceride detection kit. The morphological changes of ER were observed by TEM. PKC-δ was silenced by specific siRNA. Western blot and RT-qPCR were applied to detect the expression of markers related to ERS, calcium disorder, steatosis and insulin resistance. The fluorescence of Ca2+ influx was recorded using fluorescence spectrophotometer. Results HQT-medicated serum significantly decreased the intracellular TG content. Furthermore, it caused significant reduction in the expression of ERS markers and an improvement in ER structure of L02 cells. PKC-δ was activated into phosphorylated PKC-δ in FFA-induced L02 hepatocytes while these changes can be reversed by HQT-medicated serum. Silencing PKC-δ in L02 cells can restore the expression and activity of SERCA2 in ER and down-regulate the expression of IP3R protein to maintain intracellular calcium homeostasis, so as to relieve FFA-induced ERS and its lipid accumulation and insulin resistance. Conclusions The results concluded that HQT-medicated serum exerts protective effects against hepatic ERS, steatosis and insulin resistance in FFA-induced L02 hepatocyte. And its potential mechanism might be down-regulating the activation of PKC-δ and stabilization of intracellular calcium. Supplementary Information The online version contains supplementary material available at 10.1186/s12906-020-03164-3.
Collapse
Affiliation(s)
- Miaoting Yang
- Department of Pharmacy, People's Hospital of Longhua, Shenzhen, 518109, Guangdong, China
| | - Zhijuan Chen
- Department of Pharmacy, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, Guangdong, China
| | - Shijian Xiang
- Department of Pharmacy, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, Guangdong, China
| | - Fan Xia
- Department of Pharmacy, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, Guangdong, China
| | - Waijiao Tang
- Department of Pharmacy, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China
| | - Xiaorui Yao
- Department of Pharmacy, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-sen University, Shantou, 515041, Guangdong, China
| | - Benjie Zhou
- Department of Pharmacy, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, Guangdong, China.
| |
Collapse
|
13
|
tBHQ Induces a Hormetic Response That Protects L6 Myoblasts against the Toxic Effect of Palmitate. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:3123268. [PMID: 32509140 PMCID: PMC7246405 DOI: 10.1155/2020/3123268] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 04/20/2020] [Indexed: 12/27/2022]
Abstract
Nutritional status, in particular overweight and obesity, as well as sedentarism and high-fat diet consumption, are important risk factors to develop chronic diseases, which have a higher impact on the elderly's health. Therefore, these nutritional problems have become a concern to human healthspan and longevity. The fatty acids obtained thru the diet or due to fatty acid synthesis during obesity accumulate within the body generating toxicity and cell death. Fat is not only stored in adipose tissue, but it can also be stored in skeletal muscle. Palmitic acid (PA) has been reported as one of the most important saturated free fatty acids; it is associated to chronic oxidative stress and increased mitochondrial ROS production causing cell death by apoptosis. In skeletal muscle, palmitate has been associated with various pathophysiological consequences, which lead to muscle deterioration during aging and obesity. Since molecules that modify redox state have been proven to prevent cellular damage by inducing a hormetic response, the aim of this study was to evaluate if tert-butylhydroquinone (tBHQ) could activate an antioxidant hormetic response that would be able to protect L6 myoblasts from palmitate toxic effect. Our results provide evidence that tBHQ is able to protect L6 myoblasts against the toxicity induced by sodium palmitate due to a synergistic activation of different signaling pathways such as Nrf2 and NF-κB.
Collapse
|
14
|
Abualhasan MN, Watson DG. Tagging Fatty Acids Via Choline Coupling for the Detection of Carboxylic Acid Metabolites in Biological Samples. CURR ANAL CHEM 2019. [DOI: 10.2174/1573411014666180516093353] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
Fatty acids and other metabolites containing a carboxyl group are of high interest
in biomedicine because of their major role in many metabolic pathways and, particularly in the
case of oxidised fatty acids, their high biological activity. Tagging carboxylic acid compounds with a
permanent positive charge such as a quaternary ammonium compound could increase the LC-MS detection
sensitivity and selectivity. This paper describes a new and novel strategy for analysing carboxylcontaining
compounds in biological samples by ESI-MS through coupling to choline.
Methods:
Coupling of carboxylic acid derivatives in biological samples was performed by coupling to
2-Fluoro-1, 3 dimethyl –pyridinium (FDMP). The variation in the fatty acid profile of five different
plasma samples was studied and was illustrated by using principal components analysis (PCA) to group
the samples. Orthogonal partial least squares discriminant analysis (OPLS-DA) modelling was then
applied to identify the fatty acids that were responsible for the variation.
Results:
The test results showed that choline coupling reactions were successful in detecting fatty acids,
oxidised fatty acids and other compounds containing carboxylic acid groups in biological samples. The
PCA results showed loadings of different fatty acids according to the plasma sample allowing identification
of the fatty acids responsible for the observed variation.
Conclusion:
A new and easy tagging method was developed to detect carboxylic acids in plasma samples.
The method proved to be precise and reproducible and can quantify fatty acid compounds to 50
ng/ml.
Collapse
Affiliation(s)
- Murad N. Abualhasan
- Faculty of Medicine and Health Sciences, Department of Pharmacy, An-Najah National University, Nablus, Palestinian Territory, Occupied
| | - David G. Watson
- University of Strathclyde, Strathclyde Institute of Pharmacy and Biomedical Sciences (SIPBS), Glasgow, United Kingdom
| |
Collapse
|
15
|
Chaliha M, Sultanbawa Y. Terminalia ferdinandiana, a traditional medicinal plant of Australia, alleviates hydrogen peroxide induced oxidative stress and inflammation, in vitro. ACTA ACUST UNITED AC 2019; 17:/j/jcim.ahead-of-print/jcim-2019-0008/jcim-2019-0008.xml. [DOI: 10.1515/jcim-2019-0008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 04/11/2019] [Indexed: 12/31/2022]
Abstract
AbstractBackgroundOxidative stress and inflammation are the underlying factors in many chronic debilitating diseases and commonly intertwined. Terminalia ferdinandiana is a traditional medicinal plant, endemic to Australia and is a rich source of many bioactive phytochemicals such as ellagic acid (EA) with known antioxidant capacity.MethodsWe investigated the in vitro antioxidant and anti-inflammatory activity of an aqueous food grade EA enriched (EAE) extract of T. ferdinandiana. Caco-2 and KERTr cell lines were treated with EAE or pure EA (used as reference control), followed by the exposure to hydrogen peroxide (H2O2). Levels of reactive oxygen species (ROS) production and gene expression of molecular markers associated with oxidative stress and inflammation were monitored.ResultsSignificant reduction in ROS production was observed in both cell types treated with 100 or 200 µg/mL EA or EAE. Treatment of cells with EAE or EA showed upregulation of mRNA expression of the antioxidative gene superoxide dismutase (SOD)-2 and downregulated the expression of inducible nitric oxide synthase (iNOS), soluble cell adhesion molecule (sICAM), and cyclooxygenase (COX)-2. Neither EAE nor EA had any effect on the constitutively expressed COX1.ConclusionsThe antioxidant and anti-inflammatory activity of T. ferdinandiana extract on mammalian cells exposed to H2O2 suggests the potential of using this traditional medicinal plant in preventing oxidative damage and inflammation related diseases.
Collapse
Affiliation(s)
- Mridusmita Chaliha
- Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, Block 10, Level 1, 39 Kessels Rd, Coopers Plains Qld 4108, Queensland, Australia
| | - Yasmina Sultanbawa
- Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, Block 10, Level 1, 39 Kessels Rd, Coopers Plains Qld 4108, Queensland, Australia
| |
Collapse
|
16
|
Kaphalia L, Srinivasan MP, Kakumanu RD, Kaphalia BS, Calhoun WJ. Ethanol Exposure Impairs AMPK Signaling and Phagocytosis in Human Alveolar Macrophages: Role of Ethanol Metabolism. Alcohol Clin Exp Res 2019; 43:1682-1694. [PMID: 31211863 DOI: 10.1111/acer.14131] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 06/10/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Chronic alcohol consumption impairs alveolar macrophage's (AM) function and increases risk for developing lung infection and pneumonia. However, the mechanism and metabolic basis of alcohol-induced AM dysfunction leading to lung infection are not well defined, but may include altered ethanol (EtOH) and reactive oxygen species metabolism and cellular energetics. Therefore, oxidative stress, endoplasmic reticulum (ER) stress, the formation of fatty acid ethyl esters [FAEEs, nonoxidative metabolites of EtOH], AMP-activated protein kinase (AMPK) signaling, and phagocytic function were examined in freshly isolated AM incubated with EtOH. METHODS AMs separated from bronchoalveolar lavage fluid samples obtained from normal volunteers were incubated with EtOH for 24 hours. AMPK signaling and ER stress were assessed using Western blotting, FAEEs by GC-MS, oxidative stress by immunofluorescence using antibodies to 4-hydroxynonenal, and phagocytosis by latex beads. Oxidative stress was also measured in EtOH-treated AMs with/without AMPK activator [5-aminoimidazole-4-carboxamide ribonucleotide (AICAR)] or inhibitor (Compound C), and in AMs incubated with FAEEs. mRNA expression for interleukins (IL-6 and IL-8), monocyte chemoattractant protein (MCP)-1, and transforming growth factor (TGF)-β was measured in AM treated with EtOH or FAEEs using RT-PCR. RESULTS EtOH exposure to AM increased oxidative stress, ER stress, and synthesis of FAEEs, decreased phosphorylated AMPK, and impaired phagocytosis. Attenuation or exacerbation of EtOH-induced oxidative stress by AICAR or Compound C, respectively, suggests a link between AMPK signaling, EtOH metabolism, and related oxidative stress. The formation of FAEEs may contribute to EtOH-induced oxidative stress as FAEEs also produced concentration-dependent oxidative stress. An increased mRNA expression of IL-6, IL-8, and MCP-1 by FAEEs is key finding to suggest a metabolic basis of EtOH-induced inflammatory response. CONCLUSIONS EtOH-induced impaired phagocytosis, oxidative stress, ER stress, and dysregulated AMPK signaling are plausibly associated with the formation of FAEEs and may participate in the pathogenesis of nonspecific pulmonary inflammation.
Collapse
Affiliation(s)
- Lata Kaphalia
- Division of Pulmonary, Critical Care Medicine, and Sleep, Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas.,Division of Allergy & Clinical Immunology, Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas
| | - Mukund P Srinivasan
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas
| | - Ramu D Kakumanu
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas
| | | | - William J Calhoun
- Division of Pulmonary, Critical Care Medicine, and Sleep, Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas.,Division of Allergy & Clinical Immunology, Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas
| |
Collapse
|
17
|
Rai V, Bose S, Saha S, Chakraborty C. Evaluation of oxidative stress and the microenvironment in oral submucous fibrosis. Heliyon 2019; 5:e01502. [PMID: 31011652 PMCID: PMC6462775 DOI: 10.1016/j.heliyon.2019.e01502] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 03/24/2019] [Accepted: 04/08/2019] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Oral Submucous fibrosis (OSF) is a chronic inflammatory mucosal disease of unknown etiology. Statistics show cases of OSF which has a high rate of overall prevalence and increase the chance of malignant transformation. As we know malignant cells is situated in a very complex microenvironment with altered metabolic pathway including intermediates which participate in oxidative stress process which enhances metabolic rewiring and promotes tumor progression. This study aims to evaluate the tumor microenvironment and their role in metabolic reprogramming. METHODS This study was conducted on the serum sample of OSF (n = 20) compared to the healthy group (n = 20) using ELISA. The serum levels of intermediate by-products of metabolic pathway and oxidative stress induced biomolecular damage products were determined. The sensitivity of results was analyzed by correlating it with markers of metabolic status (Glucose, Total cholesterol, Total protein). RESULTS Metabolic pathway intermediates molecules like Fatty Acids (FAA), Ascorbic acid, Citrate, Oxaloacetate (OAA), levels were significantly high in the serum of OSF cases. This indicated that intermediates act as a metabolic switch that drives cells to adapt malignant transformation pathway. Markers related to oxidative DNA damage (8-hydroxy-2' -deoxyguanosine), Oxidative lipid peroxidation (8-epi-Prostaglandin F2α), and Protein carbonyl were significantly up-regulated. This significant increase in oxidative stress marker revealed the reprogramming of the metabolic pathway for fulfilling the nutritional requirement of cancer cells. A further significant correlation was observed with metabolic products confirmed altered metabolic status. CONCLUSION Our findings could identify the differentiating intermediate pathway metabolites and oxidative damage to biomolecules that are leading to rewiring of metabolism in the OSF group. Findings described in the study can be helpful to explain further the molecular aspects that lead to the progression of OSF towards carcinogenesis.
Collapse
Affiliation(s)
- Vertika Rai
- School of Medical Science and Technology, IIT Kharagpur, India
| | - Surajit Bose
- Awadh Dental College and Hospital, Jamshedpur, India
| | - Satadal Saha
- School of Medical Science and Technology, IIT Kharagpur, India
| | | |
Collapse
|
18
|
Umeda R, Takanari H, Ogata K, Matsumoto S, Kitano T, Ono K, Tokumaru O. Direct free radical scavenging effects of water-soluble HMG-CoA reductase inhibitors. J Clin Biochem Nutr 2019; 64:20-26. [PMID: 30705508 PMCID: PMC6348410 DOI: 10.3164/jcbn.18-48] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Accepted: 05/11/2018] [Indexed: 12/18/2022] Open
Abstract
3-Hydroxy-3-methylglutaryl coenzyme A reductase inhibitors, statins, are widely used for preventing cardiovascular and cerebrovascular diseases by controlling blood cholesterol level. Additionally, previous studies revealed the scavenging effects of statins on free radicals. We assessed direct scavenging activities of two water-soluble statins, fluvastatin and pravastatin, on multiple free radicals using electron spin resonance spectrometry with spin trapping method. We estimated reaction rate constants (kfv for fluvastatin, and kpv for pravastatin). Superoxide anion was scavenged by fluvastatin and pravastatin with kfv and kpv of 4.82 M−1s−1 and 49.0 M−1s−1, respectively. Scavenging effects of fluvastatin and pravastatin on hydroxyl radical were comparable; both kfv and kpv were >109 M−1s−1. Fluvastatin also eliminated tert-butyl peroxyl radical with relative kfv of 2.63 to that of CYPMPO, whereas pravastatin did not affect tert-butyl peroxyl radical. Nitric oxide was scavenged by fluvastatin and pravastatin with kfv and kpv of 68.6 M−1s−1 and 701 M−1s−1, respectively. Both fluvastatin and pravastatin had scavenging effects on superoxide anion, hydroxyl radical and nitric oxide radical. On the other hand, tert-butyl peroxyl radical was scavenged only by fluvastatin, suggesting that fluvastatin might have more potential effect than pravastatin to prevent atherosclerosis and ischemia/reperfusion injury via inhibiting oxidation of lipids.
Collapse
Affiliation(s)
- Ryohei Umeda
- Department of Pathophysiology, Oita University Faculty of Medicine, 1-1 Idaigaoka, Hasama-machi, Yufu, Oita 879-5593, Japan
| | - Hiroki Takanari
- Department of Pathophysiology, Oita University Faculty of Medicine, 1-1 Idaigaoka, Hasama-machi, Yufu, Oita 879-5593, Japan.,Clinical Research Center for Diabetes, Tokushima University Hospital, 2-50-1 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Kazue Ogata
- Department of Anesthesiology, Oita University Faculty of Medicine, 1-1 Idaigaoka, Hasama-machi, Yufu, Oita 879-5593, Japan
| | - Shigekiyo Matsumoto
- Department of Anesthesiology, Oita University Faculty of Medicine, 1-1 Idaigaoka, Hasama-machi, Yufu, Oita 879-5593, Japan
| | - Takaaki Kitano
- Department of Anesthesiology, Oita University Faculty of Medicine, 1-1 Idaigaoka, Hasama-machi, Yufu, Oita 879-5593, Japan
| | - Katsushige Ono
- Department of Pathophysiology, Oita University Faculty of Medicine, 1-1 Idaigaoka, Hasama-machi, Yufu, Oita 879-5593, Japan
| | - Osamu Tokumaru
- Department of Physiology, Faculty of Welfare and Health Sciences, Oita University, 700 Dan-noharu, Oita 870-1192, Japan
| |
Collapse
|
19
|
Di Cara F, Bülow MH, Simmonds AJ, Rachubinski RA. Dysfunctional peroxisomes compromise gut structure and host defense by increased cell death and Tor-dependent autophagy. Mol Biol Cell 2018; 29:2766-2783. [PMID: 30188767 PMCID: PMC6249834 DOI: 10.1091/mbc.e18-07-0434] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The gut has a central role in digestion and nutrient absorption, but it also serves in defending against pathogens, engages in mutually beneficial interactions with commensals, and is a major source of endocrine signals. Gut homeostasis is necessary for organismal health and changes to the gut are associated with conditions like obesity and diabetes and inflammatory illnesses like Crohn's disease. We report that peroxisomes, organelles involved in lipid metabolism and redox balance, are required to maintain gut epithelium homeostasis and renewal in Drosophila and for survival and development of the organism. Dysfunctional peroxisomes in gut epithelial cells activate Tor kinase-dependent autophagy that increases cell death and epithelial instability, which ultimately alter the composition of the intestinal microbiota, compromise immune pathways in the gut in response to infection, and affect organismal survival. Peroxisomes in the gut effectively function as hubs that coordinate responses from stress, metabolic, and immune signaling pathways to maintain enteric health and the functionality of the gut-microbe interface.
Collapse
Affiliation(s)
- Francesca Di Cara
- Department of Cell Biology, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Margret H Bülow
- Development, Genetics and Molecular Physiology, LIMES (Life and Medical Sciences), University of Bonn, D-53115 Bonn, Germany
| | - Andrew J Simmonds
- Department of Cell Biology, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | | |
Collapse
|
20
|
Kosmas CE, Silverio D, Tsomidou C, Salcedo MD, Montan PD, Guzman E. The Impact of Insulin Resistance and Chronic Kidney Disease on Inflammation and Cardiovascular Disease. CLINICAL MEDICINE INSIGHTS-ENDOCRINOLOGY AND DIABETES 2018; 11:1179551418792257. [PMID: 30083062 PMCID: PMC6071166 DOI: 10.1177/1179551418792257] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 07/07/2018] [Indexed: 12/13/2022]
Abstract
There is extensive evidence showing that insulin resistance (IR) is associated
with chronic low-grade inflammation. Furthermore, IR has been shown to increase
the risk for cardiovascular disease (CVD), even in nondiabetic patients, and is
currently considered as a “nontraditional” risk factor contributing to CVD by
promoting hypertension, oxidative stress, endothelial dysfunction, dyslipidemia,
and type 2 diabetes mellitus. However, chronic kidney disease (CKD) is also
considered a state of low-grade inflammation. In addition, CKD is considered an
IR state and has been described as an independent risk factor for the
development of CVD, as even early-stage CKD is associated with an estimated 40%
to 100% increase in CVD risk. There is also strong evidence indicating that
inflammation per se plays a crucial role in both the initiation and progression
of CVD. Given the above, the combined effect of IR and CKD may significantly
increase the risk of inflammation and CVD. This review aims to focus on the
complex interplay between IR, CKD, inflammation, and CVD and will present and
discuss the current clinical and scientific data pertaining to the impact of IR
and CKD on inflammation and CVD.
Collapse
Affiliation(s)
- Constantine E Kosmas
- Division of Cardiology, Department of Medicine, Mount Sinai Hospital, New York, NY, USA
| | - Delia Silverio
- Cardiology Clinic, Cardiology Unlimited PC, New York, NY, USA
| | - Christiana Tsomidou
- Department of Medicine, General Clinic of Piraeus "Hippocrates", Piraeus, Greece
| | - Maria D Salcedo
- Cardiology Clinic, Cardiology Unlimited PC, New York, NY, USA
| | - Peter D Montan
- Cardiology Clinic, Cardiology Unlimited PC, New York, NY, USA
| | - Eliscer Guzman
- Division of Cardiology, Department of Medicine, Montefiore Medical Center, Bronx, NY, USA
| |
Collapse
|
21
|
Tsang CK, Chen M, Cheng X, Qi Y, Chen Y, Das I, Li X, Vallat B, Fu LW, Qian CN, Wang HY, White E, Burley SK, Zheng XFS. SOD1 Phosphorylation by mTORC1 Couples Nutrient Sensing and Redox Regulation. Mol Cell 2018; 70:502-515.e8. [PMID: 29727620 PMCID: PMC6108545 DOI: 10.1016/j.molcel.2018.03.029] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 10/12/2017] [Accepted: 03/26/2018] [Indexed: 12/31/2022]
Abstract
Nutrients are not only organic compounds fueling bioenergetics and biosynthesis, but also key chemical signals controlling growth and metabolism. Nutrients enormously impact the production of reactive oxygen species (ROS), which play essential roles in normal physiology and diseases. How nutrient signaling is integrated with redox regulation is an interesting, but not fully understood, question. Herein, we report that superoxide dismutase 1 (SOD1) is a conserved component of the mechanistic target of rapamycin complex 1 (mTORC1) nutrient signaling. mTORC1 regulates SOD1 activity through reversible phosphorylation at S39 in yeast and T40 in humans in response to nutrients, which moderates ROS level and prevents oxidative DNA damage. We further show that SOD1 activation enhances cancer cell survival and tumor formation in the ischemic tumor microenvironment and protects against the chemotherapeutic agent cisplatin. Collectively, these findings identify a conserved mechanism by which eukaryotes dynamically regulate redox homeostasis in response to changing nutrient conditions.
Collapse
Affiliation(s)
- Chi Kwan Tsang
- Rutgers Cancer Institute of New Jersey, Rutgers, State University of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08903, USA; Department of Pharmacology, Robert Wood Johnson Medical School, Rutgers, State University of New Jersey, 675 Hoes Lane, Piscataway, NJ 08854, USA; Clinical Neuroscience Institute, The First Affiliated Hospital, Jinan University, 613 Huangpu Avenue West, Guangzhou, Guangdong 510632, China
| | - Miao Chen
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Xin Cheng
- Rutgers Cancer Institute of New Jersey, Rutgers, State University of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08903, USA
| | - Yanmei Qi
- Rutgers Cancer Institute of New Jersey, Rutgers, State University of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08903, USA
| | - Yin Chen
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Ishani Das
- Rutgers Cancer Institute of New Jersey, Rutgers, State University of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08903, USA
| | - Xiaoxing Li
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Brinda Vallat
- Institute for Quantitative Biomedicine and Department of Chemistry and Chemical Biology, Rutgers, State University of New Jersey, Piscataway, NJ 08854 USA
| | - Li-Wu Fu
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Chao-Nan Qian
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China
| | - Hui-Yun Wang
- Rutgers Cancer Institute of New Jersey, Rutgers, State University of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08903, USA; State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China; Department of Pharmacology, Robert Wood Johnson Medical School, Rutgers, State University of New Jersey, 675 Hoes Lane, Piscataway, NJ 08854, USA
| | - Eileen White
- Rutgers Cancer Institute of New Jersey, Rutgers, State University of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08903, USA; Department of Molecular Biology and Biochemistry, Rutgers, State University of New Jersey, 604 Allison Road, Piscataway, NJ 08854, USA
| | - Stephen K Burley
- Rutgers Cancer Institute of New Jersey, Rutgers, State University of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08903, USA; Institute for Quantitative Biomedicine and Department of Chemistry and Chemical Biology, Rutgers, State University of New Jersey, Piscataway, NJ 08854 USA
| | - X F Steven Zheng
- Rutgers Cancer Institute of New Jersey, Rutgers, State University of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08903, USA; Department of Pharmacology, Robert Wood Johnson Medical School, Rutgers, State University of New Jersey, 675 Hoes Lane, Piscataway, NJ 08854, USA.
| |
Collapse
|
22
|
Guthrie G, Tackett B, Stoll B, Martin C, Olutoye O, Burrin DG. Phytosterols Synergize With Endotoxin to Augment Inflammation in Kupffer Cells but Alone Have Limited Direct Effect on Hepatocytes. JPEN J Parenter Enteral Nutr 2017; 42:37-48. [PMID: 28792854 DOI: 10.1177/0148607117722752] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 06/30/2017] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Phytosterols are implicated in the development of parenteral nutrition-associated liver disease. A newly proposed mechanism for phytosterol-mediated parenteral nutrition-associated liver disease is through phytosterol-facilitated hepatic proinflammatory cytokine release following exposure to intestinally derived bacteria. Whether the proinflammatory effects are liver cell specific is not known. AIM To determine if phytosterols cause inflammation in hepatocytes or Kupffer cells independently or require costimulation by lipopolysaccharide (LPS). METHODS In an in vivo study, neonatal piglets on parenteral nutrition for 11 days received an 8-hour infusion of LPS. In the in vitro studies, neonatal piglet Kupffer cells and hepatocytes were treated with media, media + 1% soy oil, or media + 1% soy oil + 100µM phytosterols. After 24-hour incubation, cells were treated with farnesoid X receptor (FXR) agonist obeticholic acid or liver X receptor (LXR) agonist GW3965 and challenged with LPS or interleukin 1β. RESULTS LPS administration in piglets led to transient increases in proinflammatory cytokines and suppression of the transporters bile salt export pump and ATP-binding cassette transporter G5. In hepatocytes, phytosterols did not activate inflammation. Phytosterol treatment alone did not activate inflammation in Kupffer cells but, combined with LPS, synergistically increased interleukin 1β production. FXR and LXR agonists increased transporter expression in hepatocytes. GW3965 suppressed proinflammatory cytokine production in Kupffer cells, but obeticholic acid did not. CONCLUSIONS LPS suppresses transporters that control bile acid and phytosterol clearance. Phytosterols alone do not cause inflammatory response. However, with costimulation by LPS, phytosterols synergistically maximize the inflammatory response in Kupffer cells.
Collapse
Affiliation(s)
- Gregory Guthrie
- USDA/ARS Children's Nutrition Research Center, Section of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Bryan Tackett
- USDA/ARS Children's Nutrition Research Center, Section of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Barbara Stoll
- USDA/ARS Children's Nutrition Research Center, Section of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Camilia Martin
- Department of Neonatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Oluyinka Olutoye
- Texas Children's Hospital, Division of Pediatric Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas, USA
| | - Douglas G Burrin
- USDA/ARS Children's Nutrition Research Center, Section of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
23
|
Chung JW, Seo WK, Kim GM, Chung CS, Lee KH, Bang OY. Free fatty acid as a determinant of ischemic lesion volume in nonarterial-origin embolic stroke. J Neurol Sci 2017; 382:116-121. [DOI: 10.1016/j.jns.2017.09.040] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 09/06/2017] [Accepted: 09/27/2017] [Indexed: 11/16/2022]
|
24
|
Ghosh A, Gao L, Thakur A, Siu PM, Lai CWK. Role of free fatty acids in endothelial dysfunction. J Biomed Sci 2017; 24:50. [PMID: 28750629 PMCID: PMC5530532 DOI: 10.1186/s12929-017-0357-5] [Citation(s) in RCA: 279] [Impact Index Per Article: 34.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 07/24/2017] [Indexed: 02/06/2023] Open
Abstract
Plasma free fatty acids levels are increased in subjects with obesity and type 2 diabetes, playing detrimental roles in the pathogenesis of atherosclerosis and cardiovascular diseases. Increasing evidence showing that dysfunction of the vascular endothelium, the inner lining of the blood vessels, is the key player in the pathogenesis of atherosclerosis. In this review, we aimed to summarize the roles and the underlying mechanisms using the evidence collected from clinical and experimental studies about free fatty acid-mediated endothelial dysfunction. Because of the multifaceted roles of plasma free fatty acids in mediating endothelial dysfunction, elevated free fatty acid level is now considered as an important link in the onset of endothelial dysfunction due to metabolic syndromes such as diabetes and obesity. Free fatty acid-mediated endothelial dysfunction involves several mechanisms including impaired insulin signaling and nitric oxide production, oxidative stress, inflammation and the activation of the renin-angiotensin system and apoptosis in the endothelial cells. Therefore, targeting the signaling pathways involved in free fatty acid-induced endothelial dysfunction could serve as a preventive approach to protect against the occurrence of endothelial dysfunction and the subsequent complications such as atherosclerosis.
Collapse
Affiliation(s)
- Arijit Ghosh
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, HKSAR, China
- Department of Biomedical Sciences, City University of Hong Kong, HKSAR, China
| | - Lei Gao
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, HKSAR, China
| | - Abhimanyu Thakur
- Department of Biomedical Sciences, City University of Hong Kong, HKSAR, China
| | - Parco M. Siu
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, HKSAR, China
| | - Christopher W. K. Lai
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, HKSAR, China
| |
Collapse
|
25
|
Saha SK, Lee SB, Won J, Choi HY, Kim K, Yang GM, Dayem AA, Cho SG. Correlation between Oxidative Stress, Nutrition, and Cancer Initiation. Int J Mol Sci 2017; 18:E1544. [PMID: 28714931 PMCID: PMC5536032 DOI: 10.3390/ijms18071544] [Citation(s) in RCA: 236] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 07/12/2017] [Accepted: 07/13/2017] [Indexed: 02/07/2023] Open
Abstract
Inadequate or excessive nutrient consumption leads to oxidative stress, which may disrupt oxidative homeostasis, activate a cascade of molecular pathways, and alter the metabolic status of various tissues. Several foods and consumption patterns have been associated with various cancers and approximately 30-35% of the cancer cases are correlated with overnutrition or malnutrition. However, several contradictory studies are available regarding the association between diet and cancer risk, which remains to be elucidated. Concurrently, oxidative stress is a crucial factor for cancer progression and therapy. Nutritional oxidative stress may be induced by an imbalance between antioxidant defense and pro-oxidant load due to inadequate or excess nutrient supply. Oxidative stress is a physiological state where high levels of reactive oxygen species (ROS) and free radicals are generated. Several signaling pathways associated with carcinogenesis can additionally control ROS generation and regulate ROS downstream mechanisms, which could have potential implications in anticancer research. Cancer initiation may be modulated by the nutrition-mediated elevation in ROS levels, which can stimulate cancer initiation by triggering DNA mutations, damage, and pro-oncogenic signaling. Therefore, in this review, we have provided an overview of the relationship between nutrition, oxidative stress, and cancer initiation, and evaluated the impact of nutrient-mediated regulation of antioxidant capability against cancer therapy.
Collapse
Affiliation(s)
- Subbroto Kumar Saha
- Department of Stem Cell and Regenerative Biotechnology, Incurable Disease Animal Model & Stem Cell Institute (IDASI), Konkuk University, Seoul 05029, Korea.
| | - Soo Bin Lee
- Department of Stem Cell and Regenerative Biotechnology, Incurable Disease Animal Model & Stem Cell Institute (IDASI), Konkuk University, Seoul 05029, Korea.
| | - Jihye Won
- Department of Stem Cell and Regenerative Biotechnology, Incurable Disease Animal Model & Stem Cell Institute (IDASI), Konkuk University, Seoul 05029, Korea.
| | - Hye Yeon Choi
- Department of Stem Cell and Regenerative Biotechnology, Incurable Disease Animal Model & Stem Cell Institute (IDASI), Konkuk University, Seoul 05029, Korea.
| | - Kyeongseok Kim
- Department of Stem Cell and Regenerative Biotechnology, Incurable Disease Animal Model & Stem Cell Institute (IDASI), Konkuk University, Seoul 05029, Korea.
| | - Gwang-Mo Yang
- Department of Stem Cell and Regenerative Biotechnology, Incurable Disease Animal Model & Stem Cell Institute (IDASI), Konkuk University, Seoul 05029, Korea.
| | - Ahmed Abdal Dayem
- Department of Stem Cell and Regenerative Biotechnology, Incurable Disease Animal Model & Stem Cell Institute (IDASI), Konkuk University, Seoul 05029, Korea.
| | - Ssang-Goo Cho
- Department of Stem Cell and Regenerative Biotechnology, Incurable Disease Animal Model & Stem Cell Institute (IDASI), Konkuk University, Seoul 05029, Korea.
| |
Collapse
|
26
|
Dhanda N, Taheri S. A narrative review of obesity and hearing loss. Int J Obes (Lond) 2017; 41:1066-1073. [PMID: 28163314 DOI: 10.1038/ijo.2017.32] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 12/18/2016] [Accepted: 01/23/2017] [Indexed: 12/20/2022]
Abstract
The comorbidities related to obesity are already extensive, but as the prevalence of obesity increases globally, so do the number of its associated conditions. The relationship between hearing impairment and obesity is a relatively recent research interest, but is significant as both conditions have the ability to substantially reduce an individual's quality of life both physically and psychologically. Obesity has a significant effect on vascular function, and this may have an impact on highly vascular organs such as the auditory system. This review aims to provide an overview of the existing literature surrounding the association between hearing loss and obesity, in order to emphasise these two highly prevalent conditions, and to identify areas of further investigation. Our literature search identified a total of 298 articles with 11 articles of relevance to the review. The existing literature in this area is sparse, with interest ranging from obesity and its links to age-related hearing impairment (ARHI) and sudden sensorineural hearing loss (SSNHL), to animal models and genetic syndromes that incorporate both disorders. A key hypothesis for the underlying mechanism for the relationship between obesity and hearing loss is that of vasoconstriction in the inner ear, whereby strain on the capillary walls due to excess adipose tissue causes damage to the delicate inner ear system. The identified articles in this review have not established a causal relationship between obesity and hearing impairment. Further research is required to examine the emerging association between obesity and hearing impairment, and identify its potential underlying mechanisms.
Collapse
Affiliation(s)
- N Dhanda
- Clinical Research Core, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha, Qatar
| | - S Taheri
- Clinical Research Core, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha, Qatar.,Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
27
|
Diamanti-Kandarakis E, Papalou O, Kandaraki EA, Kassi G. MECHANISMS IN ENDOCRINOLOGY: Nutrition as a mediator of oxidative stress in metabolic and reproductive disorders in women. Eur J Endocrinol 2017; 176:R79-R99. [PMID: 27678478 DOI: 10.1530/eje-16-0616] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Revised: 09/20/2016] [Accepted: 09/27/2016] [Indexed: 12/12/2022]
Abstract
Nutrition can generate oxidative stress and trigger a cascade of molecular events that can disrupt oxidative and hormonal balance. Nutrient ingestion promotes a major inflammatory and oxidative response at the cellular level in the postprandial state, altering the metabolic state of tissues. A domino of unfavorable metabolic changes is orchestrated in the main metabolic organs, including adipose tissue, skeletal muscle, liver and pancreas, where subclinical inflammation, endothelial dysfunction, mitochondrial deregulation and impaired insulin response and secretion take place. Simultaneously, in reproductive tissues, nutrition-induced oxidative stress can potentially violate delicate oxidative balance that is mandatory to secure normal reproductive function. Taken all the above into account, nutrition and its accompanying postprandial oxidative stress, in the unique context of female hormonal background, can potentially compromise normal metabolic and reproductive functions in women and may act as an active mediator of various metabolic and reproductive disorders.
Collapse
Affiliation(s)
| | - Olga Papalou
- Department of Endocrinology and Diabetes Center of ExcellenceEUROCLINIC, Athens, Greece
| | - Eleni A Kandaraki
- Endocrine Unit3rd Department of Internal Medicine, University of Athens Medical School, Athens, Greece
| | - Georgia Kassi
- Endocrine Unit3rd Department of Internal Medicine, University of Athens Medical School, Athens, Greece
| |
Collapse
|
28
|
Gene-metabolite network analysis in different nonalcoholic fatty liver disease phenotypes. Exp Mol Med 2017; 49:e283. [PMID: 28082742 PMCID: PMC5291835 DOI: 10.1038/emm.2016.123] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 07/12/2016] [Accepted: 08/08/2016] [Indexed: 12/17/2022] Open
Abstract
We sought to identify common key regulators and build a gene-metabolite network in different nonalcoholic fatty liver disease (NAFLD) phenotypes. We used a high-fat diet (HFD), a methionine-choline-deficient diet (MCDD) and streptozocin (STZ) to establish nonalcoholic fatty liver (NAFL), nonalcoholic steatohepatitis (NASH) and NAFL+type 2 diabetes mellitus (T2DM) in rat models, respectively. Transcriptomics and metabolomics analyses were performed in rat livers and serum. A functional network-based regulation model was constructed using Cytoscape with information derived from transcriptomics and metabolomics. The results revealed that 96 genes, 17 liver metabolites and 4 serum metabolites consistently changed in different NAFLD phenotypes (>2-fold, P<0.05). Gene-metabolite network analysis identified ccl2 and jun as hubs with the largest connections to other genes, which were mainly involved in tumor necrosis factor, P53, nuclear factor-kappa B, chemokine, peroxisome proliferator activated receptor and Toll-like receptor signaling pathways. The specifically regulated genes and metabolites in different NAFLD phenotypes constructed their own networks, which were mainly involved in the lipid and fatty acid metabolism in HFD models, the inflammatory and immune response in MCDD models, and the AMPK signaling pathway and response to insulin in HFD+STZ models. Our study identified networks showing the general and specific characteristics in different NAFLD phenotypes, complementing the genetic and metabolic features in NAFLD with hepatic and extra-hepatic manifestations.
Collapse
|
29
|
Lonardo A, Ballestri S, Guaraldi G, Nascimbeni F, Romagnoli D, Zona S, Targher G. Fatty liver is associated with an increased risk of diabetes and cardiovascular disease - Evidence from three different disease models: NAFLD, HCV and HIV. World J Gastroenterol 2016; 22:9674-9693. [PMID: 27956792 PMCID: PMC5124973 DOI: 10.3748/wjg.v22.i44.9674] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 09/29/2016] [Accepted: 10/30/2016] [Indexed: 02/06/2023] Open
Abstract
Fatty liver, which frequently coexists with necro-inflammatory and fibrotic changes, may occur in the setting of nonalcoholic fatty liver disease (NAFLD) and chronic infections due to either hepatitis C virus (HCV) or human immunodeficiency virus (HIV). These three pathologic conditions are associated with an increased prevalence and incidence of cardiovascular disease (CVD) and type 2 diabetes (T2D). In this multidisciplinary clinical review, we aim to discuss the ever-expanding wealth of clinical and epidemiological evidence supporting a key role of fatty liver in the development of T2D and CVD in patients with NAFLD and in those with HCV or HIV infections. For each of these three common diseases, the epidemiological features, pathophysiologic mechanisms and clinical implications of the presence of fatty liver in predicting the risk of incident T2D and CVD are examined in depth. Collectively, the data discussed in this updated review, which follows an innovative comparative approach, further reinforce the conclusion that the presence of fatty/inflamed/fibrotic liver might be a shared important determinant for the development of T2D and CVD in patients with NAFLD, HCV or HIV. This review may also open new avenues in the clinical and research arenas and paves the way for the planning of future, well-designed prospective and intervention studies.
Collapse
|
30
|
Morgan A, Mooney K, Mc Auley M. Obesity and the dysregulation of fatty acid metabolism: implications for healthy aging. Expert Rev Endocrinol Metab 2016; 11:501-510. [PMID: 30058918 DOI: 10.1080/17446651.2016.1245141] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The population of the world is aging. In 2010, an estimated 524 million people were aged 65 years or older representing eight percent of the global population. By 2050, this number is expected to nearly triple to approximately 1.5 billion, 16 percent of the world's population. Although people are living longer, the quality of their lives are often compromised due to ill-health. Areas covered: Of the conditions which compromise health as we age, obesity is at the forefront. Over half of the global older population were overweight or obese in 2010, significantly increasing the risk of a range of metabolic diseases. Although, it is well recognised excessive calorie intake is a fundamental driver of adipose tissue dysfunction, the relationship between obesity; intrinsic aging; and fat metabolism is less understood. In this review we discuss the intersection between obesity, aging and the factors which contribute to the dysregulation of whole-body fat metabolism. Expert commentary: Being obese disrupts an array of physiological systems and there is significant crosstalk among these. Moreover it is imperative to acknowledge the contribution intrinsic aging makes to the dysregulation of these systems and the onset of disease.
Collapse
Affiliation(s)
- Amy Morgan
- a Department of Chemical Engineering , University of Chester, Thornton Science Park , Chester , UK
| | - Kathleen Mooney
- b Faculty of Health and Social Care , Edge Hill University , Lancashire , UK
| | - Mark Mc Auley
- a Department of Chemical Engineering , University of Chester, Thornton Science Park , Chester , UK
| |
Collapse
|
31
|
Feriani A, Hachani R, Kaabi B, Ncir M, El Feki A, Allagui MS. Protective effects of Zygophyllum albumextract against deltamethrin-induced hyperglycemia and hepato-pancreatic disorders in rats. Can J Physiol Pharmacol 2016; 94:1202-1210. [DOI: 10.1139/cjpp-2016-0132] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
The current study was designed to investigate the possible mechanism involved in hyperglycemia induced by chronic exposure to deltamethrin (DLM) in rat and to assess whether this damage is amenable to modulation by Zygophyllum album. DLM, a synthetic pyrethroid pesticide, was administrated at a dose of 4 mg/kg body mass, during 60 days. Compared with control, DLM showed a significant increase of blood glucose (p ≤ 0.01) and glycosylated hemoglobin levels (p ≤ 0.01) and a clear decrease (p ≤ 0.01) of insulin and total hemoglobin levels. In addition, hepatic glycogen content and the activity of hexokinase decreased (p ≤ 0.01), whereas the activities of glucose-6-phosphatase and glycogen phosphorylase were significantly increased (p ≤ 0.01). Moreover, pancreatic lipid peroxidation (TBARS level) was higher (p ≤ 0.01) and oxidative stress biomarkers (SOD, CAT, GPx, and GSH) were altered owing to DLM toxicity. However, Z. album, when combined with DLM, significantly ameliorated almost all the hepato-pancreatic disorders induced by DLM alone. Furthermore, Z. album supplement was found to be effective in preserving the normal histological appearance of hepatic and pancreatic tissue. In conclusion, this study suggested that, owing to its antioxidant effects, methanolic extract of Z. album (MEZAL) can potentially prevent the hyperglycemia observed in DLM-treated group.
Collapse
Affiliation(s)
- Anouar Feriani
- Laboratory of Animal Ecophysiology, Faculty of Science of Sfax, 3018 Sfax, Tunisia
- Research Unit of Macromolecular Biochemistry and Genetics, Faculty of Sciences of Gafsa, 2112 Gafsa, Tunisia
| | - Rafik Hachani
- Laboratory of Vascular Pathologies, Integrated Physiology Unit, Faculty of Sciences of Bizerte, 7021 Jarzouna, Tunisia
| | - Belhassen Kaabi
- Laboratory of Epidemiology and Veterinary Microbiology, Institut Pasteur de Tunis, BP 74, 1002 Belvedere-Tunis, Tunisia
| | - Marwa Ncir
- Laboratory of Animal Ecophysiology, Faculty of Science of Sfax, 3018 Sfax, Tunisia
| | - Abdelfattah El Feki
- Laboratory of Animal Ecophysiology, Faculty of Science of Sfax, 3018 Sfax, Tunisia
| | | |
Collapse
|
32
|
Fan Y, Wei F, Zhou Y, Zhang H. Association of non-alcoholic fatty liver disease with impaired endothelial function by flow-mediated dilation: A meta-analysis. Hepatol Res 2016; 46:E165-73. [PMID: 26179110 DOI: 10.1111/hepr.12554] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2015] [Revised: 06/24/2015] [Accepted: 07/12/2015] [Indexed: 02/06/2023]
Abstract
AIM Endothelial dysfunction was observed in patients with non-alcoholic fatty liver disease. The aim of the present study was to evaluate the extent of endothelial function impairment in patients with non-alcoholic fatty liver disease. METHODS We systematically searched through the PubMed, Embase, China National Knowledge Infrastructure and Wanfang databases for studies dated up to January 2015. All observational studies that compared endothelial function between non-alcoholic fatty liver disease patients and healthy controls were included. Endothelial function was measured by brachial artery flow-mediated dilation and nitrate-induced dilatation techniques. RESULTS Eleven observational studies were identified. Meta-analysis indicated that non-alcoholic fatty liver disease patients were associated with a reduction in brachial artery flow-mediated dilation (weighted mean difference, -4.82%; 95% confidence interval [CI], -5.63 to -4.00) compared with the healthy controls. Subgroup analysis indicated that the patients with non-alcoholic steatohepatitis or a body mass index of more than 30 kg/m(2) appeared to have a greater reduction in their flow-mediated dilation. However, no significant differences were observed in the nitrate-induced dilation (weighted mean difference, -0.4%; 95% confidence interval, -2.19 to 1.39). CONCLUSION Non-alcoholic fatty liver disease is associated with a greater risk of endothelial dysfunction as indicated by the decreased brachial artery flow-mediated dilation. Assessment of endothelial function may be recommended to identify subclinical atherosclerosis and to create subgroups of non-alcoholic fatty liver disease patients who are at higher risk of future cardiovascular events.
Collapse
Affiliation(s)
- Yu Fan
- Institute of Molecular Biology and Translational Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, China
| | - Feifei Wei
- Institute of Molecular Biology and Translational Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, China
| | - Yongjing Zhou
- Institute of Molecular Biology and Translational Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, China
| | - Heng Zhang
- Institute of Molecular Biology and Translational Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, China
| |
Collapse
|
33
|
Ditzel EJ, Li H, Foy CE, Perrera AB, Parker P, Renquist BJ, Cherrington NJ, Camenisch TD. Altered Hepatic Transport by Fetal Arsenite Exposure in Diet-Induced Fatty Liver Disease. J Biochem Mol Toxicol 2016; 30:321-30. [PMID: 26890134 DOI: 10.1002/jbt.21796] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 01/09/2016] [Accepted: 01/15/2016] [Indexed: 12/21/2022]
Abstract
Non-alcoholic fatty liver disease can result in changes to drug metabolism and disposition potentiating adverse drug reactions. Furthermore, arsenite exposure during development compounds the severity of diet-induced fatty liver disease. This study examines the effects of arsenite potentiated diet-induced fatty liver disease on hepatic transport in male mice. Changes were detected for Mrp2/3/4 hepatic transporter gene expression as well as for Oatp1a4/2b1/1b2. Plasma concentrations of Mrp and Oatp substrates were increased in arsenic exposure groups compared with diet-only controls. In addition, murine embryonic hepatocytes and adult primary hepatocytes show significantly altered transporter expression after exposure to arsenite alone: a previously unreported phenomenon. These data indicate that developmental exposure to arsenite leads to changes in hepatic transport which could increase the risk for ADRs during fatty liver disease.
Collapse
Affiliation(s)
- Eric J Ditzel
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, USA.
| | - Hui Li
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, USA
| | - Caroline E Foy
- School of Animal and Comparative Biomedical Sciences, The University of Arizona, Tucson, AZ, 85721, USA
| | - Alec B Perrera
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, USA
| | - Patricia Parker
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, USA
| | - Benjamin J Renquist
- School of Animal and Comparative Biomedical Sciences, The University of Arizona, Tucson, AZ, 85721, USA
| | - Nathan J Cherrington
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, USA.,Southwest Environmental Health Sciences Center, The University of Arizona, Tucson, AZ, 85721, USA
| | - Todd D Camenisch
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, USA.,Southwest Environmental Health Sciences Center, The University of Arizona, Tucson, AZ, 85721, USA.,Steele Children's Research Center, Arizona Health Sciences Center, The University of Arizona, Tucson, AZ, 85724, USA.,Sarver Heart Center, The University of Arizona, Tucson, AZ, 85724, USA.,Bio5 Institute, The University of Arizona, Tucson, AZ, 85721, USA
| |
Collapse
|
34
|
Guan Y, Cao WJ, Zhang ZY. Relationship between gut microbiota and non-alcoholic fatty liver disease. Shijie Huaren Xiaohua Zazhi 2015; 23:5797-5802. [DOI: 10.11569/wcjd.v23.i36.5797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The incidence of non-alcoholic fatty liver disease (NAFLD) has been increasing during these years. As we understand more about gut microbiota, the relationship between gut microbiota and NAFLD has been revealed. Both animal experiments and clinical studies show that gut microbiota can not only act on NAFLD via the gut-liver axis and two-hit theory, but also play an important role in liver inflammation and hepatic fibrosis. Experiments also indicate that using probiotics, prebiotics, berberine and antibiotics to regulate gut microbiota can relieve inflammation, lower body mass index and improve insulin resistance, which can be a new treatment for NAFLD and other metabolic diseases.
Collapse
|
35
|
Chang J, Lu Y, Boswell WT, Boswell M, Caballero KL, Walter RB. Molecular genetic response to varied wavelengths of light in Xiphophorus maculatus skin. Comp Biochem Physiol C Toxicol Pharmacol 2015; 178:104-115. [PMID: 26460196 PMCID: PMC4662885 DOI: 10.1016/j.cbpc.2015.10.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 10/01/2015] [Accepted: 10/05/2015] [Indexed: 12/14/2022]
Abstract
Xiphophorus fishes represent a model often utilized to study UVB induced tumorigenesis. Recently, varied genetic responses to UVB exposure have been documented in the skin of female and male Xiphophorus, as have differences in UVB response in the skin of different parental species and for interspecies hybrids produced from crossing them. Additionally, it has been shown that exposure to "cool white" fluorescent light induces a shift in the genetic profiles of Xiphophorus skin that is nearly as robust as the UVB response, but involves a fundamentally different set of genes. Given these results and the use of Xiphophorus interspecies hybrids as an experimental model for UVB inducible melanoma, it is of interest to characterize genes that may be transcriptionally modulated in a wavelength specific manner. The global molecular genetic response of skin upon exposure of the intact animal to specific wavelengths of light has not been investigated. Herein, we report results of RNA-Seq experiments from the skin of male Xiphophorus maculatus Jp 163 B following exposure to varied 50nm wavelengths of light ranging from 300-600nm. We identify two specific wavelength regions, 350-400nm (88 genes) and 500-550nm (276 genes), that exhibit transcriptional modulation of a significantly greater number of transcripts than any of the other 50nm regions in the 300-600nm range. Observed functional sets of genes modulated within these two transcriptionally active light regions suggest different mechanisms of gene modulation.
Collapse
Affiliation(s)
- Jordan Chang
- Molecular Bioscience Research Group and Xiphophorus Genetic Stock Center, Department of Chemistry and Biochemistry, Texas State University, San Marcos, TX 78666, USA.
| | - Yuan Lu
- Molecular Bioscience Research Group and Xiphophorus Genetic Stock Center, Department of Chemistry and Biochemistry, Texas State University, San Marcos, TX 78666, USA.
| | - William T Boswell
- Molecular Bioscience Research Group and Xiphophorus Genetic Stock Center, Department of Chemistry and Biochemistry, Texas State University, San Marcos, TX 78666, USA.
| | - Mikki Boswell
- Molecular Bioscience Research Group and Xiphophorus Genetic Stock Center, Department of Chemistry and Biochemistry, Texas State University, San Marcos, TX 78666, USA.
| | - Kaela L Caballero
- Molecular Bioscience Research Group and Xiphophorus Genetic Stock Center, Department of Chemistry and Biochemistry, Texas State University, San Marcos, TX 78666, USA.
| | - Ronald B Walter
- Molecular Bioscience Research Group and Xiphophorus Genetic Stock Center, Department of Chemistry and Biochemistry, Texas State University, San Marcos, TX 78666, USA.
| |
Collapse
|
36
|
Hafizi Abu Bakar M, Kian Kai C, Wan Hassan WN, Sarmidi MR, Yaakob H, Zaman Huri H. Mitochondrial dysfunction as a central event for mechanisms underlying insulin resistance: the roles of long chain fatty acids. Diabetes Metab Res Rev 2015; 31:453-75. [PMID: 25139820 DOI: 10.1002/dmrr.2601] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Revised: 04/19/2014] [Accepted: 07/23/2014] [Indexed: 12/25/2022]
Abstract
Insulin resistance is characterized by hyperglycaemia, dyslipidaemia and oxidative stress prior to the development of type 2 diabetes mellitus. To date, a number of mechanisms have been proposed to link these syndromes together, but it remains unclear what the unifying condition that triggered these events in the progression of this metabolic disease. There have been a steady accumulation of data in numerous experimental studies showing the strong correlations between mitochondrial dysfunction, oxidative stress and insulin resistance. In addition, a growing number of studies suggest that the raised plasma free fatty acid level induced insulin resistance with the significant alteration of oxidative metabolism in various target tissues such as skeletal muscle, liver and adipose tissue. In this review, we herein propose the idea of long chain fatty acid-induced mitochondrial dysfunctions as one of the key events in the pathophysiological development of insulin resistance and type 2 diabetes. The accumulation of reactive oxygen species, lipotoxicity, inflammation-induced endoplasmic reticulum stress and alterations of mitochondrial gene subset expressions are the most detrimental that lead to the developments of aberrant intracellular insulin signalling activity in a number of peripheral tissues, thereby leading to insulin resistance and type 2 diabetes.
Collapse
Affiliation(s)
- Mohamad Hafizi Abu Bakar
- Department of Bioprocess Engineering, Faculty of Chemical Engineering, Universiti Teknologi Malaysia, Skudai, Johor, Malaysia
| | - Cheng Kian Kai
- Department of Bioprocess Engineering, Faculty of Chemical Engineering, Universiti Teknologi Malaysia, Skudai, Johor, Malaysia
| | - Wan Najihah Wan Hassan
- Department of Bioprocess Engineering, Faculty of Chemical Engineering, Universiti Teknologi Malaysia, Skudai, Johor, Malaysia
| | - Mohamad Roji Sarmidi
- Institute of Bioproduct Development, Universiti Teknologi Malaysia, Skudai, Johor, Malaysia
| | - Harisun Yaakob
- Institute of Bioproduct Development, Universiti Teknologi Malaysia, Skudai, Johor, Malaysia
| | - Hasniza Zaman Huri
- Department of Pharmacy, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
- Clinical Investigation Centre, 13th Floor Main Tower, University Malaya Medical Centre, Lembah Pantai, Kuala Lumpur, Malaysia
| |
Collapse
|
37
|
Sulforaphane protects human umbilical vein cells against lipotoxicity by stimulating autophagy via an AMPK-mediated pathway. J Funct Foods 2015. [DOI: 10.1016/j.jff.2015.03.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
38
|
Hernández I, Domínguez-Pérez M, Bucio L, Souza V, Miranda RU, Clemens DL, Gomez-Quiroz LE, Gutiérrez-Ruiz MC. Free fatty acids enhance the oxidative damage induced by ethanol metabolism in an in vitro model. Food Chem Toxicol 2015; 76:109-15. [DOI: 10.1016/j.fct.2014.12.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 12/05/2014] [Accepted: 12/06/2014] [Indexed: 12/30/2022]
|
39
|
Yin J, Luo Y, Deng H, Qin S, Tang W, Zeng L, Zhou B. Hugan Qingzhi medication ameliorates hepatic steatosis by activating AMPK and PPARα pathways in L02 cells and HepG2 cells. JOURNAL OF ETHNOPHARMACOLOGY 2014; 154:229-239. [PMID: 24735863 DOI: 10.1016/j.jep.2014.04.011] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Revised: 03/15/2014] [Accepted: 04/05/2014] [Indexed: 06/03/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Hugan Qingzhi tablet (HQT), a lipid- lowering traditional Chinese medicine formula, has been used for the prevention and treatment of nonalcoholic fatty liver (NAFLD). AIM OF THE STUDY This study was realized to evaluate the effects of HQT-medicated serum on hepatic steatosis using in vitro experiments with cells and explore the relevant mechanisms with method of serum pharmacology. MATERIALS AND METHODS A model of hepatic steatosis in the L02 and HepG2 cells was induced by free fatty acid (FFA). The components in the HQT-medicated serum were assayed by high-performance liquid chromatography. Intracellular lipid droplets were detected by Oil Red O staining, and their ultrastructure was examined by transmission electron microscope. The biochemical parameters, including triglyceride (TG), lactate dehydrogenase (LDH), aspartate aminotransferase (AST) and alanine aminotransferase (ALT), total antioxidant capacity (T-AOC), malondialdehyde (MDA), superoxide dismutase (SOD) and glutathione (GSH), were measured with commercial kits. Furthermore, the expression of adiponectin, AMP-activated protein kinase (AMPK) phosphorylation, sterol regulatory element-binding protein 1 (SREBP-1), peroxisome proliferator activated receptor-α (PPARα), carnitine palmitoyltransferase 1 (CPT-1), and acetyl-CoA oxidase 1 (ACOX1) was analyzed by Western blot and/or quantitative reverse transcription-polymerase chain reaction (qRT-PCR). RESULTS Moderate- and high-dose HQT-medicated serum reduced (P<0.05 or P<0.01) the accumulation of lipid droplets and the cellular TG content in L02 and HepG2 cells. They caused significant reductions (P<0.01) in LDH, AST, ALT and MDA and significant increase (P<0.05 or P<0.01) in T-AOC in the culture medium. They also caused increase (P<0.05 or P<0.01) in GSH level and SOD activity in FFA-induced steatotic L02 and HepG2 cells. Furthermore, moderate- and high-dose HQT-medicated serum enhanced (P<0.01) adiponectin expression in a concentration-dependent manner and increased (P<0.05 or P<0.01) the phosphorylation of AMPK and the expression of PPARα, CPT-1, and ACOX1, and reduced (P<0.05 or P<0.01) the expression of SREBP-1. CONCLUSION The results suggested that HQT-medicated serum exerts a preventive effect against hepatic steatosis, and the potential mechanism might be activation of AMPK and PPARα pathways.
Collapse
Affiliation(s)
- JinJin Yin
- Center for Drug Research and Development, Zhujiang Hospital, Southern Medical University, Guangdong, Guangzhou 510282, PR China
| | - YanQin Luo
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| | - HouLiang Deng
- Center for Drug Research and Development, Zhujiang Hospital, Southern Medical University, Guangdong, Guangzhou 510282, PR China
| | - ShuMin Qin
- Institute of Digestive Disease, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, PR China
| | - WaiJiao Tang
- Center for Drug Research and Development, Zhujiang Hospital, Southern Medical University, Guangdong, Guangzhou 510282, PR China
| | - Lu Zeng
- Center for Drug Research and Development, Zhujiang Hospital, Southern Medical University, Guangdong, Guangzhou 510282, PR China
| | - BenJie Zhou
- Center for Drug Research and Development, Zhujiang Hospital, Southern Medical University, Guangdong, Guangzhou 510282, PR China.
| |
Collapse
|
40
|
Saremi B, Winand S, Friedrichs P, Kinoshita A, Rehage J, Dänicke S, Häussler S, Breves G, Mielenz M, Sauerwein H. Longitudinal profiling of the tissue-specific expression of genes related with insulin sensitivity in dairy cows during lactation focusing on different fat depots. PLoS One 2014; 9:e86211. [PMID: 24465964 PMCID: PMC3897665 DOI: 10.1371/journal.pone.0086211] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Accepted: 12/09/2013] [Indexed: 01/13/2023] Open
Abstract
In dairy cows the milk associated energy output in early lactation exceeds the input via voluntary feed intake. To spare glucose for mammary lactose synthesis, peripheral insulin sensitivity (IS) is reduced and fat mobilization is stimulated. For these processes a link between IS and the endocrine functions of adipose tissue (AT) is likely; we thus aimed to characterise the mRNA expression from bovine AT derived proteins and receptors that are related to IS according to the literature in metabolically active tissues plus systemic IS throughout lactation. Conjugated linoleic acids (CLA) reduce milk fat thus decreasing the milk drain of energy and potentially dampening lipolysis, but may also affect IS. Subcutaneous (s.c.) AT and liver from pluriparous cows receiving either control fat or CLA supplement (100 g/day from 1 to 182 days in milk each) were biopsied covering week -3 to 36 relative to parturition. In an additional trial with primiparous cows treated analogously and slaughtered on days in milk 1, 42 or 105, samples from liver, udder, skeletal muscle and 3 visceral and 3 s.c. AT were obtained and assayed for mRNA abundance of adiponectin, its receptors, leptin, leptin receptor, PPARγ, PPARγ2, IL-6, and TNF-α. In pluriparous animals, the mRNA abundance of most of the target genes decreased after parturition in s.c. AT but increased in liver. In primiparous cows, AT depot specific differences were mostly related to retroperitoneal AT; adiponectin receptor 1 and TNF-α were affected predominantly. CLA effects in primiparous cows were largely limited to decreased PPARγ2 mRNA abundance in udder tissue. In pluriparous cows, insulin secretion was increased by CLA resulting in decreased systemic IS but without consistent changes in tissue target mRNA abundance. The temporal gene expression profiles from the adipokines and related receptors support their coactive function in adapting to the needs of lactation.
Collapse
Affiliation(s)
- Behnam Saremi
- Institute of Animal Science, Physiology and Hygiene Unit, University of Bonn, Bonn, Germany
| | - Sarah Winand
- Institute of Animal Science, Physiology and Hygiene Unit, University of Bonn, Bonn, Germany
| | - Paula Friedrichs
- Institute of Animal Science, Physiology and Hygiene Unit, University of Bonn, Bonn, Germany
| | - Asako Kinoshita
- Clinic for Cattle, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Jürgen Rehage
- Clinic for Cattle, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Sven Dänicke
- Institute of Animal Nutrition, Friedrich-Loeffler-Institute (FLI), Federal Research Institute for Animal Health, Braunschweig, Germany
| | - Susanne Häussler
- Institute of Animal Science, Physiology and Hygiene Unit, University of Bonn, Bonn, Germany
| | - Gerhard Breves
- Department of Physiology, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Manfred Mielenz
- Institute of Animal Science, Physiology and Hygiene Unit, University of Bonn, Bonn, Germany
| | - Helga Sauerwein
- Institute of Animal Science, Physiology and Hygiene Unit, University of Bonn, Bonn, Germany
| |
Collapse
|
41
|
Li S, Li J, Shen C, Zhang X, Sun S, Cho M, Sun C, Song Z. tert-Butylhydroquinone (tBHQ) protects hepatocytes against lipotoxicity via inducing autophagy independently of Nrf2 activation. BIOCHIMICA ET BIOPHYSICA ACTA 2014; 1841:22-33. [PMID: 24055888 PMCID: PMC3884638 DOI: 10.1016/j.bbalip.2013.09.004] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 08/23/2013] [Accepted: 09/10/2013] [Indexed: 12/30/2022]
Abstract
Saturated fatty acids (SFAs) induce hepatocyte cell death, wherein oxidative stress is mechanistically involved. Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) is a master transcriptional regulator of cellular antioxidant defense enzymes. Therefore, Nrf2 activation is regarded as an effective strategy against oxidative stress-triggered cellular damage. In this study, tert-butylhydroquinone (tBHQ), a widely used Nrf2 activator, was initially employed to investigate the potential protective role of Nrf2 activation in SFA-induced hepatoxicity. As expected, SFA-induced hepatocyte cell death was prevented by tBHQ in both AML-12 mouse hepatocytes and HepG2 human hepatoma cells. However, the protective effect of tBHQ is Nrf2-independent, because the siRNA-mediated Nrf2 silencing did not abrogate tBHQ-conferred protection. Alternatively, our results revealed that autophagy activation was critically involved in the protective effect of tBHQ on lipotoxicity. tBHQ induced autophagy activation and autophagy inhibitors abolished tBHQ's protection. The induction of autophagy by tBHQ exposure was demonstrated by the increased accumulation of LC3 puncta, LC3-II conversion, and autophagic flux (LC3-II conversion in the presence of proteolysis inhibitors). Subsequent mechanistic investigation discovered that tBHQ exposure activated AMP-activated protein kinase (AMPK) and siRNA-mediated AMPK gene silencing abolished tBHQ-induced autophagy activation, indicating that AMPK is critically involved in tBHQ-triggered autophagy induction. Furthermore, our study provided evidence that tBHQ-induced autophagy activation is required for its Nrf2-activating property. Collectively, our data uncover a novel mechanism for tBHQ in protecting hepatocytes against SFA-induced lipotoxicity. tBHQ-triggered autophagy induction contributes not only to its hepatoprotective effect, but also to its Nrf2-activating property.
Collapse
Affiliation(s)
- Songtao Li
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, 150081, P. R. China
| | - Jiaxin Li
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612
| | - Chen Shen
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612
| | - Ximei Zhang
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612
| | - Shan Sun
- Department of Bioengineering, College of Engineering, University of Illinois at Chicago, IL60607
| | - Michael Cho
- Department of Bioengineering, College of Engineering, University of Illinois at Chicago, IL60607
| | - Changhao Sun
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, 150081, P. R. China
| | - Zhenyuan Song
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612
- Department of Pathology, University of Illinois Medical Center, Chicago, IL 60612
| |
Collapse
|
42
|
Niu J, Gilliland MGF, Jin Z, Kolattukudy PE, Hoffman WH. MCP-1and IL-1β expression in the myocardia of two young patients with Type 1 diabetes mellitus and fatal diabetic ketoacidosis. Exp Mol Pathol 2013; 96:71-9. [PMID: 24246157 DOI: 10.1016/j.yexmp.2013.11.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Revised: 11/04/2013] [Accepted: 11/04/2013] [Indexed: 01/01/2023]
Abstract
Convincing evidence exists for the early onset of diabetic cardiomyopathy and coronary artery disease (CAD) as distinct forms of cardiac disease in young patients with Type 1 diabetes mellitus (T1DM) and the pre-stages of T2DM, forms of dysregulated insulin signaling. Progression of both chronic cardiac conditions is mediated by oxidative stress and low grade inflammation. This study reports the expression of monocyte chemotactic protein-1 (MCP-1) chemokine and the interleukin (IL)-1β inflammatory cytokine in two young patients with suboptimal metabolic control and fatal diabetic ketoacidosis (DKA), two age-matched overweight/obesity cases and two age-matched controls. In addition, markers of oxidative stress, apoptosis, collagen deposition and cardiomyocyte hypertrophy were studied. Significant expression of MCP-1 and IL-1β was seen in the myocardia of the T1DM/DKA cases, with lesser amounts expressed in the overweight/obesity myocardia. All of the other markers except cardiomyocyte hypertrophy were expressed to a significantly greater extent in the T1DM/DKA and overweight/obesity cases in comparison to the age-matched controls. Cardiomyocyte hypertrophy was significantly greater in the overweight/obesity cases than in the T1DM/DKA or the control cases.
Collapse
Affiliation(s)
- Jianli Niu
- Burnett School of Biomedical Sciences, College of medicine, University of Central Florida, Orlando, FL, USA
| | - M G F Gilliland
- Department of Pathology and Laboratory Medicine, The Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Zhuqing Jin
- School of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Pappachan E Kolattukudy
- Burnett School of Biomedical Sciences, College of medicine, University of Central Florida, Orlando, FL, USA
| | - William H Hoffman
- Department of Pediatrics, Section of Pediatric Endocrinology, Georgia Regents University, Augusta, GA, USA.
| |
Collapse
|
43
|
Huang KW, Leu HB, Wang YJ, Luo JC, Lin HC, Lee FY, Chan WL, Lin JK, Chang FY. Patients with nonalcoholic fatty liver disease have higher risk of colorectal adenoma after negative baseline colonoscopy. Colorectal Dis 2013; 15:830-5. [PMID: 23398678 DOI: 10.1111/codi.12172] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Accepted: 12/15/2012] [Indexed: 12/13/2022]
Abstract
AIM The study aimed to determine whether nonalcoholic fatty liver disease (NAFLD) is an independent risk factor of adenoma after negative baseline colonoscopy. METHOD A retrospective cohort study was conducted on 1522 health-check individuals who underwent two consecutive colonoscopies at Taipei Veterans General Hospital between 2003 and 2010. Those developing an adenoma after an initial negative baseline colonoscopy (adenoma group) were compared with those in whom the second colonoscopy was negative (nonadenoma group). Anthropometric measurements, biochemical tests and the presence of NAFLD were compared between the two groups. RESULTS The adenoma group had a higher prevalence of NAFLD than the nonadenoma group (55.6% vs 38.8%; P < 0.05). On multivariate logistic regression analysis, NAFLD was an independent risk factor (OR = 1.45, 95% CI: 1.07-1.98) for adenoma formation after a negative baseline colonoscopy. The risk of colorectal adenoma increased when NAFLD patients had other morbidities including metabolic syndrome, hypertension or smoking (OR = 2.85, 4.03 and 4.17). CONCLUSION NAFLD is an independent risk factor for colorectal adenoma formation after a negative baseline colonoscopy. The risk is higher in individuals with NAFLD and other comorbidities, such as hypertension, smoking or metabolic syndrome.
Collapse
Affiliation(s)
- K-W Huang
- Department of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Masuoka HC, Chalasani N. Nonalcoholic fatty liver disease: an emerging threat to obese and diabetic individuals. Ann N Y Acad Sci 2013; 1281:106-22. [PMID: 23363012 PMCID: PMC3646408 DOI: 10.1111/nyas.12016] [Citation(s) in RCA: 194] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common liver disease in the Western world and its incidence is increasing rapidly. NAFLD is a spectrum ranging from simple steatosis, which is relatively benign hepatically, to nonalcoholic steatohepatitis (NASH), which can progress to cirrhosis. Obesity, insulin resistance, type 2 diabetes mellitus, and dyslipidemia are the most important risk factors for NAFLD. Due to heavy enrichment with metabolic risk factors, individuals with NAFLD are at significantly higher risk for cardiovascular disease. Individuals with NAFLD have higher incidence of type 2 diabetes. The diagnosis of NAFLD requires imaging evidence of hepatic steatosis in the absence of competing etiologies including significant alcohol consumption. Liver biopsy remains the gold standard for diagnosing NASH and for determining prognosis. Weight loss remains a cornerstone of treatment. Weight loss of ∼5% is believed to improve steatosis, whereas ∼10% weight loss is necessary to improve steatohepatitis. A number of pharmacologic therapies have been investigated to treat NASH, and agents such as vitamin E and thiazolidinediones have shown promise in select patient subgroups.
Collapse
Affiliation(s)
- Howard C Masuoka
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | |
Collapse
|